You are on page 1of 492

Handbook of Pediatric Hematology

and Oncology
Handbook
of Pediatric
Hematology
and Oncology
Children’s Hospital &
Research Center Oakland

Dr. Caroline A. Hastings


Division of Hematology, Oncology and BMT, Children’s Hospital &
Research Center Oakland, Oakland, CA, USA

Dr. Joseph C. Torkildson


Division of Hematology, Oncology and BMT, Children’s Hospital &
Research Center Oakland, Oakland, CA, USA

Dr. Anurag K. Agrawal


Division of Hematology, Oncology and BMT, Children’s Hospital &
Research Center Oakland, Oakland, CA, USA

Third Edition
This edition first published 2021
© 2021 John Wiley & Sons Ltd
Wiley‐Blackwell (1e, 2012)
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 law. Advice on how to obtain permission to reuse material from this title is available at
http://www.wiley.com/go/permissions.
The right of Dr. Caroline A. Hastings, Dr. Joseph C. Torkildson, and Dr. Anurag K. Agrawal to be identified as
the author(s) of this work has been asserted in accordance with law.
Registered Offices
John Wiley & Sons, Inc., 111 River Street, Hoboken, NJ 07030, USA
John Wiley & Sons Ltd., The Atrium, Southern Gate, Chichester, West Sussex, PO19 8SQ, UK
Editorial Office
9600 Garsington Road, Oxford, OX4 2DQ, UK
For details of our global editorial offices, customer services, and more information about Wiley products, visit
us at www.wiley.com.
Wiley also publishes its books in a variety of electronic formats and by print‐on‐demand. Some content that
appears in standard print versions of this book may not be available in other formats.
Limit of Liability/Disclaimer of Warranty
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 scientific method,
diagnosis, or treatment by physicians for any particular patient. 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. While the publisher and
authors have used their best efforts in preparing this work, they 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 merchantability or fitness for a particular purpose. No
warranty may be created or extended by sales representatives, written sales materials, or promotional
statements for this work. The fact that an organization, website, or product is referred to in this work as a
citation and/or potential source of further information does not mean that the publisher and authors endorse
the information or services the organization, website, or product may provide or recommendations it may
make. This work is sold with the understanding that the publisher is not engaged in rendering professional
services. The advice and strategies contained herein may not be suitable for your situation. You should consult
with a specialist where appropriate. Further, readers should be aware that websites listed in this work may have
changed or disappeared between when this work was written and when it is read. Neither the publisher nor
authors shall be liable for any loss of profit or any other commercial damages, including but not limited to
special, incidental, consequential, or other damages.
Library of Congress Cataloging‐in‐Publication Data
Names: Hastings, Caroline, 1960– author. | Torkildson, Joseph C., author. |
Agrawal, Anurag K. (Anurag Kishor), author. | Children’s Hospital &
Research Center Oakland (Oakland, Calif.)
Title: Handbook of pediatric hematology and oncology : Children’s Hospital
& Research Center Oakland / Dr Caroline A. Hastings, Dr Joseph C.
Torkildson, Anurag K. Agrawal.
Description: Third edition. | Hoboken, NJ : Wiley-Blackwell, 2020. |
Includes bibliographical references and index.
Identifiers: LCCN 2020023659 (print) | LCCN 2020023660 (ebook) | ISBN
9781119210740 (paperback) | ISBN 9781119210764 (adobe pdf) | ISBN
9781119210757 (epub)
Subjects: MESH: Hematologic Diseases | Child | Neoplasms | Handbook
Classification: LCC RJ411 (print) | LCC RJ411 (ebook) | NLM WS 39 | DDC
618.92/994–dc23
LC record available at https://lccn.loc.gov/2020023659
LC ebook record available at https://lccn.loc.gov/2020023660
Cover Design: Wiley
Cover Images: © Billion Photos/Shutterstock
Set in 9.25/11.5pt Minion Pro by SPi Global, Pondicherry, India

10 9 8 7 6 5 4 3 2 1
On a day‐to‐day basis, the patients and their families continue to show us how to live gracefully
in even the most unbearable of times and inspire us to endeavor for improved outcomes. Our
experiences have taught us the magnitude of remembering our roles: “to cure sometimes, to
relieve often, to comfort always.” (anonymous, fifteenth century)
Contents

Preface, ix
Acknowledgments, xi

1 Approach to the Anemic Child, 1

2 Hemolytic Anemia, 15

3 Sickle Cell Disease, 27

4 Thalassemia, 51

5 Transfusion Medicine, 63

6 Chelation Therapy, 79

7 Approach to the Bleeding Child, 85

8 Von Willebrand Disease, 95

9 Hemophilia, 103

10 Thrombophilia, 111

11 The Neutropenic Child, 119

12 Thrombocytopenia, 133

13 Evaluation of the Child with a Suspected Malignancy, 157

14 Oncologic Emergencies, 173

15 Acute Leukemias, 191

16 Central Nervous System Tumors, 211

17 Hodgkin and Non‐Hodgkin Lymphoma, 227

18 Wilms Tumor, 239

19 Neuroblastoma, 245
viii Contents

20 Sarcomas of the Soft Tissues and Bone, 253

21 Germ Cell Tumors, 265

22 Rare Tumors of Childhood, 275

23 Histiocytic Disorders, 287

24 Hematopoietic Stem Cell Transplantation, 295

25 Supportive Care of the Child with Cancer, 313

26 Central Venous Catheters, 323

27 Management of Fever in the Child with Cancer, 333

28 Acute Pain Management in the Inpatient Setting, 347

29 Palliative Care, 361

30 Chemotherapy Basics, 377

31 Guide to Procedures, 393

32 Treatment of Chemotherapy Extravasations, 403

Formulary, 409
Index, 489
Preface

The pace of change in the field of pediatric and observing the myriad variations in dis-
hematology, oncology, and hematopoietic ease and individual nuances that are not
cell therapies is staggering. Molecular biol- addressed in large studies or case reports,
ogy, genomics, and biochemistry have accel- all the while expanding foundational
erated the knowledge and understanding of knowledge.
disease states and further highlight the com- This handbook represents the work of
plex interplay of clinical, genetic, and social our colleagues at Children’s Hospital &
factors that constantly challenge us in the Research Center Oakland toward this
rapid application of novel findings to treat endeavor. The guidelines offered here have
patients with the goal of improved outcomes. been used to instruct medical students,
This translation of knowledge to the unique pediatric residents, nurses, pediatricians,
patient before us, the true art of the physi- and hematology/oncology fellows for over
cian, encompassing experience, knowledge, 25 years. This handbook provides clinical
intuition, and understanding of the individ- approaches for common problems in pedi-
ual needs and goals of patients and families, atric hematology, oncology, hematopoietic
can be overwhelming. What is needed is a stem cell transplant, and newer cellular ther-
practical, tested approach to analyze and apies; knowledge to organize and evaluate
address these problems to ensure timely the care of your patients; and a framework
evaluation, competent clinical care, and to incorporate ever‐expanding psychosocial
avoidance of pitfalls that might negatively needs, clinical studies, medical treatments,
impact the patient or future treatment and science. All of these are essential com-
options. This practical approach is achieved ponents that encompass the care of the child
by spending time with patients and families with blood disorders and cancer.
Acknowledgments

We are grateful to Yoram Unguru, MD, MS, Coleman Abadi, Assistant Professor of pedi-
MA, for submission of the expert case and atrics at UCSF Benioff Children’s Hospital
teaching guide in Chapter 29. Dr. Unguru is an Oakland; Dr. Cheryl Peretz, senior research
attending physician in the Division of Pediatric fellow and clinical instructor at UCSF
Hematology/Oncology at the Herman & Benioff Children’s Hospital Oakland; and
Walter Samuelson Children’s Hospital at Sinai Dr. Monica Davini, attending physician in
and Chairman of the Sinai Hospital Ethics the Division of Pediatric Hematology/
Committee, as well as attending at the Johns Oncology at Banner—University Medical
Hopkins Berman Institute of Bioethics. Center, Tucson Campus in Tucson, Arizona.
We are also extremely appreciative of These cases appear in Chapters 18, 15,
patient cases submitted by Dr. Christina and 11, respectively.
1 Approach to the
Anemic Child
Anemia is the condition in which the cell indices are very helpful in the diagnosis
concentration of hemoglobin or the red
­ and classification of anemia. They allow for
cell mass is reduced below normal. Anemia ­classification by the cell size (mean corpus­
results in a physiological decrease in the cular volume [MCV]), give the distribution
oxygen‐carrying capacity of the blood and of cell size (red cell distribution width
reduced oxygen supply to the tissues. Causes [RDW]), and may give important diagnos­
of anemia are increased loss or destruction tic clues if specific morphological abnor­
of red blood cells (RBCs) or a significant malities are present (e.g., sickle cells, target
decreased rate of production. When evalu­ cells, and spherocytes). The MCV, RDW,
ating a child with anemia, it is important and reticulocyte count are helpful in the
to determine if the problem is isolated to differential diagnosis of anemia. A high
one cell line (e.g., RBCs) or multiple cell RDW, or anisocytosis, is seen in stress
lines (i.e., RBCs, white blood cells [WBCs], erythropoiesis and is often suggestive of
or platelets). When two or three cell lines iron deficiency or hemolysis. A normal or
are affected, it may indicate bone marrow low reticulocyte count is an inappropriate
involvement (e.g., leukemia, metastatic dis­ response to anemia and suggests impaired
ease, and aplastic anemia), sequestration red cell production. An elevated reticulo­
(i.e., hypersplenism), immune deficiency, cyte count suggests blood loss, hemolysis,
or an immune‐mediated process (e.g., or sequestration.
hemolytic anemia and immune thrombocy­ The investigation of anemia requires
topenic purpura). the following steps:
1. The medical history of the anemic child
(Table 1.2), as certain historical points
Evaluation of anemia may 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, physi­ with particular attention to acute and
cal examination, and laboratory assessment chronic effects of anemia.
(see 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, reticu­
age (Table 1.1). The blood smear and red locyte count, and RBC morphology.

Handbook of Pediatric Hematology and Oncology – Children’s Hospital & Research Center Oakland,
Third Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
© 2021 John Wiley & Sons Ltd. Published 2021 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 Red cell aplasia Reticulocytosis
Chronic blood loss Malignancy Post splenectomy
Thalassemia, α or β Infection Liver disease
Hemoglobin E Renal failure Aplastic anemia
Lead toxicity^ Hypersplenism Bone marrow failure
Chronic disease/Infection Drugs syndromes
Severe malnutrition Acute blood loss Hypothyroidism
Sideroblastic anemia Hemolysis Down syndrome
Red cell enzyme deficiency Preleukemia
Red cell membrane defects Syndromes with elevated Hgb F
Megaloblastic anemia
Aplastic anemia
Folic acid deficiency
DBA, TEC
Malabsorption, drugs
Vitamin B12 deficiency
Evaluations Dietary
To Consider Pernicious anemia

Iron Studies Reticulocyte count Liver function tests


RDW, FEP, ferritin, Complete metabolic panel Thyroid function tests
transferrin saturation, TIBC Red cell enzyme panel Hemoglobin electrophoresis
Hemoglobin electrophoresis G6PD, Pyruvate kinase Folic acid level
Lead level Osmotic fragility/Ektacytometry Red cell
Family studies Coombs test (DAT) Serum
Check newborn screen Hemoglobin electrophoresis Vitamin B12 level
Oral iron challenge Bone marrow aspirate 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 dehydro­
genase deficiency; DAT, direct antiglobulin test).
*Refer to Table 1.1 for age‐based normal values.
^Microcytosis with lead toxicity has been noted secondary to concomitant iron deficiency; see text.

Consideration should also be given to the


Interventions
WBC and platelet counts as well as their
respective morphologies.
Oral iron challenge
4. Determination of an etiology of the ane­
An oral iron challenge may be indicated in
mia by additional studies as needed (see
the patient with significant iron depletion,
Figures 1.1–1.3).
as documented by moderate‐to‐severe
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 mo 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; Penttilä I, et al. Suomen Lääkärilehti 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.

a­ nemia and deficiencies in circulating and Administration of an oral iron challenge


storage iron forms (such as an elevated is quite simple: first, draw a serum iron level;
total iron‐binding capacity [TIBC], low second, administer a dose of iron (3 mg/kg
serum iron, low transferrin saturation, and elemental iron) orally; third, draw another
low ­ferritin). Iron absorption is impaired in serum iron level 30–60 minutes later. The
­certain chronic disorders (autoimmune dis­ serum level is expected to increase by at
eases such as systemic lupus erythematosus, least 100 mcg/dL if absorption is adequate.
peptic ulcer disease, ulcerative colitis, and The oral iron challenge is a quick and easy
Crohn’s disease), by certain medications method to assess appropriateness of oral
(antacids and histamine‐2 blockers), and iron to treat iron deficiency—a safer,
by environmental factors such as lead cheaper, yet equally efficacious method of
toxicity. treatment as parenteral iron.
Indications for an oral iron challenge
include any condition in which a poor Parenteral iron therapy
response to oral iron is being questioned, such Due to the potential risks of older par­
as in: noncompliance, severe anemia sec­ enteral iron preparations (specifically
ondary to dietary insufficiency (excessive high‐molecular‐weight iron dextran),
milk intake), and ongoing blood loss. practitioners have moved to newer (and
­
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 the abovementioned
factors or maternal iron deficiency
Ethnicity
African‐American Hgb S, C; α‐ and β‐thalassemia; G6PD deficiency
Mediterranean α‐ and β‐thalassemia; G6PD deficiency
Southeast Asian α‐ and β‐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 (also predisposed
to iron deficiency)
Sex
Male X‐linked enzymopathies (i.e., G6PD deficiency)
Early jaundice (<24 h of age) Isoimmune, infectious
Persistent jaundice Suggests hemolytic anemia
Diet (Usually >6 mo unless history of
prematurity)
Pica behavior Lead toxicity, iron deficiency
Excessive milk intake Iron deficiency
Macrobiotic/vegan diets Vitamin B12 deficiency
Goat 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

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, decreased red cell production
Parvovirus Anemia with reticulocytopenia

Abbreviations: EPO, erythropoietin; TORCH, toxoplasmosis, other, rubella, cytomegalovirus


(CMV), herpes simplex virus; G6PD, glucose‐6‐phosphate dehydrogenase deficiency.
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 (i.e.,
malar and maxillary bones thalassemia major, congenital hemolytic
anemia)
Icteric sclerae Congenital hemolytic anemia, hyperhemolytic
crisis associated with infection (i.e., red cell
enzyme deficiencies, red cell membrane
defects, thalassemias, hemoglobinopathies),
autoimmune hemolytic anemia
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,
hematologic malignancies, portal
hypertension
Extremities Radial limb dysplasia Fanconi anemia
Spoon nails Iron deficiency
Triphalangeal thumbs Red cell aplasia

perceived safer) f­ ormulations including fer­ in pediatric patients, total iron replacement
ric gluconate and iron sucrose. Three addi­ is feasible in 1–2 doses of LMW iron
tional compounds have been approved, one ­dextran and has been shown safe. Refer to
only in Europe (iron isomaltoside) and two the Formulary for calculation of LMW iron
in the United States (ferumoxytol and fer­ dextran dosing.
ric carboxymaltose). These newer agents Severe allergic reactions can occur with
have the potential benefit of total dose iron dextran and therefore a LMW product
replacement in a very short and single infu­ should be preferentially utilized. A test dose
sion as compared to ferric gluconate and (10–25 mg) should be given prior to the
iron sucrose which require multiple doses. first dose with observation of the patient
Low‐molecular‐weight (LMW) iron dextran for 30–60 minutes prior to administering
is approved as a total dose infusion for the remainder of the dose. A common side
adults in Europe but not the United States. effect is mild to moderate arthralgias the
Due to the smaller dose generally required day after drug administration, especially
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
at 1 week Review smear Hospitalization
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 β-thalassemia major
Iron malabsorption Hemoglobinopathy
Iron studies Sickle cell disease
Oral iron challenge TEC
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 proto­
porphyrin; TIBC, total iron‐binding capacity; DAT, direct antiglobulin test; IBD, inflammatory bowel
disease).

in patients with autoimmune disease. by a maximum dose beyond which there


Acetaminophen frequently alleviates the is increased risk of adverse events.
arthralgias. Iron dextran is contraindicated Ferumoxytol and ferric carboxymaltose
in patients with rheumatoid arthritis. have both received approval by the United
Iron sucrose or ferric gluconate should States Food and Drug Administration for the
be considered in patients for whom multiple treatment of iron deficiency anemia as well
doses are more feasible. Both are limited as renal insufficiency in adults. Both have
Approach to the Anemic Child 7

Low hemoglobin concentration

Low
Elevated or normal
reticulocyte
reticulocyte count Infection
count
TORCH
Coombs test (DAT) Congenital hypoplastic anemia
Transcobalamin II deficiency

Positive Immune hemolytic anemia


ABO incompatibility
Rh incompatibility
Minor blood group incompatibility
Negative

Microcytic

α-thalassemia syndrome
Normocytic
Macrocytic Chronic intrauterine blood loss
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‐phosphate dehydrogenase deficiency; TORCH, toxoplasmosis, other, rubella,
cytomegalovirus, herpes simplex virus).

the benefit of total dose replacement given regarding utilization in pediatric patients
as a rapid infusion due to slow release of are limited regarding appropriate per kg
elemental iron. Neither is immunogenic and dosage, although the infusions appear safe and
therefore no test dose is required. Studies therefore will likely replace use of iron
8 Chapter 1

sucrose and ferric gluconate in the future


Transfusion therapy
due to the convenience of single dose
replacement. Refer to the Formulary for
Children with very severe anemia (i.e.,
dosing parameters for these products.
hgb < 5 g/dL) may require treatment with
red cell transfusion, depending on the
Erythropoietin underlying disease and baseline hemo­
globin status, duration of anemia, rapidity
Recombinant human erythropoietin (EPO) of onset, and hemodynamic stability. The
stimulates proliferation and differentiation pediatric literature is scarce as to the best
of erythroid precursors, with an increase in method of transfusing such patients.
heme synthesis. This increased prolifera­ However, it appears to be common prac­
tion creates an increased demand in iron tice to give slow transfusions to children
availability and can result in a functional with cardiovascular compromise (i.e., gal­
iron deficiency if not given with iron lop rhythm, pulmonary edema, excessive
therapy. tachycardia, and poor perfusion) while
Indications for EPO include end‐stage being monitored in an ICU setting.
renal disease, anemia of prematurity, ane­ Transfusions are given in multiple small
mia of chronic disease, anemia associated volumes, sometimes separated by several
with treatment for AIDS, and autologous hours, with careful monitoring of the vitals
blood donation. EPO use for the treatment and fluid balance. For those children who
of chemotherapy‐induced anemia remains have gradual onset of severe anemia, with­
controversial and is not routinely recom­ out cardiovascular compromise, continu­
mended in pediatric patients (see ous transfusion of 2 mL/kg/h has been
Chapter 25). EPO has also been used in shown to be safe and result in an increase
autoimmune hemolytic anemia with low in the hematocrit of 1% for each 1 mL/kg
production as well as in chemotherapy‐ of transfused packed RBCs (based on RBC
induced anemia when the family has reli­ storage method). The hemoglobin should
gious beliefs that preclude transfusion. be increased to a normal value to avoid
The use of EPO in this latter setting may further cardiac compromise (i.e., hgb
­
reduce transfusion exposures but has not 8–12 g/dL). Again, the final endpoint may
been validated. be dependent on several factors including
The most common side effect of EPO nature of anemia, ongoing blood loss or lack
administration is hypertension, which may of production, baseline hemoglobin, and
be somewhat alleviated with changes in the volume to be transfused. Care should be
dose and duration of administration. taken to avoid unnecessary exposure to mul­
Typical starting dose of EPO is 150 U/kg tiple blood donors by maximal use of the
three times a week (IV) or subcutaneous unit of blood, proper division of units in the
(SC). CBCs and reticulocyte counts are blood bank, and avoidance of opening extra
checked weekly. Higher doses, and more units for small quantities to meet a total vol­
frequent dosing, may be necessary. Response ume. See Chapter 5 for product preparation,
is usually seen within 1–2 weeks. Adequate ordering, and premedication. A posttransfu­
iron intake (3 mg/kg/day orally or inter­ sion hemoglobin can be checked if necessary
mittent parenteral therapy) should be pro­ at any point after the transfusion has been
vided to optimize efficacy and prevent iron completed. Waiting for “reequilibration” is
deficiency. anecdotal and unnecessary.
Approach to the Anemic Child 9

should be given with vitamin C (i.e., orange


Case studies for review
juice) and not milk to improve absorption. If
there is a low likelihood of iron deficiency, a
1. You are seeing a 1‐year‐old for their well‐
family history of thalassemia or sickle cell
child check in clinic. As part of routine
disease, or a suggestive newborn screen, an
screening, a fingerstick hemoglobin is
empiric trial of oral iron supplementation
recommended.
should not be performed. Similarly, if there
a. What questions in the history might
are signs that are consistent with a hemolytic
help screen for anemia?
process or a significant underlying disorder,
b. What about the physical examination?
further workup should be done. In these
Multiple questions in the history can be
cases, it would be correct to next perform a
helpful. Dietary screening for excessive milk
CBC. If there are concerns for sickle cell dis­
intake is important in addition to asking
ease or thalassemia, it would be reasonable
about intake of iron‐rich foods such as green
to also perform hemoglobin electrophoresis.
leafy vegetables and red meat. One should
If there are concerns for hemolysis, labs
also ask about pica behavior such as eating
including reticulocyte count, total bilirubin,
dirt or ice and include questions regarding
lactate dehydrogenase, and a direct Coombs
the age of the house to help screen for lead
should be performed. Finally, if there is con­
paint exposure and ingestion. Any sources
cern for a systemic illness such as leukemia,
of blood loss should also be explored includ­
a manual differential should be requested.
ing blood in the urine or stool as well as
Further workup for iron deficiency (ferritin,
­frequent gum or nose bleeding (more likely
TIBC) as well as lead toxicity (lead level)
in an older child). Finally, family history
could be included or deferred until the ane­
should be explored regarding anemia dur­
mia is better characterized utilizing the
ing pregnancy, previous history of iron
MCV and RDW on the CBC.
deficiency in siblings, and history of
­
hemoglobinopathies. 2. You are seeing a 15‐month‐old for a
Physical examination to search for ane­ routine well‐child check visit. The family
mia should be focused. Pallor, especially notes that the child has been well but
subconjunctival, perioral, and periungual, appears pale to them. You ask the same
should be checked. Tachycardia, if present, questions as reviewed in case one, and the
would be more consistent with acute anemia family notes a recent viral illness with no
rather than well‐compensated chronic ane­ evidence of hemolysis, as there has been no
mia. Splenomegaly, scleral icterus, and jaun­ history of dark‐colored urine or jaundice.
dice may point to an acute or chronic The dietary history is unremarkable and on
hemolytic picture. review, the 1‐year fingerstick hemoglobin
You do the fingerstick hemoglobin in was normal at 11.4 g/dL with a normal lead
clinic and it is 10.2 g/dL. The history is not level at that time. There has been no noted
suggestive of iron deficiency and the exam is diarrhea or blood loss, and the child is not
unremarkable. on any medications. On exam, the child is
c. What are the reasonable next steps? pale but well appearing with a normal heart
Depending on the prevalence of iron defi­ rate, respiratory rate, and blood pressure
ciency in your population, it would be rea­ without scleral icterus or other signs of
sonable at this point to give a 1‐month trial jaundice. There is no lymphadenopathy or
of oral iron therapy. The family should be hepatosplenomegaly. There are no pete­
counseled that oral iron tastes bad and chiae or bruising.
10 Chapter 1

a. What are some common diagnostic viral or secondary to TEC. Coombs is vital
considerations? to help determine if this is an underlying
b. What are the next laboratory steps? warm autoimmune hemolytic anemia, not­
Microcytic causes of anemia, notably iron ing that the Coombs will not be positive in
deficiency anemia and secondary lead toxic­ all cases. Complete metabolic panel will
ity as well as thalassemia syndromes, have look at many important aspects including
generally been ruled out with a history of a renal function (to rule out aHUS), as well
normal recent hemoglobin as well as a reas­ as bilirubin and AST which may be ele­
suring dietary history. Congenital causes of vated along with the LDH if there is ongo­
anemia, especially Diamond–Blackfan ane­ ing hemolysis. Similarly, the urinalysis will
mia, a pure red cell aplasia, have also been help rule out blood loss as well as hemoly­
ruled out with a previous normal hgb. sis. Parvovirus PCR is not vital given the
Megaloblastic anemias are also less likely to lack of clinical history, but this infection
develop in the preceding 3 months after a should remain in the differential diagnosis.
normal hemoglobin. Hemolytic anemia, Type and screen is important as the child
especially warm autoimmune hemolytic may require blood transfusion depending
anemia (AIHA), remains a possibility even on the level of anemia and the potential
without a clinical history of jaundice in presence of hemolysis.
patients with slow hemolytic rates. Many
Lab results include the following:
other diagnostic possibilities remain at this
point including viral suppression secondary 4.7 retic 11.9%
to infections such as parvovirus, syndromes 10.4 279
14.2
that may have bicytopenia or pancytopenia
in addition to the clinical presentation of Normal LDH, total, and indirect bilirubin
anemia including severe aplastic anemia Normal UA
and acute leukemias, as well as transient Negative direct Coombs
erythroblastopenia of childhood (TEC). Normal CMP
Atypical hemolytic uremic syndrome c. What is the likely diagnosis?
(aHUS) is an unlikely possibility, but should The elevated reticulocyte count in the set­
be in the differential diagnosis in addition to ting of a low hemoglobin should always
occult blood loss. make the practitioner first think of a hemo­
The initial laboratory workup should lytic anemia, either a warm antibody or cold
include labs which will help make the diag­ agglutinin. Yet, in this case, there are no
nosis and potentially treat the patient: other supporting laboratories for a hemo­
CBC/diff, reticulocyte count, Coombs test, lytic process. With a slow hemolytic process,
complete metabolic panel (CMP), LDH, it is possible that the UA, LDH, and indirect
type and screen, fecal occult blood, parvo­ bilirubin could be normal but the direct
virus PCR, and urinalysis (UA). CBC will Coombs should be helpful in the majority of
help evaluate the level of anemia noted warm antibody‐mediated cases. In the case
clinically as well as determine if there are of a cold agglutinin (usually associated with
more cytopenias which will help direct the Mycoplasma infection), it is possible there
differential diagnosis. The reticulocyte would be no other positive lab findings.
count will be helpful in determining if the The most likely diagnosis here is TEC in
bone marrow is responding correctly to the recovery phase. TEC occurs due to unclear
anemia, as it should be high with a hemo­ reasons and may present with concomitant
lytic process and suppressed if this is post­ neutropenia leading to workup for acute
Approach to the Anemic Child 11

lymphoblastic leukemia and aplastic anemia. 2. You are seeing a patient in the hospital
Patients will have a decrease in erythroid admitted at two months of age for sepsis
precursors in the bone marrow and therefore rule out secondary to high fever with asso­
a decrease in peripheral reticulocytes. TEC ciated decreased feeds and decreased urine
is an indolent process and thus patients will output. The child has a urinary tract infec­
generally present with a well‐compensated tion. A CBC is checked the hgb is 9.0 g/dl.
but often severe normocytic anemia. Spon­ There is no relevant family history, the
taneous recovery occurs with reticulocyto­ mother received good prenatal care and
sis, and thus TEC in the recovery phase can was not anemic during pregnancy and the
be mistaken for a hemolytic anemia. baby was born full term. What is the next
best step?
a. Empirically start iron therapy
Multiple choice questions b. Plan to repeat the CBC prior to
discharge
1. You are seeing a patient at their one year c. Plan to repeat the CBC in 1‐2 months
clinic visit and perform a finger stick hemo­ as an outpatient
globin. The hgb is 9.2 g/dl. The child has d. Provide reassurance to the family
been well without any recent illnesses or Explanation: Although anemia in a young
significant findings on exam. What is the infant with an underlying infection can be
next best step? due to bone marrow suppression, this level
a. Empirically start iron therapy of hemoglobin is normal for age. The physi­
b. Obtain further history ologic nadir of infancy occurs around 2‐3
c. Check a full CBC/diff months of age as fetal hemoglobin goes
d. Plan to repeat in one month away (fetal hemoglobin lives 60‐90 days as
e. Perform a lead level compared to hemoglobin A which lives
Explanation: It is first important to under­ 90‐120 days). A certain level of anemia
stand underlying risk factors for anemia. occurs prior to the bone marrow ramping
These could include a dietary intake of sig­ up production of hemoglobin A red blood
nificant milk consumption which would cells. Choice a. is incorrect as iron deficiency
lead to iron deficiency or a family history of would be unlikely in this age although can
thalassemia. Also it is important to ask occur in a very preterm infant or if the
about a bleeding history to determine if mother had severe anemia during preg­
there are ongoing losses. It important to ask nancy. Choice b. and c. are not necessary
about any history of jaundice or scleral since the hb is normal for age. The answer is d.
icterus which would point to a hemolytic
anemia as well as other inflammatory condi­ 3. You are seeing a patient in follow up. At
tions which could lead to anemia of chronic the one year visit the finger stick hemoglobin
disease. Choice a. would likely be correct was 9.4 g/dl. Given a nutritional history
after obtaining more history; choice c. is of significant milk intake you previously
reasonable in cases where the history is not decided to empirically start iron therapy at a
suggestive or the hgb does not increase with dose of 3 mg elemental iron daily and see the
empiric iron therapy; choice d. would not be child today, one month later for a repeat finger
correct without other interventions; choice e. stick hemoglobin. The repeat hemoglobin is
is important to do but lead toxicity itself now 10.2 g/dl. What is the next best step?
does not lead to anemia, rather it is concom­ a. Stop iron therapy
itant iron deficiency. The answer is b. b. Send a full CBC/diff
12 Chapter 1

c. Continue iron therapy and see the Explanation: As iron deficiency is the most
patient back in one month likely cause of anemia, an empiric trial of
d. Continue iron therapy and see the iron is the correct first step. Assessing com­
patient back in 2‐3 months pliance with iron therapy and nutritional
Explanation: Given the level of hemoglobin recommendations is very important in fol­
and nutritional history, iron deficiency is low up especially if the hgb is not improved.
the most likely diagnosis. Reticulocyte counts In many cases the family is unable to give
should increase in 1‐2 days after commence­ the iron well and the excessive milk intake
ment of iron therapy and hemoglobin continues. Assuming a reliable family as in
should rise within one week. A rise in hb in the case above, it is important to now check
one month is reassuring that the diagnosis a CBC/diff to further characterize the ane­
and treatment plan are correct. Choice a. is mia. It would also be important to test for
incorrect because it is important to continue ongoing losses at this point with a urinalysis
iron therapy for 2‐3 months after resolution and fecal occult blood test. Choice a. is
of anemia to replete liver iron stores; as this incorrect since iron therapy should show
child is still anemic it is too early to stop iron improvement in the hgb in 1 week and if
therapy. Choice b. is unnecessary at this that is not the case it is best to avoid iron
point since the hgb is appropriately increas­ overload by unnecessarily continuing iron.
ing with therapy but may be necessary at a Choice b. though correct is not as good a
later point if the anemia persists on iron choice as choice c. Choice d. is reasonable
therapy. Choice c. is reasonable although although it would be first important to
given the amount of time of iron therapy check for microcytosis on the full CBC—if
required to replete stores and the improve­ microcytic then it would be reasonable to
ment seen to date, it is not necessary to see test iron studies (ferritin, TIBC) in addition
the patient so frequently. The answer is d. to checking hemoglobin electrophoresis for
β‐thalassemia trait given the non‐respon­
4. You are seeing a patient in follow up. At siveness to iron therapy. The answer is c.
the one year visit the finger stick hemo­
globin was 9.4 g/dl. Given a nutritional his­ 5. You are seeing a 15-month-old for a rou­
tory of significant milk intake you previously tine clinic visit. The family notes the child
decided to empirically start iron therapy at a has been more tired and pale recently. There
dose of 3 mg elemental iron daily and see the was an antecedent viral illness about one
child today, one month later for a repeat fin­ month prior but the child is now otherwise
ger stick hemoglobin. The repeat hemo­ well. The child had a normal finger stick hb
globin is now 9.2 g/dl. The mother states at one year of age, eats a varied diet, has had
that the child is taking the iron well, with no noted blood loss and had no signs of
vitamin C, and she has decreased milk jaundice. The child is pale and tachycardic
intake to 16 oz (480 ml) on average in a but otherwise well appearing without LAD
24 hour period. What is the next best step? or HSM. A finger stick hemoglobin is 5.4 g/
a. Continue iron therapy for another dl. Follow up CBC shows a normocytic ane­
month and recheck hb at that time mia with normal WBC/diff and platelet
b. Check a CBC/diff count. The retic count is very low. What is
c. Stop iron therapy and check a CBC/ the most likely diagnosis?
diff a. Transient erythroblastopenia of
d. Stop iron therapy and check iron childhood
studies in addition to a CBC/diff b. Iron deficiency anemia
Approach to the Anemic Child 13

c. Hemolytic anemia with a normocytic anemia but typically


d. Anemia of chronic disease ­presents with additional cytopenias. Often
e. Aplastic anemia these patients have macrocytosis. The
Explanation: Transient erythroblastopenia answer is a.
of childhood (TEC) is a common etiology of
normocytic anemia in infants and is due to
unclear causes, possibly infection triggered Suggested reading
bone marrow suppression. The normal
MCV in addition to the low reticulocyte Camaschella, C. (2015). Iron deficiency: new
count in a well appearing child is suggestive insights into diagnosis and treatment.
of this diagnosis. TEC in recovery phase Hematology Am. Soc. Hematol. Educ. Program
presents with a high reticulocyte and can be 1: 8–13.
confused with hemolytic anemia. Choice b. DeLoughery, T.G. (2014). Microcytic anemia.
N Engl. J. Med. 371: 1324–1331.
is less likely since the anemia is normocytic
Kaneva, K., Chow, E., Rosenfeld, C.G., and Kelly,
and not microcytic. Choice c. is unlikely M.J. (2017). Intravenous iron sucrose for chil­
without a history of jaundice, scleral icterus dren with iron deficiency anemia. J. Pediatr.
or dark urine and with a low reticulocyte Hematol. Oncol. 39: e259–e262.
count. Choice d., although possible, is not Wang, M. (2016). Iron deficiency and other types
suggested by a normal history without other of anemia in infants and children. Am. Fam.
medical diagnoses. Choice e. is also possible Physician 93: 270–278.
2 Hemolytic Anemia

Red blood cells (RBCs) normally live for is quite variable, with most severe cases pre-
about 100–120 days in the circulation. senting in the newborn period or early
Hemolytic anemia results from a reduced childhood and milder cases presenting in
red cell survival due to increased destruc- adulthood.
tion. To compensate for a reduced RBC life Several membrane protein defects are
span, the bone marrow increases its output responsible for HS, and most result in the
of red cells, a response mediated by erythro- instability of spectrin, one of the major red
poietin. Destruction of red cells can be cell skeletal membrane proteins. Structural
intravascular (within the circulation) or changes that result as a consequence of pro-
extravascular (by phagocytic cells of the tein deficiency lead to membrane instability,
bone marrow, liver, or spleen). Hemolytic loss of surface area, abnormal membrane
anemia may be inherited (i.e., thalassemias, permeability, and decreased red cell deform-
hemoglobinopathies, red cell enzyme defi- ability. Metabolic depletion accentuates the
ciencies, or membrane defects) or acquired defect in HS cells, which accounts for an
(immune‐mediated, associated with infec- increase in osmotic fragility after a 24‐hour
tion, or medication‐related). It can be incubation of whole blood at 37°C. The
chronic or acute. Some types of low‐grade splenic sinusoids prevent passage of nonde-
chronic hemolytic anemias can have acute formable spherocytic red cells. This explains
exacerbations, such as a child with glu- the occurrence of splenomegaly in HS and
cose‐6‐phosphate dehydrogenase (G6PD) the therapeutic effect of splenectomy.
deficiency with an exposure to fava beans or Patients with HS have a mild‐to‐mod-
other oxidative stress. erate chronic hemolytic anemia. Red cell
indices reveal a normal to low mean cor-
puscular volume (MCV) depending on the
Red cell membrane disorders number of microspherocytes. Cellular
dehydration increases the mean corpuscu-
Hereditary spherocytosis (HS) is the most lar hemoglobin concentration (MCHC)
common congenital red blood cell mem- characteristically >36%, which can be a
brane disorder. The typical patient with HS helpful diagnostic clue. The red cell distri-
has intermittent jaundice, and hemolytic or bution width (RDW) is elevated because of
red cell aplastic episodes associated with the variable presence of microspherocytes
viral infections, splenomegaly, and chole- and reticulocytes in proportion to the
lithiasis. However, the clinical presentation degree of hemolysis. The peripheral blood

Handbook of Pediatric Hematology and Oncology – Children’s Hospital & Research Center Oakland,
Third Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
© 2021 John Wiley & Sons Ltd. Published 2021 by John Wiley & Sons Ltd.
16 Chapter 2

smear can be highly suggestive with the For pediatric patients who have excessive
presence of spherocytes, although this can splenic size, an additional consideration for
be a normal finding in the patient with splenectomy is to diminish the risk of trau-
severe anemia and a resultant reticulocy- matic splenic rupture. The risks of splenec-
tosis. Osmotic fragility tests and ektacy- tomy must be considered before any clinical
tometry studies show characteristic decision is made regarding the procedure.
findings for HS, with increased red cell Red cell survival returns to normal val-
f ragility in hypotonic environments.
­ ues after splenectomy unless an accessory
Confirmatory testing is done with RBC spleen develops. Although an increased
band 3 protein reduction. Band 3 is the number of spherocytes can be seen in the
most abundant transmembrane protein peripheral blood after splenectomy and the
found in the erythrocyte. This study is osmotic fragility is more abnormal, the
done by flow cytometry utilizing a fluores- hemoglobin value is normal. Platelet counts
cent dye to bind the band 3 protein. It is frequently increase to more than 1000 × 109/l
93% specific for the diagnosis of HS, immediately after splenectomy, but return
though other diseases can test positive to normal levels over several weeks. No
(i.e., congenital dyserythropoietic anemia therapeutic interventions are required for
type II, Southeast Asian ovalocytosis, postsplenectomy thrombocytosis in patients
hereditary pyropoikilocytosis). with HS.
As with other hemolytic anemias, To minimize the risk of sepsis due to
affected individuals are susceptible to hypo- Haemophilus influenza and Streptococcus
plastic crises during viral infections. Human pneumoniae, the splenectomy procedure
parvovirus B19, a frequent pathogen and the (when necessary) is often postponed until
organism responsible for erythema infectio- after the child’s fifth or sixth birthday and
sum (fifth disease), selectively invades when fully immunized. Patients should be
erythroid progenitor cells and may result in immunized against these organisms in addi-
a transient arrest in red cell proliferation tion to Neisseria meningitidis prior to sple-
(see Chapter 1). Recovery begins within nectomy and receive penicillin prophylaxis
7–10 days after infection and is usually com- following the procedure. The increase in
pleted by 4–6 weeks. If the initial presenta- penicillin‐resistant strains of S. pneumoniae
tion of a patient with HS is during an aplastic has raised questions regarding the use of
crisis, a diagnosis of HS might not be con- prophylactic penicillin. No studies have
sidered because the reticulocyte count will determined the frequency of this problem in
be low and the peripheral blood smear may children receiving prophylactic penicillin
be nondiagnostic. Anemia may also be more after splenectomy. Dietary supplementation
marked in patients with any underlying with folic acid (1 mg/day) is recommended
hemolytic disorder. A family history of HS due to high cellular turnover.
should be explored; if it is positive, the Paroxysmal nocturnal hemoglobinuria
patient should be evaluated for HS after (PNH) is an acquired hemolytic anemia
recovery from the aplastic episode. with periods of increased hemolysis in addi-
Splenectomy is often considered for tion to risk of venous thrombosis and is
patients who have severe hemolysis requir- associated with aplastic anemia. The red
ing transfusions or repeated hospitalization. blood cell backbone has a deficiency in gly-
In patients with mild hemolysis, the deci- cosylphosphatidylinositol (GPI) antigen
sion to perform splenectomy should be structure leading to complement activation
delayed; in many cases, it is not required. secondary to deficiency of CD55 or CD59.
Hemolytic Anemia 17

patient’s clinical course. Unlike HS patients,


Red cell enzyme deficiencies
PK‐deficient patients, although they
improve after splenectomy, do not have
Glucose is the primary metabolic substrate
complete correction of their hemolytic ane-
for the red cell. Because the mature red cell
mia. As with all hemolytic anemias, these
does not contain mitochondria, it can
patients should have dietary supplementa-
metabolize glucose only by anaerobic mech-
tion with folic acid (1 mg/day) to prevent
anisms. The two major metabolic pathways
megaloblastic complications associated with
within the red cell are the Embden–
relative folate deficiency. Immunization
Meyerhof pathway (EMP) and the hexose
against H. influenza, S. pneumonia, and N.
monophosphate shunt.
meningitidis should be given, as well as life-
Red cell morphological changes are min-
long penicillin prophylaxis in the splenecto-
imal in patients with red cell enzyme defi-
mized patient.
ciency involving the EMP. Red cell indices
G6PD deficiency is the most common
are usually normocytic and normochromic.
X‐linked red cell enzyme deficiency, with par-
The reticulocyte count is elevated in propor-
tial expression in the female population and
tion to the extent of hemolysis. Because
full expression in the affected male popula-
many enzyme activities are normally
tion. The distribution of G6PD deficiency
increased in young red cells, a mild defi-
is worldwide, with the highest incidence
ciency in one of the enzymes may be
in Africans and African‐Americans.
obscured by the reticulocytosis.
Mediterraneans, American Indians, Southeast
Pyruvate kinase (PK) deficiency is the
Asians, and Sephardic Jews are also affected.
most common enzyme deficiency in the
In African‐Americans, 12% of the male popu-
EMP. The inheritance pattern of this disor-
lation has the deficiency, 18% of the female
der is autosomal recessive. Homozygotes
population is heterozygous, and 2% of the
usually have hemolytic anemia with spleno-
female population is homozygous. In
megaly, whereas heterozygotes are usually
Southeast Asians, G6PD deficiency is found
asymptomatic. The disorder is found world-
in approximately 6% of the male population.
wide, although it is most common in
Most likely, the prevalence of this enzyme
Caucasians of Northern European descent.
abnormality confers resistance to malaria,
The range of clinical expression is variable,
thus its geographic distribution.
from severe neonatal jaundice to a fully
Many variants of G6PD deficiency are
compensated hemolytic anemia. Anemia is
known and have been characterized at the
usually normochromic and normocytic, but
biochemical and molecular levels. A variant
macrocytes may be present shortly after a
found in Mediterraneans is associated with
hemolytic crisis, reflecting erythroid hyper-
chronic hemolytic anemia. Other variants
plasia and early release of immature red
are associated with an unstable enzyme that
cells. The osmotic fragility of red cells is
has normal levels in young red cells. These
normal to slightly reduced. Diagnosis is
variants result in hemolysis only in associa-
confirmed by a quantitative assay for PK, by
tion with an oxidant challenge (as found in
the measurement of enzyme kinetics and
African‐Americans). In some cases of G6PD
glycolytic intermediates, and by family
deficiency, hemolysis may be triggered by
studies.
the oxidant intermediates generated during
Splenectomy is a therapeutic option for
viral or bacterial infections or after inges-
PK‐deficient patients. As with HS, the deci-
tion of oxidant compounds. Shortly after
sion should be made on the basis of the
exposure to the oxidant, hemoglobin is
18 Chapter 2

oxidized to methemoglobin and eventually chronic hemolysis, dietary supplementation


denatured, forming intracellular inclusions with folic acid (1 mg/day) is recommended.
called Heinz bodies that attach to the red Use of vitamin E, 500 mg/day, may improve
cell membrane. This portion of the mem- red cell survival in patients with chronic
brane may be removed by reticuloendothe- hemolysis.
lial cells resulting in a “bite” cell that has a
shortened survival owing to its loss of mem-
brane components. To compensate for Autoimmune hemolytic anemia
hemolysis, red cell production is increased
and thus the reticulocyte count is increased. In addition to intrinsic causes of hemolytic
Individuals with the Mediterranean or anemia, patients may develop an autoanti-
Asian forms of G6PD deficiency, in addition body or alloantibody toward their red blood
to being sensitive to infections and certain cells. The underlying cause for this antibody
drugs, often have a chronic, moderately formation is often idiopathic or precipitated
severe anemia, with non‐spherocytic red by a secondary condition including drugs,
cells and jaundice. Hemolysis usually starts infection, autoimmune diseases, or an onco-
in early childhood. Reticulocytosis is pre- logic process. A positive direct antiglobulin
sent and can increase the MCV. test (DAT, direct Coombs) is pathogno-
When a hemolytic crisis occurs in G6PD monic for immune‐mediated hemolysis
deficiency (or favism), pallor, scleral icterus, with the appropriate clinical and laboratory
hemoglobinemia, hemoglobinuria, and findings (i.e., jaundice, scleral icterus, ele-
splenomegaly may be noted. Plasma hapto- vated bilirubin, and anemia with reticulocy-
globin and hemopexin concentrations are tosis). Fortunately, many 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. However, AIHA may
the fragmented bite cells and polychromat- be a presenting feature in chronic immuno-
ophilic cells. Red cell indices may be nor- logic or hematologic conditions including
mal. Special stains can detect Heinz bodies autoimmune lymphoproliferative disease,
in the cells during the first few days of common variable immune deficiency, and
hemolysis. systemic lupus erythematosus.
A diagnosis of G6PD deficiency should In the DAT, the patient’s erythrocytes are
be suspected based on family history, eth- washed and then incubated with specific
nicity, laboratory features, physical findings, antiglobulin antisera (usually anti‐IgG and
and recent exposure to oxidants with result- anti‐C3d). Agglutination indicates a posi-
ant acute hemolysis. The diagnosis is con- tive test confirming the presence of antibod-
firmed by a quantitative enzyme assay or by ies or complement attached to the surface of
molecular analysis of the gene. Since reticu- the patient’s red blood cells. In patients with
locytes may have a normal level of G6PD severe immune‐mediated hemolytic ane-
enzyme activity, screening tests during acute mia, the DAT is often strongly positive,
hemolysis may be falsely elevated; therefore, although the strength of the reaction does
it is important to test once the hemolytic cri- not always correlate to the severity of the
sis has ended and the patient again has disease. Similarly, up to 80% of patients will
mature red blood cells. Treatment is directed have antibodies in the serum as well, meas-
toward supportive care during the acute ured by the indirect Coombs (indirect anti-
event and counseling regarding prevention globulin test, IAT). In the IAT, test
of future hemolytic crises. In patients with erythrocytes are incubated with donor
Hemolytic Anemia 19

serum, washed, and then incubated with therapies. Cold agglutinins are more likely in
specific antiglobulin antisera. Agglutination older patients though tend to be acute in chil-
again indicates a positive test indicating dren and chronic in adults.
antibody in the patient’s sera against a for- Finally, also secondary to infection, chil-
eign red cell antigen. Of note, patients with- dren may develop paroxysmal cold hemo-
out symptoms of hemolysis may have a globinuria, a transient condition which is
positive DAT or IAT; therefore, screening is often unrecognized with complement acti-
only recommended in the setting of clinical vation rather than IgG or IgM. This is spe-
and laboratory signs of hemolysis. In cific to the P antigen on the RBC membrane.
approximately 5–10% of cases, patients may Diagnosis is made in the DAT negative
have an AIHA with a negative DAT. patient with serum antibody demonstrated
The initiation of autoimmunity is poorly by the Donath–Landsteiner test. The major-
understood. Viral syndromes are often pro- ity of patients have self‐resolution though
posed as a culprit, although causation has corticosteroids may be required.
been hard to prove. A majority of cases of
AIHA in pediatrics are due to “warm” anti-
bodies, so named because they react at Hemolytic disease of the
37 °C. These are often secondary to a viral newborn
syndrome, although patients with an under-
lying autoimmune disease or oncologic pro- Intrinsic causes of hemolytic anemia can
cess can also present with a warm AIHA. present as jaundice in the newborn period.
Immune hemolytic anemia may also be seen These syndromes must be differentiated
after allogeneic transplant, generally sec- from hemolytic disease of the newborn
ondary to autoantibodies formed by donor (HDN), in which alloimmunization in the
cells against donor RBCs. The formation of mother occurs due to foreign RBC antigens
IgG antibodies leads to extravascular hemol- from the fetus. RBC antigens can either be
ysis in which pieces of the red cell mem- major (ABO) or minor (Rh, Kell, Duffy,
brane are sequentially removed during etc.). For ABO hemolytic disease, typically
passages through the spleen. Patients may the mother is type O and the fetus is type A;
also develop DAT positive hemolytic ane- anti‐A antibodies subsequently produced by
mia and immune‐mediated thrombocyto- the mother then traverse the placenta lead-
penia (Evans syndrome). ing to hemolytic anemia in the fetus.
Patients may also develop AIHA second- RhoGAM® (Rho[D] immune globulin) has
ary to cold agglutinins, often secondary to virtually eliminated hemolytic disease in the
infection with Mycoplasma, and occurs sec- Rh‐negative (D‐negative) mother with a
ondary to IgM and complement (C3d) rather Rh‐positive (D‐positive) fetus, although is
than IgG. IgM autoantibodies react with poly- still possible in the mother not receiving
saccharide antigens on the RBC surface, spe- prenatal care. Many cases of HDN are now
cifically I/i heavy chains. IgM binding occurs due to other minor RBC antigens with vary-
at lower temperatures in the periphery lead- ing levels of clinical severity. AIHA in the
ing to acrocyanosis. Intravascular hemolysis is mother can also lead to HDN. In this case,
rare with complement fixation leading to maternal IgG antibodies traverse the pla-
removal of RBCs in the liver. Corticosteroids centa and are transferred to the fetus. If the
are generally ineffective in cold agglutinin mother is DAT positive but does not have
syndrome; intravenous immune globulin clinical signs of hemolytic anemia, there is
(IVIG) and avoidance of the cold are first‐line usually no risk to the fetus.
20 Chapter 2

●● Anemia unresponsive to iron


Microangiopathic hemolytic
supplementation
anemia
●● Medications

●● Environmental exposures
Microangiopathic hemolytic anemias are
●● Ethnicity
due to extracorpuscular abnormalities and
●● Dietary history
are not associated with antibody formation.
The physical exam should be complete,
Causes include disseminated intravascular
but focused on:
coagulation (DIC), thrombotic thrombocy-
●● Skin color (pallor, jaundice, and icteric
topenic purpura/hemolytic uremic syn-
sclerae)
drome (TTP/HUS), transplant‐associated
●● Facial bone changes (extramedullary
microangiopathy, preeclampsia, malignant
hematopoiesis)
hypertension, valvular abnormalities, and
●● Abdominal fullness and splenomegaly
march hemoglobinuria. In these cases, red
The laboratory evaluation includes:
blood cells travel through damaged blood
●● Complete blood count, RBC indices, and
vessels or heart valves or are damaged by the
reticulocyte count
formation of an intravascular fibrin mesh
●● Peripheral blood smear (assess for frag-
due to hypercoagulability, leading to frag-
mented forms or evidence of inherited anemia
mentation (e.g., schistocytes) and intravas-
with specific morphological abnormalities)
cular hemolysis. In the case of TTP,
●● Bilirubin, AST, lactate dehydrogenase
ADAMTS13 is a useful evaluation as defi-
(LDH)
ciency can occur in hereditary TTP second-
●● Coombs test, direct and indirect (to exclude
ary to mutation and in acquired TTP
antibody‐mediated red cell destruction)
secondary to autoantibodies.
●● Urinalysis (for heme, bilirubin)
Transplant‐associated microangiopathy
●● Free plasma hemoglobin, haptoglobin
is increasingly being recognized, often sec-
●● Parvovirus PCR (if history is suspicious)
ondary to calcineurin inhibitors in alloge-
Specific tests for diagnosis may include:
neic transplant, though it has also been
●● Osmotic fragility
noted in patients after autologous transplan-
●● Ektacytometry
tation, likely secondary to irradiation,
●● RBC band 3
multiagent chemotherapy, or viral reactiva-
●● Red cell enzyme defects (G6PD and PK)
tion leading to endothelial injury.
●● Red cell membrane defects (HS)

●● CD 55/59

Evaluation ●● ADAMTS13

The osmotic fragility test is used to meas-


The evaluation of hemolytic anemia includes ure the osmotic resistance of red cells. Red
a thorough history assessing for evidence of cells are incubated under hypotonic condi-
chronic hemolytic anemia and possible pre- tions, and their ability to swell before lysis is
cipitants of an acute event (see Figure 2.1). determined. The osmotic fragility of red cells
The family history is equally important is increased when the surface area to volume
and questions to ask include: ratio of the red cells is decreased, as in hered-
●● History of newborn jaundice itary spherocytosis, in which membrane
●● Gallstones instability results in membrane loss and
●● Splenomegaly or splenectomy decreased surface area. Conversely, osmotic
●● Episodes of dark urine and/or yellow fragility is decreased in liver disease, as the
skin/sclerae ratio of the red cell surface area to volume is
Hemolytic Anemia 21

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

RBC enzyme studies


Consider
Ceruloplasmin
CD 55/59
DIC
Coagulation Family studies
Hemoglobin Enzyme deficiency
screen Osmotic fragility or
(fibrinogen, electrophoresis PK deficiency
ektacytometry
PT, PTT, FDP) G6PD deficiency
Platelet count Wilson disease
Blood cultures PNH
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 anti-
globulin test; ANA, anti‐nuclear antibody; DIC, disseminated intravascular coagulation; PT, prothrom-
bin 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; PNH, paroxysmal nocturnal hemoglobinuria).

increased. Ektacytometry measures the In chronic hemolysis, such as that associated


deformability of red cells subjected simulta- with hereditary spherocytosis, splenectomy
neously to shear stress and osmotic stress. is often recommended to decrease the
degree of splenic destruction and level of
anemia and to decrease the incidence of bili-
Treatment rubin gallstones. This therapy must be now
weighed against the potential long‐term
Determination of a treatment plan relies on complications of splenectomy including risk
identification of the underlying cause of the for infection, thrombosis, and pulmonary
anemia and the degree of acute hemolysis. hypertension. In other forms of inherited
22 Chapter 2

anemias in which the hemolysis is more sig- fatigued over the last few days with a red
nificant and even life‐threatening, such as color to the urine. Fingerstick hemoglobin
thalassemia or some forms of enzymopa- at the pediatrician’s office reveals a hemo-
thies, chronic transfusion therapy is recom- globin of 5 g/dl prior to transfer to the ED.
mended. Other general measures include On the basis of this history and hemoglobin,
folic acid replacement due to high cell turn- it appears that the child is suffering from a
over, avoidance of oxidant chemicals and hemolytic anemia.
drugs, and iron chelation therapy as indi- a. What initial lab studies will help con-
cated for transfusion‐related iron overload. firm the diagnosis and also help with the
Immune hemolytic anemias resulting in initial treatment plan?
acute, symptomatic anemia require more Initial lab studies should include a complete
immediate and aggressive therapy. The blood count with reticulocyte count. The
underlying disease, if present and identifiable, reticulocyte count is an important first step
warrants treatment. Additionally, the use of to confirm that the patient is undergoing
corticosteroids in high doses is frequently hemolysis, which should present with a low
necessary (starting with prednisone at hemoglobin and a resultant increase in the
2–4 mg/kg/day). Very high doses for 2–4 days reticulocyte count. A low reticulocyte count
(up to 10 mg/kg/day [max 1 g]) may be neces- in this setting should lead to consideration
sary prior to decreasing back to 2 mg/kg/day. of alternative diagnoses of decreased red
Splenectomy and immunosuppressive drugs cell production, such as viral suppression
(such as rituximab) have also been successful (although one would not expect hemolysis).
in steroid‐resistant disease. Immune globulin A complete metabolic panel as well as lac-
infusions do not contribute to resolution of tate dehydrogenase (LDH) should be done
the disease, but should be considered in to ensure that the patient is actually suffer-
patients with severe hemolysis who are ing from jaundice (elevated total bilirubin)
requiring transfusion or are having a poor and hemolysis (elevated LDH and AST). A
response to transfusion. Plasmapheresis can DAT/IAT is an important first step to deter-
be considered in patients with severe IgG‐ mine if the patient has an immune or non-
induced AIHA and should always be com- immune hemolytic anemia. If the DAT is
bined with an immune suppressive drug positive, the blood bank should be asked to
to ensure both antibody production and elute the antibody to determine if this is
­antibody titer reduction. Microangiopathic IgM, IgG, or possibly complement-medi-
hemolytic anemias can also be severe and ated hemolysis.
life‐threatening. Treatment should again first The patient is noted to have a hemo-
be directed toward the primary disorder to globin of 4.6 g/dl with 12.6% reticulocytes.
remove the cause of trauma, if possible. One should first determine if the patient
Transfusions are frequently necessary and is having an appropriate bone marrow
splenectomy may be needed in some patients response to anemia by calculating the retic-
with severe hypersplenism. ulocyte index (RI):

RI Reticulocyte count %
Case studies for review
current hemoglobin
1. You are seeing a 6-year-old child in the expected hemoglobin
emergency department. The family notes
that the child has been jaundiced and In this case, the RI is 12.6% × (4.6/13) = 4.5.
Hemolytic Anemia 23

An RI ≥ 3.0 is consistent with an appro- ued hemolysis and the potential for
priate bone marrow response to anemia, increased, bystander hemolysis with trans-
and therefore helps to rule out bone marrow fusion. Because of the cardiac instability, it is
dysfunction in this case. Modern blood cell advisable to give the transfusion slowly and
analyzers have the ability to calculate the monitor for worsening cardiac function.
absolute reticulocyte count and the fraction Consideration can be given to IVIG infu-
of “immature” reticulocytes directly. sion concurrently with the packed red blood
Patients who are demonstrating an appro- cell (PRBC) transfusion. Finally, a change in
priate response to hemolysis will have an therapy would be advisable at this point
elevated absolute reticulocyte count and with an immunosuppressant drug such as
immature reticulocyte fraction; these will be rituximab (monoclonal antibody to CD20),
low or normal in patients with an inade- cyclosporine, or cyclophosphamide.
quate response.
Other labs include a total bilirubin of 2. You have been consulted on a now 4-day-
6.7 mg/dl, LDH of 936 U/l (reference range old full-term neonate with profound jaun-
313–618 U/l), and AST of 161 U/l (reference dice (unconjugated hyperbilirubinemia)
range 8–43 U/l). DAT is noted to be positive that has risen from 11 mg/dl at 72 hours to
for IgG and C3d. 20 mg/dl. The neonatology team has initi-
b. What is the likely diagnosis? ated phototherapy, but is asking for hema-
With the positive DAT to IgG and comple- tology recommendations.
ment and clinical and laboratory signs of a. What are some background questions
hemolysis, warm antibody-mediated AIHA that may be clarifying?
is the likely diagnosis. It should be noted It is important to consider questions which
that a positive DAT without clinical and can help determine if the jaundice is physi-
laboratory signs of hemolysis is not suffi- ologic or non-physiologic. A conjugated
cient for the diagnosis of AIHA. hyperbilirubinemia is never physiologic and
c. What should be the initial treatment should prompt early referral. An unconju-
plan? gated hyperbilirubinemia in the first 14 days
The patient is started on steroid therapy, of life in the neonate can be physiologic sec-
intravenous (IV) methylprednisolone 1 mg/ ondary to liver immaturity and may be pre-
kg twice a day (BID). After a couple of days, cipitated by prematurity, poor feeding,
the hemoglobin has continued to decrease Asian ethnicity, infection, breastmilk jaun-
to 3 g/dl even though the methylpredniso- dice, and drugs which impact liver metabo-
lone has been increased to 4 mg/kg BID and lism. A family history of extended
the patient is showing signs of symptomatic phototherapy may be indicative of physio-
anemia and congestive heart failure. logic or non-physiologic conditions. Family
d. How should your treatment change at history of enlarged spleen, splenectomy,
this point? gallstones and/or cholecystectomy, or jaun-
Since the patient has a falling hemoglobin dice at an older age could by indicative of a
with clinical signs of cardiac instability and membranopathy (i.e., hereditary spherocy-
volume overload, the patient should be trans- tosis or elliptocytosis), while jaundice at an
fused. The term “least incompatible unit” has older age could be indicative of an enzymo-
been used in the past but is a misnomer if pathy (i.e., G6PD, PK deficiency, or Gilbert
phenotypically matched blood is given. The syndrome). It is important to ensure that the
patient may not have a normal increase in mother received prenatal care and was tested
hemoglobin with transfusion due to contin- for Rh incompatibility. ABO incompatibility
24 Chapter 2

as well as minor red cell antigen incompati- 28 year old mother. The baby appears jaun-
bility (i.e., Kell) may also lead to jaundice, diced but otherwise well so a CBC and bili-
though not as severe as Rh incompatibility rubin are checked. The CBC shows an anemia
(with minor antigen incompatibility not to 9.4 g/dl (MCV 110) with normal WBC/
being as severe as ABO incompatibility). diff and platelets. Total bilirubin is 10.2 mg/dl
Gilbert syndrome is due to a mild deficiency (all indirect). Mother’s blood type is O, the
of uridine diphosphate glucuronosyltrans- baby is A. What is the next best test to order?
ferase (UGT1A1), which conjugates biliru- a. Blood culture
bin in the liver. UGT1A1 is lacking in b. Reticulocyte count
Crigler–Najjar syndrome type I leading to c. Direct antiglobin test
severe hyperbilirubinemia after birth, while d. G6PD screening
Crigler–Najjar type II maintains approxi- Explanation: The most likely diagnosis in
mately 10% activity and milder symptoms. this scenario is hemolytic disease of the
b. Given the high bilirubin level, what newborn (HDN) which is typically due to
are some follow up studies that should be ABO incompatibility. Generally the mother
considered? is O blood type and the baby is either A or B
Hemoglobin levels, reticulocyte count, and blood type. Although reticulocytosis is
a DAT (direct Coombs) can be helpful. present, a positive direct Coombs (direct
Mothers who are type O blood type with a antiglobin test; DAT) is diagnostic. HDN
type A infant are most likely (type B less typically presents in the first 24 hours with
likely) to have a newborn with HDN. A nor- minimal anemia and jaundice. With good
mal hemoglobin level makes hemolysis less prenatal care, Rh incompatibility is quite
likely, while a low hemoglobin with an ele- rare though if present would lead to a more
vated reticulocyte makes hemolysis likely. severe presentation. Choice a. is incorrect as
A positive DAT would be consistent with the baby is otherwise well appearing without
HDN though may not always be present. fevers. Choice b. is reasonable to include
Low haptoglobin levels can also be a helpful with the DAT. Choice d. is more likely in the
diagnostic clue. Nonimmune mediated male newborn of Mediterranean or Asian
causes should be considered if there is not descent with prolonged jaundice (the
blood group incompatibility or a positive African G6PD type does not lead to neona-
DAT. G6PD deficiency is the most likely tal jaundice). The answer is c.
cause in the male infant of African-
American or Mediterranean descent and 2. You are seeing a patient in the emergency
may also be present in a lyonized or homozy- department who was noted to have scleral
gous female and can be diagnosed based on icterus, jaundice and pallor. A CBC is done
red blood enzyme levels. Hereditary sphero- with a hemoglobin of 5.8 g/dl and a reticulo-
cytosis can be diagnosed based on an ele- cyte percent of 14.8%. WBC/diff and platelets
vated MCHC (>36 g/dl) with follow-up are within normal range. Direct antiglobin
confirmatory genetic testing (RBC band 3). test (DAT, direct Coombs) is positive for IgG
and C3d. What is the best next step?
a. Admit for observation
Multiple choice questions b. Give a PRBC transfusion secondary
to the anemia
1. You are seeing a 1-day-old, 3.2 kg male c. Start steroids, 2 mg/kg/day methyl-
infant in the newborn nursery, born at ­prednisolone
38 weeks gestational age to a G2P2 healthy d. Give PRBCs and start steroids
Hemolytic Anemia 25

Explanation: This is a classic presentation anemia, the history is most suggestive of


for warm antibody autoimmune hemolytic G6PD deficiency in an African‐American
anemia (AIHA). A positive DAT with retic- male patient with a recent oxidative stress.
ulocytosis is pathognomic for this diagnosis, G6PD is the most common X‐linked red cell
especially when positive to IgG and comple- enzyme deficiency. A hemolytic crisis can
ment. The underlying etiology is often not occur with oxidative stress, secondary to
found but can but acutely related to a previ- illness or oxidant foods or medications.
ous infection or may be due to an underly- Hemoglobin is oxidized to methemoglobin
ing autoimmune condition such as SLE. As and then denatured, forming intracellular
there is an autoimmune etiology, steroids inclusions called Heinz bodies. This portion
are the appropriate treatment with transfu- of the RBC may be removed by the reticu-
sion reserved to patients with severe anemia loendothelial system leading to “bite cells.”
not responsive to steroid therapy or with The answer is a.
cardiorespiratory compromise. PRBC trans-
fusion can increase hemolysis and also lead 4. What would be the most appropriate
to bystander hemolysis, worsening the next step in the above case?
underlying condition. If blood is required a. Start IV steroids
the blood bank should use the PRBC unit b. Admit for observation
that has the least cross‐reactivity. The ster- c. Give a PRBC transfusion
oid dose should be increased as needed if d. Reassure the family and provide close
there is no response to the initial dose. The follow up
answer is c. Explanation: The patient is actively
hemolyzing but is recovering from his ill-
3. You are seeing a 5-year-old African‐ ness and has a reassuring exam with resolu-
American male in clinic who presents with tion of his scleral icterus. Although he is
pallor after a viral illness. He has now anemic he is well compensated and having
recovered from this illness. Mom notes that an appropriate reticulocyte response. If
his eyes appeared a bit yellow prior but have there are concerns from the family’s or
normalized now. She notes that he has had clinician’s standpoint it would be reasonable
yellowing of his eyes a few times prior and to admit for observation but this is not
she remembers a maternal uncle with simi- necessary if close follow up can be ensured.
lar complaint. On exam the child is pale and Choice a. is incorrect as this is a non‐
tachycardic with splenomegaly but otherwise immune hemolytic anemia and there would
has a reassuring exam. Blood counts reveal be no benefit to steroid administration.
a hgb of 7.2 g/dl, retic of 11.1%, normal Choice c. would be reasonable if the patient
WBC/diff, normal platelets. Chemistries were more symptomatic or if the family pre-
show an elevated LDH, indirect bilirubin ferred transfusion but is not necessary at
and AST. DAT is negative. What is the most this point in an otherwise stable‐appearing
likely diagnosis? patient. The answer is d.
a. G6PD deficiency
b. Hereditary spherocytosis 5. You are following a 14-year-old female
c. PK deficiency who was admitted for further work up of
d. Hemolytic uremic syndrome worsening ability to concentrate at school.
e. Disseminated intravascular coagulation She has otherwise felt okay although her
Explanation: Although all of the above con- school performance has dropped in the
ditions can cause a non‐immune hemolytic last six months. There are no underlying
26 Chapter 2

psychosocial triggers and her exam is normal. membranopathy and enzyme deficiency,
On routine labs her hemoglobin is 10.4 g/dl, respectively, and would be reasonable to do
bilirubin is 2.4 mg/dl (all indirect), AST if the ceruloplasmin is not diagnostic. For
and ALT are mildly elevated, BUN/Cr are choice e., an underlying autoimmune condi-
normal. You add on a retic and it is 2.4%. tion such as SLE generally leads to an anemia
Thyroid function studies are normal. of chronic disease rather than a hemolytic
Coagulation studies are normal. What is the process. The answer is c.
next best step?
a. Provide reassurance
Suggested reading
b. Send osmotic fragility studies
c. Check ceruloplasmin
Cappellini, M.D. and Fiorelli, G. (2008).
d. Send RBC enzyme panel Glucose‐6‐phosphate dehydrogenase defi-
e. Check ESR and ANA ciency. Lancet 371: 64–74.
Explanation: Wilson’s disease can often pre- Ciepiela, O. (2018). Old and new insights into the
sent with subtle findings that remain undiag- diagnosis of hereditary spherocytosis. Ann.
nosed for many years. Patients will Wilson’s Transl. Med. 6: 339.
may present with a mild, nonimmune hemo- Grace, R.F. and Glader, B. (2018). Red cell mem-
lytic anemia which can be a clue to the diag- brane disorders. Pediatr. Clin. North Am. 65:
nosis. Bronzing of the skin, liver failure and 579–595.
Kayser‐Fleischer rings due to copper deposi- Haley, K. (2017). Congenital hemolytic anemia.
Med. Clin. North Am. 101: 361–374.
tion are more often see at a later stage of dis-
Lauer, B.J. and Spector, N.D. (2011).
ease presentation. Thyroid problems can
Hyperbilirubinemia in the newborn. Pediatr.
also lead to subtle neuropsychiatric symp- Rev. 32: 341–349.
toms as well as anemia so it is important to Liebman, H.A. and Weitz, I.C. (2017).
rule that out at presentation. Choice a. is Autoimmune hemolytic anemia. Med. Clin.
incorrect as the patient has a mild anemia North Am. 101: 351–359.
with signs of hemolysis. Choice b. and d. Noronha, S.A. (2016). Acquired and congenital
are reasonable when trying to rule out a hemolytic anemia. Pediatr. Rev. 37: 235–246.
3 Sickle Cell Disease

Sickle cell disease (SCD) refers to a group of severe complications, although children
genetic disorders that share a common who have vaso‐occlusion and other compli­
feature: hemoglobin S (hgb S) alone or in cations generally have a more severe course.
combination with another abnormal Increased leukocyte count, decreased hemo­
hemoglobin. The sickle cell diseases are globin with concomitant increased reticu­
inherited in an autosomal codominant locytosis, as well as frequency and severity
manner. The molecular defect in hgb S is of vaso‐occlusive episodes (VOEs) are
due to the substitution of valine for glutamic associated with increased morbidity and
­
acid at the sixth position of the β‐globin mortality.
chain on chromosome 11. This substitution Alpha‐thalassemia (α‐thalassemia; fre­
results in polymerization of hemoglobin quency 1–3% in African‐Americans) may
causing the red cells to transform from be coinherited with sickle cell trait or dis­
deformable biconcave discs into rigid, ease. Individuals who have both α‐thalas­
sickle‐shaped cells. Hypoxia, acidosis, and semia and sickle cell anemia are less anemic
hypertonicity facilitate polymer formation. than those who have sickle cell anemia alone
The most common combinations of due to a more similar concentration of
abnormal hemoglobins are (i) hgb SS, (ii) α‐ and β‐globulins. However, α‐thalassemia
hgb SC, and (iii) hgb S with a β‐thalassemia, trait does not appear to prevent frequency or
either Sβ+ or Sβ0. The most severely affected severity of vaso‐occlusive complications,
individuals have either hgb SS or Sβ0 (no resulting in eventual end‐organ damage.
normal β‐globin production). Individuals Sickle cell disease is not uncommon, as
with hgb Sβ+ have decreased β‐globin the genetic mutation occurred as a protective
production and less severe disease, whereas mechanism from malaria in those with sickle
children who have hgb SC have intermediate cell trait. In African‐Americans, the fre­
severity of disease. There is phenotypic quency of genetic alteration is quite high: 8%
overlap between hgb SS and hgb SC; some have the hgb S gene, 4% the hgb C gene, and
children with hgb SC are more symptomatic 1% the β‐thalassemia gene. Approximately 1
than children with hgb SS. There are in 600 African‐American infants has sickle
many variables to expression of this hemo­ cell anemia. Sickle cell disease also occurs in
globinopathy including haplotype, hgb F children from the Middle East, India, Central
concentration, and other as yet to be deline­ and South America, and the Caribbean.
ated factors. Currently it is not possible to All children who have sickle cell hemo­
predict the severity of disease in advance of globinopathies have a variable degree of

Handbook of Pediatric Hematology and Oncology – Children’s Hospital & Research Center Oakland,
Third Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
© 2021 John Wiley & Sons Ltd. Published 2021 by John Wiley & Sons Ltd.
28 Chapter 3

hemolytic anemia and vaso‐occlusive tissue sickle cell centers with access to multidisci­
ischemia resulting in numerous clinical plinary providers, with provision of age‐
complications. When subjected to a low‐ based assessments. Patients should be
oxygen state, hgb S polymerizes and the red referred following positive newborn
cells become altered into a rigid sickled screening for a definitive diagnosis with
form. The pathophysiology of vaso‐ hemoglobin electrophoresis and possibly
occlusion is complex and involves many thalassemia genotyping. Frequent visits are
factors and pathways including chronic encouraged to provide education to pri­
inflammation, oxidative stress, altered mary caregivers on the disease process,
hemostasis of the coagulation system, subtle signs and symptoms of infection,
hemolysis‐associated reduction in nitric pain, and complications at various points
oxide availability, and increased viscosity. of growth and development, as well as pro­
Organs most sensitive to the ischemia and vide intervention as needed. A multidisci­
reperfusion injury are the lungs, spleen, plinary team approach to care including
kidneys, bone marrow, eyes, brain, and the intervention and support by nurses, social
heads of the humeri and femurs. Sickling workers, genetic counselors, and therapists
has both acute and long‐term implications is critical to comprehensive care. Important
for organ function. Cerebral vascular disease elements of care are detailed in this chapter
can be subtle, causing only abnormal and include:
neuropsychological testing or it can be ●● Ensuring immunizations are up to
catastrophic, resulting in hemiparesis, coma, date, anticipating development of splenic
or death; acute pulmonary sickling causes dysfunction.
lung injury leading to restrictive lung ●● Early initiation of penicillin and educa­

disease and eventually pulmonary tion regarding recognition of infection.


hypertension; osteonecrosis of the femoral ●● Frequent nutritional and developmental

head can be debilitating, resulting in the assessments; children may have delayed
need for hip replacement; untreated growth and development including sexual
retinopathy can lead to blindness; and, development.
sickle cell nephropathy can cause asymp­ ●● Assessments of school or work perfor­

tomatic proteinuria, an early sign of the risk mance, neuropsychiatric assessments as


of eventual renal failure. indicated.
Now that newborn hemoglobinopathy ●● Monitoring of CBCs and degree of
testing is mandatory in all states, children hemolysis as well as liver/kidney function.
are diagnosed early and receive appropriate ●● Pulmonary monitoring (pulmonary func­

care before they are at risk for complications. tion tests, oxygen saturation is tested but
All infants who have an electrophoretic may not correlate with PaO2 in steady state);
pattern of hgb FS at birth will have some asthma is associated with an increased rate of
form of sickle cell disease. complications including pain, acute chest
syndrome (ACS), stroke, and pulmonary
hypertension.
Preventive care ●● Transcranial Doppler (TCD) ultrasonog­

raphy as primary stroke prevention.


Preventive care is a critical aspect of ensur­ ●● Cardiac function assessments, echocardi­

ing optimal health for children, teens, and ogram (ECG).


adults with sickle cell disease. Optimally, ●● Monitoring for sickle retinopathy and

patients should be cared for in specialized associated complications.


Sickle Cell Disease 29

●● Bone health monitoring. S. pneumoniae infection that does not change


●● Red cell phenotype, anticipating red cell with age.
transfusion at some point. Additional bacteria that cause morbidity
●● Early intervention and education regard­ and mortality include Haemophilus influen-
ing newer preventive/curative therapies zae, Neisseria meningitidis, Mycoplasma
including initiation of hydroxyurea (HU) pneumoniae, Staphylococcus aureus,
and potential clinical trials, as well as gene Salmonella species, Escherichia coli, and
therapy and stem cell transplant. Streptococcus pyogenes. The S. pneumoniae
●● Management of pain in the outpatient and H. influenzae vaccines have importantly
setting. resulted in a lowered case rate of sepsis from
these organisms. Meningococcal vaccination
should also be given during the adolescent
Fever and infection in sickle period. Viral infections, particularly parvo­
cell disease virus B19, can cause severe aplastic crises
as well as ACS in patients with sickle cell
Susceptibility to infection is increased not disease.
only because of loss of splenic function due All patients with a fever (≥38.3°C) should
to infarction but also because of other have a complete evaluation and laboratories
acquired immunologic abnormalities. This including CBC with differential, reticulocyte
can result in life‐threatening episodes of count, blood culture, urinalysis, and urine
sepsis. Recognition of this susceptibility and culture (see Figure 3.1). A chest radiograph
aggressive medical management have should also be obtained. Meningitis can
resulted in an increased life span for most occur in children with sickle cell disease, but
patients. routine lumbar puncture without physical
Most children with sickle cell disease are signs of meningitis is not warranted.
identified at birth, started on prophylactic Urosepsis is common in sickle cell patients
penicillin by age 2 months, and aggressively of all ages. Our practice remains that all
monitored and treated for signs of infection. infants (up to 2–3 years of age) should be
However, with the increasing concern for admitted and cultures followed for 48 hours.
bacterial antibiotic resistance, health care These patients should not be discharged
providers need to be vigilant when con­ during this time even if they appear well and
fronted with an infant or a child who has are afebrile. Some practitioners would
fever (≥38.3°C) and/or appears ill. Overall, recommend admission in all patients with a
Streptococcus pneumoniae is responsible for fever. More recent data show the benefit of
>80% of the morbidity of infection. In some vaccination against encapsulated organisms,
areas of the United States, up to 50% of pneu­ with very low rates of sepsis in young
mococcal isolates are penicillin‐resistant. patients with sickle cell disease. Therefore, a
Infections can precipitate VOEs and other more liberal policy in regard to admission
complications of sickle cell anemia and, in can be considered in the fully vaccinated
this population, can quickly become fulmi­ infant if appropriate follow‐up can be
nant. Although the American Academy of ensured.
Pediatrics guidelines recommend the discon­ ACS should be high on the differential in
tinuation of penicillin prophylaxis after 6 all children with sickle cell disease with
years of age, our institutional practice is to fever. While physical examination is
continue it as long as possible (until compli­ essential, 60% of pulmonary infiltrates in
ance becomes an issue) due to the high risk of children with sickle cell disease and fever
30 Chapter 3

Rapid assessment
history & physical

Septic appearing child Infant, child less than


2–3 years of age:
Non-toxic appearing child

Rapid assessment
for degree of respiratory and History (last dose of penicillin):
cardiovascular compromise; Cardiovascular and respiratory status,
meningismus; splenomegaly, jaundice, pain;
cardiovascular monitoring and neurologic screen for weakness and
pulse oximetry altered mental status

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
Catheterized urine culture Oxygen Catheterized urine culture
Consider LP Chest X-ray Consider LP
Type and cross match ABG Chest X-ray
Chest X-ray

Antibiotics Antibiotics
Fluid bolus to Ceftriaxone 50 mg/kg IV push
maintain blood pressure Ceftriaxone 50 mg/kg
or rapid infusion (plus azithromycin if CXR findings
Note: fluid overload can lead
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).

will be missed on exam alone; therefore, access, ceftriaxone can be given intramuscu­
chest radiography is essential. If there is lar (IM) while access is being obtained. Of
another possible source of infection, appro­ note, studies have shown that the higher the
priate cultures should be obtained. Rapid temperature, the more likely the risk for
antibiotic treatment with parenteral ceftriax­ bacterial sepsis.
one should occur in the emergency depart­ Children who do not appear septic, in
ment after obtaining labs and blood cultures. whom there is a low index of suspicion, vac­
If there is difficulty with intravenous (IV) cinated, and who are older than 2–3 years of
Sickle Cell Disease 31

age (per institutional preference) can be feet. This is seen exclusively in infants and
treated as outpatients with IV/IM ceftriax­ children (<5 years of age). It presents with
one while awaiting culture results, only if pain, low‐grade fever, and diffuse nonpit­
close monitoring can be assured (parents ting edema of the dorsum of the hands and
can be contacted by telephone and daily feet, which extends to the fingers. One or
evaluation is easily done while still febrile). more extremities may be affected at one
If all cultures are negative after 2–3 days and time. Radiographic changes (periostitis and
the child is afebrile without clinical symp­ subperiosteal new bone formation with per­
toms, the antibiotics and follow‐up can be iosteal elevation) may appear 1 week or
discontinued. more after clinical presentation. Therapy is
All patients who appear ill should be supportive, with analgesics, hydration, and
hospitalized and treated for presumed infec­ parental reassurance. Although the swelling
tion. Children may develop increasing or radiographic changes may persist for
symptoms after being evaluated and hospi­ weeks, the syndrome is almost always self‐
talized, and should be monitored closely. limited. Transfusions and antibiotics are not
ACS commonly occurs after hydration and necessary unless there is concern for infec­
is frequently precipitated by a vaso‐occlu­ tion or the medical condition worsens.
sive pain episode. Occasionally, there is a precipitating
event such as hypoxia (e.g., obstructive sleep
apnea), fever, viral illness, or dehydration
Vaso‐occlusive episodes that leads to the VOE. Few children have
frequent pain episodes; it should not be
The bones and joints are the major sites of taken for granted that each of these episodes is
pain in sickle cell disease. In trabecular similar, and a source for each painful event
bones, such as the vertebrae, infarction can should be sought. If the pain changes or is
occur and eventually lead to collapse of the very persistent, the child should be
vertebral plates and compression. The classic reevaluated. If a young child has a symptom
radiographic appearance is of “fish mouth” that is interpreted as pain, such as refusal to
disc spaces and the “step” sign (a depression walk or a limp, also consider that the cause
in the central part of the vertebral body). may be due to an acute central nervous
Back pain is a common symptom in sickle system (CNS) event.
cell disease, likely as a result of recurrent The most common type of pain is
infarction and vertebral compression. musculoskeletal pain. The pain may be of
Infarction in the long bones can cause swell­ any configuration: isolated, multifocal,
ing and edema in the overlying soft tissues. It symmetric, migratory, or associated with
may be difficult to differentiate a VOE from erythema or swelling. There can be low‐
acute osteomyelitis. Although uncommon, grade fevers, possibly associated with other
infection should be considered. Osteomyelitis clinical symptoms. It can sometimes be
may be ruled out by close clinical observa­ difficult to distinguish pain from infection,
tion, blood cultures, and, occasionally, aspi­ synovitis, or another pathologic process.
ration of the affected area. Plain radiographs Children are frequently seen with abdo­
are not helpful in the early stages of infection minal pain. A surgical abdomen needs to
and bone scans may not differentiate a sim­ be considered. Children who have sickle
ple infarct from osteomyelitis. cell anemia have a high incidence of chole­
Dactylitis, or hand–foot syndrome, cystitis. Also consider pancreatitis, u ­ rinary
refers to painful swelling of the hands and tract infection, pelvic inflammatory disease,
32 Chapter 3

and pneumonia presenting as abdominal the development of chronic pain syn­


pain. Ileus and ACS are frequent complica­ dromes. In addition to specific treatment
tions of abdominal VOEs. for the acute pain event, treatment with
VOEs can last for many days. Do not hydroxyurea as a preventive intervention
assume that the episode is controlled when should be considered in all patients with
the acute administration of analgesia is effec- sickle cell disease (see section on
tive. Children with these symptoms may hydroxyurea).
have a medical problem that needs aggres­
sive treatment beyond therapy for their
pain. A VOE is a diagnosis of exclusion. The Outpatient
pain needs to be treated while there is a
workup for potential other causes of pain Mild pain
beyond the VOE itself. ●● Increase fluids to 1–1.25× maintenance.
A reliable tool should be used to assess a Water, fruit juice, and fruit drinks as well as
child’s level of pain. The assessment should decaffeinated soda are recommended.
be modified depending on the age and ●● Acetaminophen without codeine (15 mg/kg

developmental level of the child, and the q4–6 hours) orally can be tried prior to
pain tool should be able to rate both using acetaminophen with codeine (1 mg
subjective and objective aspects of the child’s codeine/kg q4–6 hours) depending on what
pain. Physicians should be familiar with medications are currently being used by the
their use and refer to them when assessing patient for pain. For many children,
children and adolescents with pain. At our acetaminophen alone is not sufficient for
institution, we use the validated adolescent vaso‐occlusive pain. Patients who utilize
pediatric pain tool (APPT). hydrocodone or oxycodone with or without
High‐risk factors for complications other acetaminophen should continue with this
than VOE in children with pain: regimen even if the pain is not severe.
●● Fever ≥101°F (38.3°C) and signs of Codeine is generally not recommended
infection. though may be the only choice when the
●● Acute pulmonary symptoms (chest pain, patient cannot present for a prescription of a
hypoxia, abnormal auscultatory exam). controlled substance.
●● Persistent vomiting. ●● Ibuprofen (10 mg/kg q6–8 hours) orally

●● Pain that is unusual for the patient. should be used routinely, every 8 hours,
●● Severe abdominal pain. even if pain is temporarily controlled.
●● Extremity weakness or loss of function. Ketorolac IV (or IM) can be given if the
●● Any neurological symptoms. patient is seen as an outpatient.
●● Severe headache. ●● Rest and heat to the area of pain.

●● Acute joint swelling. ●● Relaxation and biofeedback exercises, such

as positive visualization and meditation.

Pain management Severe pain


●● IV hydration: bolus to correct fluid losses,

Following are the recommendations for then IV with D5W 1/2 NS (dextrose 5%
the management of vaso‐occlusive events. water, 0.45% normal saline) at 1 to 1.25×
However, many children and adolescents maintenance (IV + PO). Use caution not to
need individualized care plans for their overhydrate, especially if there are any
routine treatment, especially those with pulmonary symptoms (overhydration can
Sickle Cell Disease 33

lead to pulmonary edema and precipitate the management of acute on chronic pain at
ACS). Note that if the patient does not a dose of 0.2 mg/kg. For patients with
appear volume‐depleted, an IV fluid bolus is frequent utilization of opioids (i.e., oral
not routinely required. hydrocodone or oxycodone), upregulation
●● Analgesics (IN, intranasal): Due to the of the NMDA receptor decreases opioid
rapid onset of action, a single dose of IN efficacy. Utilization of ketamine as a NMDA
fentanyl, 1–2 μg/kg, has been shown receptor antagonist allows greater opioid
beneficial while IV access is being obtained. efficacy in addition to being synergistic due
Given the very short duration of effect, IV to benefits in pain, anxiety, and depression.
therapy should be utilized as soon as IV Side effects of single low‐dose ketamine
access is available. include dysphoria and disassociation which
●● Analgesics (IV): Morphine: 0.1–0.15 generally are mild and can be managed by
mg/kg/dose q3–4 hours. In children <2 benzodiazepines, if necessary.
years who have not been exposed to narcotic Upon presentation to the clinic or
analgesics, 0.05–0.1 mg/kg should be used. emergency room, the child or adolescent
The pharmacokinetics of morphine differs should be rapidly assessed and treated for
between individual children and doses must pain. Initially, both morphine and ketorolac
be titrated for individual patients. Both should be given at the maximum
underdosing and overdosing need to be recommended doses. If the pain worsens
avoided. Careful management is required! again, repeat the parenteral narcotic dose; it
Previous medication history is often a good is unlikely that oral analgesics will be
starting place. For patients with a previous successful if this regimen is not effective. If
intolerance to morphine secondary to pain relief can be achieved and maintained
nausea or pruritus, IV dilaudid for 3 hours, administer an oral narcotic and
(hydromorphone) should be utilized while observe for 1 hour. If this regimen is
calculating the dose based on morphine successful, give a prescription of narcotic
equivalents. Dilaudid is approximately and nonsteroidal anti‐inflammatory medi­
8× stronger than morphine, so a dose of cations for several days (10–15 doses). There
0.01–0.02 mg/kg should be utilized. should be follow‐up within 72 hours. If this
●● Ketorolac: (child) IV: 1 mg/kg/load, then regimen is not successful, if other symptoms
0.5 mg/kg q6 hours (max 30 mg); (adult) IV: develop with hydration, or a fever develops,
30–60 mg/load, then 15–30 mg/kg q6 hours. the patient should be admitted, treated, and
Gastrointestinal (GI) bleeding has been observed for possible evolution to ACS or
reported, and an H2 blocker such as famo­ other life‐threatening infection.
tidine is required if ketorolac is given IV.
Morphine and ketorolac can be given simul­
taneously or in succession for the initial treat­ Inpatient
ment of pain in children. Added to narcotic
therapy, ketorolac increases analgesia and has As noted in the preceding text, the administra­
a narcotic sparing effect. Ketorolac is very tion of narcotics to acutely ill patients requires
useful in pain control, but should not be used careful monitoring. A plan for pain manage­
for more than 5 days. ment should be defined on admission and
●● Adjuvant medications (IV): We and other followed for the duration of hospitalization.
centers have begun utilizing ketamine, an A recent statement by the Joint Commission
N‐methyl‐d‐aspartic acid (NMDA) receptor regarding pain management notes the
antagonist, as an adjuvant medication for importance of both pharmacologic as well
34 Chapter 3

as non‐pharmacologic strategies. Routine alternative to morphine and preferred by


inpatient care is outlined in the following text. some patients.
●● Low‐dose continuous ketamine drips

General management are being used with success to manage acute


●● Correct acute losses due to dehydration. severe pain requiring hospitalization. Recom­
Do not overhydrate children who have mended dose is 1–5 mcg/kg/min. These doses
pulmonary symptoms. are typically well‐tolerated, though some
●● Total fluid intake (IV + PO) should be patients may experience disassociation at
1–1.25× daily maintenance fluids; do not higher doses which can be mitigated with
overhydrate. benzodiazepines. Ketamine may be initiated
●● Monitor daily weight, record strict intake at the outset to potentiate opioid efficacy
and output (I&O), and observe for signs of and can be continued safely for 7–10 days
fluid overload. of hospitalization and discontinued without
●● Encourage ambulation, sitting up in bed, a taper.
and taking deep breaths. ●● Objective measurement of response to

●● Incentive spirometry (IS), 10 times an analgesic therapy through utilization of


hour, every hour while awake. tools such as the APPT is vital to ensure that
the patient is having the appropriate
Analgesics response to the therapy prescribed.
●● Pain medication should be administered Side effects of narcotics include respira­
on a fixed‐time schedule, with an interval tory depression, nausea, vomiting, pruritus,
that does not extend beyond the duration of hypotension, constipation, inappropriate
the analgesic effect. Do not give narcotics on secretion of anti‐diuretic hormone, and
a PRN (as needed) basis except for when the change in seizure threshold. Low‐dose
patient is ready for discharge. naloxone drip (i.e., 1 μg/kg/h) has been used
●● Titration schedules require a written for severe pruritus and urinary retention due
plan, close observation, and a flow sheet to morphine. The most common side effects
to monitor effectiveness; sedation and of nausea, vomiting, and pruritus resolve
hypoventilation can lead to hypoxia and over time. Diphenhydramine may be used
increased sickling and pain in those safely in this setting and can have a synergis­
receiving excess narcotics, while patients tic positive effect with morphine. We suggest
being underdosed will have continued pain oral administration, though it can be given
as well as potential splinting increasing the IV. Meperidine should not be substituted for
risk of atelectasis which can lead to ACS. morphine. Metabolites of meperidine can
●● Patient‐controlled analgesia (PCA): This accumulate and cause seizures, especially
is the method of choice for controlling pain if used over a longer period at high doses.
in children who are ≥7 years of age or older. A plan for withdrawal should be a part of
PCA protocols may be hospital‐specific. discharge planning; ­clonidine hydrochloride
Generally, there is an initial bolus (0.1 mg/kg has been used s­ uccessfully in this setting, but
morphine), and an on‐demand dose (0.01– should be carefully monitored.
0.05 mg/kg morphine) with a 10–20‐minute All patients who are hospitalized for pain
lock out, and a 1 hour maximum of 0.1– should have an incentive spirometer at the
0.15 mg/kg of morphine. At night, there bedside. Ensure that the patient has been
may be a need for a continuous low‐dose instructed on its usage and can demonstrate
infusion (0.01–0.02 mg/kg/h morphine) in it appropriately. IS is effective in reducing
addition to the bolus dosing. Dilaudid is an the incidence of ACS in the setting of VOE.
Sickle Cell Disease 35

Alternative methods of pain control


(behavior modification, relaxation, visuali­ Acute chest syndrome/
zation, self‐hypnosis, local heat, and trans­ pneumonia
cutaneous electrical nerve stimulation) are
helpful adjuvants in the outpatient and ACS is defined as the development of a new
inpatient setting, but should not replace pulmonary infiltrate in the presence of fever
standard therapy. Children should have or respiratory symptoms. As chest radio­
access to a psychologist who is experienced graph changes may be delayed, the diagnosis
in the management of sickle cell pain. may not be apparent at presentation.
Drug addiction is extremely rare and Approximately 50% of children with sickle
should not be a primary concern. The goal cell disease will experience ACS. ACS is
should be to provide patients with adequate frequently caused by community‐acquired
relief by understanding the pharmacology pathogens such as chlamydia, mycoplasma,
of the medications, drug tolerance, and and other bacterial or viral organisms. In
physical dependence. Drug tolerance is older children, adolescents, and young
common, and withdrawal symptoms after adults, ACS may be more commonly associ­
hospitalization are probably underreported ated with vaso‐occlusion, infarction, fat
by patients. All patients should start a narcotic embolism, or in situ sickling. These episodes
taper while in the hospital and complete are characterized by chest pain, fever,
this as an outpatient. On average, patients hypoxia, and pulmonary infiltrates.
admitted for the management of VOE Although ACS usually improves with medi­
require 3–7 days of inpatient care. cal management, it can present with or pro­
gress rapidly to respiratory failure (acute
respiratory distress syndrome), requiring
Complementary therapies mechanical ventilation and emergent
exchange transfusion. Preventative therapy
●● Patient with acute pain and rare hospitali­ with hydroxyurea has been shown to
zation may be successfully managed with decrease the incidence of ACS and should be
analgesics alone. initiated in all patients with sickle cell ane­
●● More complicated patients with longer mia (see section on hydroxyurea).
or more frequent admission would bene­ Sixty percent of children who have pneu­
fit from the addition of complementary monia on X‐ray will be missed by physical
therapies to help manage pain with the exam alone. In addition, many children
recognition that opioid therapy alone is with a negative chest film on admission
not sufficient to manage chronic pain syn­ will develop findings after hydration. All
dromes which can lead to hyperalgesia children who have symptoms of pulmonary
and allodynia and have strong psychosocial disease, such as fever, shortness of breath,
elements. Recognition that a multimodal, tachypnea, chest pain, cough, wheezing,
interdisciplinary team approach is required rales, or dullness to percussion, should have
is vital. an assessment of oxygen saturation, a chest
●● Potential avenues for complementary radiograph, receive parenteral antibiotics,
therapy for patients with vaso‐occlusive and be admitted to the hospital (see
pain include acupuncture, massage therapy, Figure 3.2). Bronchodilators and IS are
cognitive behavioral therapy, biofeedback/ helpful adjuncts in treatment and preven­
relaxation therapy, warm packs or whirlpool tion of worsening ACS. If there is an infil­
therapy, and distraction therapies trate, the patient should be closely observed
36 Chapter 3

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 echocardiography

Figure 3.2 Sickle cell disease with acute pulmonary infiltrate (abbreviations: CBC, complete blood
count; ABG, arterial blood gas; ACS, acute chest syndrome).

for hypoxia and progression of pulmonary and mortality. Children should have oxygen
infiltrates, with repeat chest radiographs. saturation monitored and, if indicated, arte­
Overhydration can lead to pulmonary edema rial blood gases. Oxygen therapy should be
and exacerbate ACS. Oversedation with used only for significant hypoxia (i.e., O2
narcotics can also lead to hypoventilation, saturation ≤ 92%). Supplemental oxygen can
increasing the risk for ACS. Narcotics should decrease erythropoietin production and lead
be used with caution in this setting. to more severe anemia. Pulmonary infec­
ACS can develop in a matter of hours and tions should be treated aggressively, and
is associated with a high rate of morbidity these children watched closely. If there is no
Sickle Cell Disease 37

improvement, or worsening anemia, a red Management


blood cell transfusion (straight or exchange, All patients with evidence of acute pulmo­
dependent upon the severity of the hypoxia, nary pathology should be admitted to the
anemia, and clinical status of the patient) may hospital. If fever is present or if an infectious
help to correct the anemia, decrease the process is suspected, antibiotic therapy
percent hgb S, and improve oxygen‐carrying should be started immediately. See
capacity to aid in reversing the pulmonary Section 3.2 and Figures 3.1 and 3.2.
sickling and improve the clinical course. ●● Oxygen

Transfusions are more effective when admin­ Hypoxemic patients (PaO2 70–80 mmHg,
istered early in the course of ACS, rather than O2 saturation ≤ 92%): 2 l/min per nasal can­
as a life‐saving measure in a critically ill child. nula. Goal should be 96% O2 saturation (not
There is a distinct difference between the 100%).
etiology of ACS in children and that of adoles­ Reevaluate arterial blood gas on oxygen.
cents and adults. In children, the incidence of ●● Antibiotics

ACS is seasonal, lower in the summer months Initiate broad spectrum antibiotic: ceftri­
with increasing rates in the winter when viral axone 50 mg/kg/d IV every 24 hours after
infections are prevalent. In adults and adoles­ blood cultures obtained (maximum 2 g/dose).
cents, ACS is frequently a complication of an Due to the frequency of atypical organ­
episode of vaso‐occlusion (without fever) due isms (e.g., chlamydia and mycoplasma), a
to pulmonary fat embolism. This event will macrolide or quinolone antibiotic should be
progress to include chest pain, fever, and a included: azithromycin 10 mg/kg on day 1,
pulmonary infiltrate (usually basilar with followed by 5 mg/kg on days 2–5 (maximum
pleural effusion). Adults and adolescents 500 mg on day 1, 250 mg on days 2–5).
more frequently need transfusions and less ●● Analgesics

frequently have a viral or bacterial infection As indicated for vaso‐occlusive pain


associated with their episode of ACS. management, administration must be
Individuals with hgb SC disease have rela­ monitored to provide the maximum pain
tively more fat in their marrow and resultantly control and prevent hypoventilation or
can have more severe pulmonary fat emboli atelectasis from splinting or narcotization.
when their course is complicated by ACS. ●● Hydration

Stroke and other central nervous system PO and IV hydration (D5W 1/2 NS) at a
events are more common in children with maximum of 1.25× maintenance. Caution
ACS in the 2‐week period after the event. should be used in patients with potential
ACS to avoid fluid overload. Monitor Is and
Laboratory evaluation of acute Os, daily weight.
chest syndrome ●● Respiratory supportive measures

●● CBC with differential and reticulocyte Continuous pulse oximetry.


count. IS, 10 times an hour, every hour while
●● Type and hold for possible blood trans­ awake (prevention of hypoventilation and
fusion (phenotypically matched, sickledex atelectasis).
negative). Albuterol aerosols q4 hours (airway
●● Chest radiograph, as often as clinically hyperreactivity common in ACS).
indicated to monitor progression of disease. ●● Physical therapy

●● Continuous pulse oximetry and baseline Warm packs.


arterial blood gas (on room air if possible). Ambulation as tolerated, sitting up in
●● Blood culture for fever. bed.
38 Chapter 3

●●Transfusion Priapism can last for hours, days, or


Straight transfusion. weeks with moderate‐to‐severe pain, or it
Exchange transfusion (for patients with can occur as a pattern of painful erections
hematocrit > 30% or moderate‐to‐severe ACS that recur over a period of days to several
to more efficiently reduce hgb S%). weeks (stuttering priapism). Chronic
nonpainful priapism can also occur.
Transfusion decreases the proportion of Since priapism can lead to impotence,
sickling red cells and increases blood oxygen early medical management is indicated. A
affinity. The main indication is worsening urologist familiar with priapism in sickle cell
respiratory function, as documented by disease should be consulted. Treatment
hypoxemia (PaO2 < 70 mmHg on room air), includes hydration, pain management, intra­
worsening chest pain, evolving clinical cavernous injection of alpha‐adrenergic
examination, or worsening infiltrates on agents, exchange transfusion, and shunting
chest radiography. For patients with chronic surgery. Oral alpha‐adrenergics (e.g., pseu­
hypoxemia, a drop of >10% from baseline is doephedrine hydrochloride 30 mg BID, twice
a reasonable level at which to transfuse. daily) have been used successfully to treat
Delays in instituting transfusion therapy, stuttering priapism. Chronic transfusion
particularly in rapidly deteriorating patients, therapy has been used in patients with recur­
should be avoided. rent priapism (maintenance of hgb S below
30–40%), although no controlled trials have
been performed proving the efficacy of this
Priapism therapy.

Forty percent of men who are homozygous


for hgb S report having priapism in their Stroke (cerebrovascular
adolescence and early adulthood. Priapism accident)
has a second peak at 21–29 years. Priapism
is defined as a painful erection that lasts for Stroke is a common event in children
more than 30 minutes. It frequently results homozygous for hgb S. Between 10 and 15%
in interference with the urinary stream. of children with sickle cell disease suffer
Priapism can be precipitated by prolonged from overt strokes. In children, strokes are
intercourse or masturbation, frequently more frequently the result of cerebral
occurs at night, and can be differentiated vascular stenosis and infarction. The mean
from a nocturnal erection by its duration age of occurrence of clinically evident stroke
and pain. Children will occasionally is 7–8 years with the highest risk occurring
complain of dysuria as the first complaint of between 2 and 9 years. Hemorrhage and
priapism. Prognosis is poorer in adolescents infarct may occur together.
and adults who have recurrent prolonged Strokes in children who have sickle cell
priapism, and if aggressive treatment has disease involve stenosis and occlusion of
not been successful, impotence may result. the major anterior arteries of the brain,
Children who have priapism generally have including the carotids. The presenting
a better prognosis than adolescents and symptoms of stroke can be dramatic and
adults and usually do not have prolonged acute, such as coma, seizure, hemiparesis,
episodes requiring aggressive therapy. hemianesthesia, visual field deficits, aphasia,
Priapism can occur with VOEs as well as or cranial nerve palsies. Subtle limb weak­
with fever and sepsis. ness (without pain) is often mistaken for an
Sickle Cell Disease 39

acute VOE, but can be due to stroke. By defi­ TCD has been shown to predict increased
nition, the symptoms must persist for at risk of stroke in children who have increased
least 24 hours to be classified as a stroke. flow velocity in major cerebral arteries.
The presentation of severe headache and Unsuspected cerebral vascular damage was
vomiting with no other neurological find­ detected by CT and MRI in 25–30% of chil­
ings can be symptoms of an intracranial dren in the Cooperative Study of Sickle Cell
hemorrhage. Disease. No clinical factors, with the excep­
Initially, a computed tomography (CT) tion of TCD, predicted the occurrence of
scan without contrast will be normal for up stroke. In this study, there was a correlation
to 6 hours after a stroke, but will rule out between first stroke and transient ischemic
intracerebral hemorrhage, abscess, tumor, attack, ACS, elevated systolic blood pressure,
or other pathology that would explain the and severe anemia. All children who can
neurological symptoms. Magnetic resonance cooperate with TCD (usually by 2 years of
imaging (MRI) and magnetic resonance age) should have an annual evaluation if the
angiography (MRA) are much more study is available locally. Normal middle cer­
sensitive methods to determine intracranial ebral artery velocity in children with sickle
infarct but can remain normal for 2–4 hours cell disease is approximately 120 cm/s.
after an event. All catastrophic neurological Children who have velocities of 170–
events should first be evaluated with CT, 199 cm/s are considered at high risk and
since it is generally more available and will those with velocities ≥200 cm/s are at highest
rule out acute pathology. risk. The Stroke Prevention Trial in Sickle
With clinical concern prior to definitive Cell Anemia (STOP) showed that transfu­
diagnosis by MRI/MRA imaging, children sion greatly reduces the risk of first stroke in
are exchange transfused to achieve a children who have abnormally high TCD
concentration of hgb S of less than 30%. velocity (≥200 cm/s). Prophylactic chronic
Blood pressure should be continually transfusion should be strongly considered if
monitored during an exchange transfusion there are two studies at least 2 weeks apart
realizing that, relative to the normal with velocities ≥200 cm/s. Compliance issues
population, children with sickle cell disease and risks of chronic transfusions must also
are hypotensive. The diagnosis of stroke in be considered when deciding on this ther­
children should initially be made based on apy. The TCD With Transfusions Changing
clinical findings and treated emergently. to Hydroxyurea (TWiTCH) trial in high‐risk
Children should be monitored until they are children with sickle cell anemia and abnor­
stable and the hemoglobin electrophoresis is mal TCD velocities (who have received at
documented. Those who have had a stroke least 1 year of chronic transfusion therapy
require chronic transfusion therapy for an and have not MRA‐defined vasculopathy)
undetermined length of time. demonstrated that treatment with hydroxyu­
Children with sickle cell disease have rea can be substituted for chronic transfu­
more frequent and more severe headaches sions to maintain normal TCD velocities and
than other children; these may be mani­ prevent primary stroke.
festations of cerebral hypoxia and vaso­ Transient ischemic attacks are defined as
dilation. Children who have severe focal neurological deficits with a vascular
headaches accompanied by vomiting need distribution persisting for less than 24 hours,
extensive evaluation with imaging as well although they typically last less than an
as neurologic and neuropsychological hour. Patients with transient ischemic
testing. attacks are treated in a similar manner to
40 Chapter 3

those who have an infarct. The diagnosis is young as 2 months of age. Classically, how­
made in retrospect when the follow‐up MRI ever, it occurs in children with hgb SS after
is negative for a persistent lesion that would the disappearance of hgb F from approxi­
explain the neurological symptoms. mately 6 months to 3 years of age when the
Silent cerebral infarcts are the most spleen becomes fibrotic due to multiple
common neurologic injury in children with infarctions. These crises can occur in older
sickle cell anemia. In contrast to overt children who have hgb SC or Sβ+ thalassemia
strokes, children with silent infarcts do not but are usually not as severe; still, fatal events
have apparent neurologic impairment on have been reported for these children. The
examination. However, children with silent incidence is between 10 and 30% and the
infarcts are at increased risk from stroke, recurrence rate is 50%. Mild events can
new and enlarged silent infarcts, poor indicate the possibility of subsequent life‐
academic achievement and lower IQ scores threatening sequestration events. Chronic
compared to children with sickle cell anemia sequestration can be a sequela with chronic
and no evidence of silent stroke on MRI of anemia and thrombocytopenia.
the brain or in siblings unaffected by sickle The clinical presentation is usually rapid
cell anemia. Regular blood transfusion in in onset; the child will typically have sudden
this population has significantly reduced the weakness, pallor of the lips and mucous mem­
incidence of recurrent cerebral infarcts. branes, breathlessness, rapid pulse, faintness,
Intracerebral hemorrhage or subarach­ and abdominal fullness. Evaluation of the
noid hemorrhage may present without focal CBC will frequently show a precipitous drop
neurologic symptoms. Exchange transfusion from the baseline hemoglobin.
should be carried out immediately. The treatment for splenic sequestration is
Arteriography is used to identify the arterial transfusion to restore circulating blood vol­
bleed and the patient may need emergent ume, usually with phenotypically matched
surgical intervention. Mortality is very high blood (unless there is a life‐threatening situa­
during the acute event (~50%). tion). As the shock is reversed and the trans­
fused blood decreases the percentage of hgb
S, the splenomegaly regresses and much of
Acute anemia the blood is remobilized with a rapid rise in
the child’s hemoglobin.
There are two common causes of acute life‐ Splenectomy should be strongly consid­
threatening anemia in sickle cell disease: ered in all children who have a splenic
splenic sequestration and aplastic crisis. sequestration crisis. Transfusions can pre­
vent recurrence until surgery can be
Splenic sequestration arranged. Some children who have hgb SS
Infants and young children who have hgb and older children who have other S hemo­
SS and Sβ0 thalassemia and older children globinopathies can have massive spleno­
(over 10 years) who have hgb SC or Sβ+ megaly with hypersplenism after an episode
thalassemia syndromes can have intras­ of splenic sequestration. If hypersplenism
plenic pooling of large amounts of blood (with resultant anemia, neutropenia, or
and platelets. This can lead to anemia, thrombocytopenia) is persistent and severe,
thrombocytopenia, hypovolemia, cardio­ splenectomy is indicated.
vascular collapse, and sudden death within Prior to splenectomy, younger children
hours of the onset of sequestration. The should have an evaluation of splenic func­
syndrome has been reported in infants as tion with a pit count and a spleen scan. In
Sickle Cell Disease 41

normally eusplenic persons, fewer than 1% causes pain that can sometimes be confused
of the circulating red cells are pitted; values with a bone infarct. Symptoms of avascular
of 2–12% may represent decreased splenic necrosis, such as limping, can sometimes
function. If they still have a functional be confused with stroke. Limping with
spleen, usually under 2 years of age, a partial weakness but without pain is stroke not
splenectomy can be performed. Children avascular necrosis.
should have appropriate immunizations, Avascular necrosis is common in all age
including pneumococcal, haemophilus, and groups, but is more frequently diagnosed in
meningococcal vaccinations, prior to sple­ the adolescent and usually involves the
nectomy. Penicillin prophylaxis is continued humeral and femoral heads. Thorough
indefinitely in all children who have been musculoskeletal examinations with a focus
splenectomized. on the hips should be performed on an
annual basis for all children with SCD. It is
Aplastic crisis more common in individuals who have hgb
Severe anemia can develop over several Sβ0 thalassemia and hgb SS. It is also seen,
days due to shortened red cell survival though with a lower frequency, in those who
without compensatory reticulocytosis.
­ have hgb Sβ+ thalassemia. Individuals who
Reticulocytopenia can last 7–10 days. The have hgb SS are more frequently affected
primary cause of transient red cell aplasia is than those with hgb SC.
parvovirus B19, though it can follow other Avascular necrosis is a result of an infarct
viral infections as well. Parvovirus B19, the in the cancellous trabeculae in the head of
most common cause of aplasia in children either the femur or the humerus. The
with hemoglobinopathies, can also cause process of necrosis and repair can be
neutropenia. While many patients recover progressive, leading to collapse of the head
spontaneously, the anemia can be profound. or arrest with varying degrees of disability
Red cell transfusion is indicated for those and sclerosis. A method of describing the
who become symptomatic from anemia or if progression of avascular necrosis is provided
the hemoglobin falls 2 g/dl or more from here:
baseline. If the ability to follow‐up is of
concern, the child should be hospitalized for Stage 0: no evidence of disease.
observation. The patient should be isolated Stage 1: X‐ray normal, bone scan abnormal,
from other patients with chronic hemolytic MRI abnormal.
anemias (sickle cell disease or thalassemia) Stage 2: X‐ray: sclerosis and cystic changes
or red cell aplasia and should not have without collapse.
pregnant caregivers (due to risk of parvo­ Stage 3: Subchondral collapse (crescent
virus infecting the fetus and resulting in a sign) without collapse.
spontaneous abortion). If the child is mildly Stage 4: Collapse and flattening of the femo­
anemic and asymptomatic, outpatient moni­ ral head without acetabular involvement,
toring is reasonable. normal joint space.
Stage 5: Joint narrowing or acetabular
involvement.
Avascular necrosis Stage 6: Increased joint narrowing or acetab­
ular involvement.
Bone pain is common in sickle cell disease.
Marrow infarcts can cause pain that can last Treatment for avascular necrosis of the
for weeks. Avascular necrosis (osteonecrosis) femur includes bed rest with crutch walking
42 Chapter 3

for 6 weeks, nonsteroidal anti‐inflammatory disease of the eye is common in sickle cell
drugs (NSAIDs), and core decompression disease. It occurs most commonly in indi­
surgery for stages 1 or 2. Extensive physical viduals who have hgb SC.
therapy has been found to be beneficial but Treatment of sickle cell retinopathy
is often difficult to perform. Decompression requires recognition. The damaged peripheral
surgery is relatively simple; a core is removed vessels are not generally appreciated by direct
from the head of the femur. The coring ophthalmic examination and require the use
begins approximately 2 cm below the of indirect binocular stereoscopic ophthal­
trochanteric ridge extending through the moscopy. Sea fanning, vitreous hemorrhage,
neck and into the head of the femur. Acutely, and retinal detachment can be observed by
this procedure provides relief of pain and is direct ophthalmoscopy. Annual ophthalmic
thought to arrest the progression of evaluations should begin at age 10 for all chil­
avascular necrosis if done in the early stages; dren with sickle cell disease. Treatment of
however, this has not been confirmed in a early neovascularization requires laser photo­
prospective trial. Higher‐stage avascular coagulation. Surgical approaches are required
necrosis requires joint replacement. for advanced lesions. Central retinal artery
occlusion can occur rarely in patients with
sickle cell disease.
Retinopathy/hyphema Hyphema is a medical emergency in
sickle cell disease. It requires immediate oph­
The eye is particularly sensitive to hypoxia. thalmic evaluation and transfusion (exchange
Vaso‐occlusion of retinal vessels and hypoxia or straight) to reduce sickling and improve
of the retina cause permanent retinal dam­ oxygenation. If a conservative approach is not
age. Blood in the anterior chamber of the successful, anterior chamber paracentesis is
eye (hyphema) becomes rapidly deoxygen­ performed to relieve intraocular pressure and
ated and permanently sickled, obstructing remove the hyphema.
the outflow from the aqueous humor. The
accumulation of aqueous humor in the ante­
rior chamber increases intraocular pressure Hyperbilirubinemia/gallstones
leading to decreased blood flow to the retina
until the perfusion pressure of the globe is Bilirubin gallstones can eventually be
reached. This leads to sudden vascular stasis detected in most patients with chronic
and blindness. The events occurring in hemolytic anemia. In sickle cell disease,
hyphema can also occur in children who gallstones occur in children as young as
have sickle cell trait. 3–4 years of age and are eventually found
Retinal vascular occlusion initially occurs in approximately 70% of patients. It may
in peripheral retinal vessels without signifi­ be difficult to differentiate between gall­
cant sequelae. It eventually leads to neovas­ bladder disease and abdominal vaso‐
cularization from the retina into the vitreous occlusive crisis in patients with recurrent
(sea fans). These abnormal vessels are fragile abdominal pain. Cholecystectomy may be
and can bleed into the vitreous causing necessary for patients with fat intolerance,
“floaters” or blindness if the hemorrhage is presence of gallstones, and recurrent
sufficient. If bleeds do not obscure vision abdominal pain.
and are unnoticed, they can cause collapse Hepatomegaly and liver dysfunction may
of the vitreous, traction on the retina, and be caused by a combination of intrahepatic
eventually cause retinal detachment. Retinal trapping of sickled cells, transfusion‐acquired
Sickle Cell Disease 43

infection, or transfusional hemosiderosis. obtained prior to any transfusion (this


The combination of hemolysis, liver dysfunc­ should be part of routine supportive care
tion, and renal tubular defects can result in and done in advance, anticipating an emer­
very high bilirubin levels. Benign cholestasis gent need for transfusion at some point).
of sickle cell disease may result in severe Sickle cell patients should receive pheno­
asymptomatic hyperbilirubinemia without typically matched blood to decrease the risk
fever, pain, leukocytosis, or hepatic failure. of alloimmunization. In addition, all blood
should be sickledex negative and leukode­
pleted. The following transfusion guidelines
Perioperative management are recommended. See also Chapter 5.
of sickle cell patients a. Patients with hgb SS or Sβ0 disease
who are undergoing any procedure other
Following are the guidelines for the man­ than myringotomy tube placement
agement of sickle cell patients who have should have a simple transfusion to
surgical procedures or require anesthesia achieve a hemoglobin of 10–12 g/dl. Do
for other purposes. General anesthesia not exceed 12 g/dl due to the risk of
results in atelectasis and hypoxia, which is stasis/sludging.
poorly tolerated by the patient with sickle b. Patients with hgb SC disease or Sβ+
cell disease; therefore, special precautions who have baseline hemoglobin below
must be taken perioperatively to decrease 10 g/dl may need transfusions to achieve
morbidity and mortality associated with a hemoglobin of 10–12 g/dl. However,
even minor surgical procedures. Other risk do not transfuse these patients prior to
factors include the presence of chronic consulting the hematologist. Many fac­
organ damage in some patients, the effects tors may affect this decision including a
of asplenia, and the propensity for sickling history of significant complications of
and obstruction of microvasculature with their sickle cell disease (i.e., pneumonia,
even mild hypoxia. The guidelines suggested VOEs, priapism, and aseptic necrosis).
in the following text are extrapolated from a c. Consult the hematologist if there is
multicenter randomized trial of transfusion confusion regarding individual patients.
(exchange vs. straight) in the perioperative 7. Patients with central venous catheters or
period. those undergoing dental procedures should
receive antibiotic prophylaxis prior to
surgery.
Preoperative care

1. Admission to the hospital the afternoon Postoperative care


prior to scheduled surgery.
2. Hydration at 1.25× maintenance (IV + PO) 1. Adequate pain management to prevent
the evening and night prior to surgery. splinting and atelectasis, with care to prevent
3. Pulse oximetry on room air (spot checks). narcosis.
4. IS at least 10 times per hour while awake. 2. Pulse oximetry for at least the first
5. Lab work: CBC with differential and 12–24 hours postoperatively.
reticulocyte count, urinalysis, type and 3. IS at least 10 times per hour while awake.
cross, chemistry panel. Be aggressive!
6. Transfusion: All sickle cell patients 4. Ambulation as early as possible, taking
should have a red cell phenotype study into account the specific surgical procedure.
44 Chapter 3

5. Lab work: CBC with reticulocyte count transfusion is indicated. Many patients can
daily. be treated with straight transfusion, though
6. Follow‐up in clinic approximately in severe cases, exchange transfusion or red
1‐week post‐hospital discharge, or sooner if cell pheresis is recommended. Studies sug­
there are ongoing problems. gest early transfusion may prevent progres­
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 oxy­ that early exchange transfusion may limit
gen‐carrying capacity and improves micro­ neuronal damage by improving local oxy­
vascular perfusion by decreasing the genation and perfusion. Chronic transfusion
proportion of sickle red cells (hgb S%). therapy reduces the rate of recurrence and is
Transfusions are given to patients with indicated in all patients after a first stroke.
sickle cell disease to stabilize or reverse an
acute medical complication, or as part of Perioperative transfusion
chronic therapy in certain situations to Patients with sickle cell anemia undergoing
prevent future complications. major surgery should be prepared in
advance by transfusion to a hemoglobin of
Indications approximately 10 g/dl and a decrease in hgb
Acute illnesses S% to approximately 60%. While standard
Splenic sequestration practice guidelines have not been developed
Transient red cell aplasia with severe for hgb SS patients undergoing minor pro­
anemia cedures or those with hgb SC, it is generally
Hyperhemolysis (infection, ACS) acceptable to not transfuse these patients,
Patients should be transfused if there is assuming they are medically stable.
evidence of cardiovascular compromise
(heart failure, dyspnea, hypertension, or Chronic transfusion therapy
marked fatigue). Generally, transfusion is Chronic transfusions are indicated in several
indicated if the hemoglobin falls below conditions in which the potential medical
5 g/dl or drops greater than 2 g/dl from the complications warrant the risks of alloim­
steady state, or with any cardiovascular munization, infection, and transfusional
instability related to anemia. iron overload. Transfusions are given every
3–4 weeks, with a goal to maintain the hgb
Sudden severe illness S% at 30–50%. While straight transfusions
ACS, hypoxia are acceptable, red cell pheresis or exchange
Stroke transfusions may be preferred to decrease the
Sepsis rate of iron overload.
Acute multiorgan failure
These life‐threatening illnesses are often Indications
accompanied by a falling hemoglobin. 1. Primary stroke prevention
Transfusion therapy has become standard 2. Pulmonary hypertension/chronic lung
medical practice in the management of disease
these illnesses. When ACS is associated 3. Frequent ACS
with hypoxia and a falling hemoglobin, 4. Chronic debilitating pain
Sickle Cell Disease 45

Other indications for transfusions: patients 2 years of age and older. Current
Transfusions are sometimes suggested recommendations for HU usage include the
for a number of conditions in which efficacy following:
is unproven, but may be considered under ●● Frequent episodes of VOE including

severe circumstances. Such circumstances dactylitis.


include: ●● ACS.

Acute priapism ●● Patients with abnormal TCD or history

Pregnancy (frequent complications) of stroke who have received at least 1 year


“Silent” cerebral infarcts of chronic transfusion and have no MRA‐
Leg ulcers defined vasculopathies; HU can substi­
tute for chronic transfusion to maintain
TCD velocities and help prevent primary
Hydroxyurea therapy stroke.
As experience grows with HU therapy,
HU was originally utilized as an antineo­ practitioners are recommending HU as
plastic agent, but due to its effects on prophylaxis in more patients. Some would
increasing fetal hemoglobin (hgb F), it has argue that all patients with hgb SS and Sβ0
gained favor in the treatment of patients thalassemia should be on HU therapy; this
with sickle cell disease. HU is a myelosup­ may become the standard of care in the
pressive agent that inhibits ribonucleotide future.
reductase leading to downstream cytotoxic­ HU therapy dosing begins at 20 mg/
ity of erythroid progenitors leading to kg/d and should be titrated based on ben­
upregulation of fetal hemoglobin produc­ eficial increment in hgb F% and resultant
tion by an unclear mechanism. In addition, increase in hemoglobin and mean cor­
hydroxyurea’s cytotoxic effects lead to leu­ puscular volume (MCV). Potential toxici­
kopenia and neutropenia, beneficial in ties with HU therapy that should be
sickle cell disease secondary to the viscous monitored closely include neutropenia,
properties of these cells during vaso‐occlu­ leukopenia, and anemia. Elevation in ala­
sion. The Multicenter Study of Hydroxyurea nine aminotransferase (ALT) and throm­
in Sickle Cell Anemia (MSH) demonstrated bocytopenia can also occur, though less
HU’s effectiveness in decreasing VOEs and commonly.
ACS in addition to increasing levels of hgb F.
A meta‐analysis demonstrated reduction in
hospital days and further studies confirmed Novel agents
decreased costs of care. In an extension of
the MSH trial, patients who had a cumula­ Glutamine
tive HU exposure of 15 years had reduced l‐glutamine is an essential amino acid
mortality compared with patients who did involved in many pathways throughout the
not receive HU. body including nitrogen transport, catabolic
Long‐term adult studies have shown no signaling, and acid–base homeostasis. It is a
significant complications of HU usage and, required element for the synthesis of other
given the impact on quality of life, addi­ amino acids, proteins, nucleic acids, nucleo­
tional data have continued to support its use tides, and hexosamines. It is a precursor for
in pediatric patients as young as 9 months the synthesis of glutathione (GSH), nicoti­
of age. It is currently approved for use in namide adenine dinucleotide (NAD), and
46 Chapter 3

arginine, all of which have a role in prevent­ episodes, recurrent ACS, stroke, and end‐
ing erythrocyte oxidative stress and con­ organ damage should be considered for
tributing to maintaining vascular tone. It unrelated bone marrow or umbilical cord
was approved in 2017 by the United States blood transplantation when a matched
Federal Drug Administration as an oral sibling donor is not available, ideally in the
agent for patients with sickle cell disease setting of a clinical trial at a center with
5 years and older to reduce the frequency of expertise in the management of patients
vaso‐occlusive events and hospitalizations. with SCD in the peri‐transplant period.
Work to date shows only a modest decre­ Given the paucity of donors for the non‐
ment in VOEs in pediatric patients as com­ Caucasian population, studies regarding
pared with adults. Combination therapy haploidentical transplant in patients with
with HU can be considered after optimizing severe disease manifestations and without a
the dose of HU, and it may be of benefit to more suitable donor are also underway.
the few individuals unable to tolerate HU. Due to underlying vasculopathy,
Crizanlizumab is a monoclonal antibody patients with SCD are at particular risk of
binding P‐selectin which impacts vascular neurovascular events during the transplant
adhesion. Upregulation of P‐selectin in period which may occur secondary to the
endothelial cells and platelets contributes to preparative chemotherapy (i.e., busulfan)
the cell–cell interactions involved in the or secondary to medications used as part
pathogenesis of vaso‐occlusion and sickle‐ of the post‐transplant immunosuppressive
cell‐related complications including pain. regimen (i.e., cyclosporine, tacrolimus,
Crizanlizumab has shown benefit in adult sirolimus). Strict management of blood
patients in a phase II study with a low inci­ pressure parameters must be utilized with
dence of adverse events. The median num­ appropriate use of antihypertensives as
ber of uncomplicated crises was decreased needed.
with the addition of crizanlizumab, though
there was no impact on hospitalization days.
Voxelotor is an agent that increases Gene therapy
hemoglobin’s affinity for oxygen, theoreti­
cally decreasing sickle hemoglobin polym­ Early phase trials have commenced on the
erization and subsequent sickling. Studies utilization of gene therapy to improve nor­
are continuing on the potential benefit of mal hemoglobin A production in order to
this intervention. decrease the proportion of sickle cells, ide­
ally resulting in a relatively asymptomatic
sickle cell trait phenotype. Patients undergo
Hematopoietic stem cell mobilization of their mononuclear cells
transplantation in sickle cell which are subsequently removed through a
disease peripheral blood apheresis or bone marrow
harvest procedure prior to processing to
Hematopoietic stem cell transplantation is alter the β‐globin. The processed cells are
curative for patients with sickle cell disease subsequently reinfused in the patient after a
and should be considered in young patients myeloablative preparative regimen. Gene
with hemoglobin SS and Sβ0 thalassemia therapy offers the benefit of a non‐allogeneic
who have a matched sibling donor. transplant with an improved safety profile
Patients with severe manifestations of and mitigates the need to find a suitable
sickle cell disease including multiple pain donor. Further study is required to deter­
Sickle Cell Disease 47

mine the safety, viability, and longevity of antibiotics, and hospital admission and
these genetically altered cells. observation. You should again reinforce
the need for the child to be on penicillin
prophylaxis. Finally, one should review the
Case studies for review child’s vaccinations to ensure they are up to
date, especially for encapsulated bacteria
1. You are a primary care physician taking including S. pneumoniae, H. influenzae, and
care of a 2-year-old with known sickle cell N. meningitidis.
disease (hgb SS). You are counseling the Regarding episodes of pain, although it is
family on particular risks for the patient at reassuring the child has had a relatively
this age. unremarkable course to date, VOE can start
a. What are some of the main areas of at any age. At 2 years of age, the child is still
discussion? at risk for developing episodes of dactylitis.
In evaluating the patient as a whole, one must The family should be advised on the signs
first discuss growth and development and the and symptoms to monitor for pain in a
risks of low hemoglobin on brain develop­ young child which may present as irritability,
ment. Assuming the patient has had previous refusal to walk, decreased appetite, as well as
laboratory studies, past hemoglobin values localized swelling and/or tenderness. A pain
can help objectively guide this discussion, as plan should be established with the family
phenotypic disease is related to level of ane­ which should include the availability of oral
mia and concomitant reticulocytosis, amount medications at home (e.g., acetaminophen,
of leukocytosis, as well as evidence of ibuprofen, Tylenol with codeine, and/or
hemolysis (total bilirubin and LDH levels). Lortab elixir), as well as adjuvant strategies
In this discussion, newer treatment modali­ such as warm bath and heating pads.
ties including hydroxyurea therapy and the Finally, the practitioner should discuss
potential for hematopoietic stem cell trans­ other potential acute complications such as
plantation should be discussed. splenic sequestration. If the child has a palpa­
Fortunately, this child has had a relatively ble spleen, the family should be advised on
benign course to date. the current size and to monitor for changes in
b. What are some of the potential acute size if the patient appears to be pale or weak
risks that should again be discussed with as a sign of splenic sequestration. In addition,
the family? if the child does appear to have these symp­
The major risks to the child include severe toms without a change in the size of the
infection, pain including dactylitis, and spleen or does not have a palpable spleen, the
splenic sequestration. These risks should all family should be advised that these symptoms
be discussed in detail with the family. should still lead to prompt evaluation, as they
In terms of severe infection, the family could be signs of an aplastic crisis.
should be aware that they should monitor
the child closely if the child appears ill and 2. You are seeing parents for a prenatal
check the temperature before administering evaluation prior to the birth of their first
antipyretics, even if given for pain alone. In child. The mother of the child has sickle cell
addition, they should be aware that if the disease with β0 thalassemia (hgb Sβ0) and
child appears ill, they should immediately the father has sickle cell trait (hgb AS). The
call the on-call physician and will likely mother has been receiving blood transfu­
require immediate evaluation including sion during the pregnancy and it has other­
blood work, chest radiography, parenteral wise gone asymptomatically.
48 Chapter 3

a. What are the important genetic A (i.e., FA). Note that in conditions car­
implications in this case? rying a β-thalassemia mutation beyond
Given the known genotypes of the parents, β-thalassemia major (which will present
you should advise that they have a 25% with newborn screen of F only), hgb electro­
chance of having a child with hgb SS, a 25% phoresis done after 6 months of age when
chance of hgb Sβ0, a 25% chance of sickle fetal hgb production has abated is necessary
cell trait, and a 25% chance of β-thalassemia for diagnosis, showing increased production
trait, dependent on which combination of of hgb F and hgb A2 (see Chapter 4 for more
β-globins is passed on to the child in a information).
Mendelian pattern. You can advise them the
hgb SS and hgb Sβ0 phenotypes should likely
be similar to the mother’s level of disease Multiple choice questions
due to lack of hgb A production in both cases,
knowing there is some level of individual 1. You are seeing a 2-year-old female in the
phenotypic variability. You can advise that ED with Hgb SS disease. She has been gener­
the child with sickle cell trait should be gen­ ally well up to this point with only one pre­
erally asymptomatic, though it is still advisa­ vious admission for dactylitis at 9 months of
ble to monitor fluid intake with high levels of age. The parents note that she has been pale
strenuous activity and at altitude; in the rare appearing over the last 24 hours with
patient, these scenarios may precipitate increased fatigue and decreased appetite.
symptoms of vaso-occlusion. The child with She has been afebrile with no URI symp­
β-thalassemia trait will also be asymptomatic toms or rash. The most important consid­
with a mild microcytic anemia that should eration to first rule out is:
not be mistaken for iron deficiency anemia a. Vaso‐occlusive episode
given the low MCV in both conditions. b. Acute chest syndrome
b. How will newborn screening be c. Splenic sequestration
useful? d. Encapsulated bacteremia
Screening for hemoglobinopathies in the e. Aplastic crisis from parvo virus
United States is now standard of care with Explanation: Although all of the above condi­
hemoglobin electrophoresis, especially due tions are possible diagnoses, the most imme­
to the utility of diagnosing SCD and diate life‐threatening complication in a young
β-thalassemia major at birth. In the normal child is splenic sequestration which can lead
newborn, fetal hemoglobin (hgb F) will pre­ to rapid, uncompensated severe anemia.
dominate with less hgb A production (i.e., Exam should reveal a spleen that is much
normal newborn screen FA; written with larger than the baseline examination. Type &
highest percentage hgb first). For the patient screen and rapid administration of PRBC
with sickle cell trait, the normal β-globin transfusion should occur to improve anemia
will produce more hgb A than the β-globin and ease the splenic sequestration. As the
with the mutation leading to the production child is without fever or pain and has no ante­
of hgb S (i.e., newborn screen FAS). For the cedent history of viral symptoms, choice a.,
patient with hgb SS or Sβ0, neither β-globin b., d. and e. are all less likely. The answer is c.
will produce hgb A (i.e., newborn screen
FS). And, for the patient with β-thalassemia 2. A 12-year-old female with Hgb SS is admit­
trait, due to the hgb F production at birth, ted to the hospital for a vaso‐occlusive crisis.
the newborn screen will be normal, as She was admitted with the complaint of ster­
the one normal β-globin will produce hgb nal, back and leg pain which is the usual loca­
Sickle Cell Disease 49

tion for her vaso‐occlusive events. She was


Suggested reading
initially afebrile without respiratory symptoms
but overnight while inpatient she has devel­
Chou, S.T. (2013). Transfusion therapy for sickle
oped fever with tachypnea, tachycardia and cell disease; a balancing act. Hematology Am.
hypoxia. Chest X‐ray reveals a white out of her Soc. Hematol. Educ. Program 1: 439–446.
R lung. The most important next step is: Kassim, A.A., Galdanci, N.A., Pruthi, S., and
a. Straight transfusion of PRBCs DeBaun, M.R. (2015). How I treat and man­
b. Exchange transfusion of PRBCs age strokes in sickle cell disease. Blood 125:
c. Initiation of antibiotics 3401–3410.
d. Initiation of oxygen therapy Piel, R.B., Steinberg, M.H., and Rees, D.C. (2017).
e. Incentive spirometry Sickle cell disease. N. Engl. J. Med. 376:
Explanation: Although all of the above are 1561–1573.
Rees, D.C., Olujohungbe, A.D., Parker, N.E. et al.
important management steps, a reduction in
(2003). Guidelines for the management of the
the hemoglobin S% is the most important step
acute painful crisis in sickle cell disease. Br. J.
to prevent significant morbidity and mortal­ Haematol. 120: 744–752.
ity. Although exchange transfusion is ideal to Vichinsky, E.P., Neumayr, L.D., Earles, A.N. et al.
reduce the hb S%, as it is overnight, it will take (2000). Causes and outcomes of the acute
time to arrange this procedure and in the chest syndrome in sickle cell disease. National
interim it is best to begin straight transfusion. Acute Chest Syndrome Group. N. Engl. J. Med.
Initiation of antibiotics and symptomatic 342: 1855–1865.
oxygen therapy are also important supportive Ware, R.E. (2015). Optimizing hydroxyurea ther­
care measures that should occur in conjunc­ apy for sickle cell anemia. Hematology Am.
Soc. Hematol. Educ. Program 1: 436–443.
tion with transfusion. The answer is a.
4 Thalassemia

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 hemo­ alpha‐globin tetramers that accumulate in
globin, resulting in a microcytic anemia. the erythroblast. These aggregates are very
The 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 pat­ anemia.
terns, there is an increase in the incidence of An excess of beta‐globin chains (alpha‐
thalassemia in North America, primarily thalassemia) leads to tetramers of beta‐glo­
because of immigration from Southeast bin, hgb H. These tetramers are more stable
Asia. Like with sickle cell anemia, develop­ and soluble, but as the red cell ages in the
ment of thalassemia is directly related to circulation and, under conditions of oxidant
evolutionary pressure secondary to malaria. stress, hgb H precipitates and interferes with
Normally, ≥95% of adult hemoglobin cell membrane function leading to an
found on electrophoresis is hgb A (α2β2). increase in hemolysis.
Two minor hemoglobins occur: 2–3.5% is
hgb A2 (α2δ2) and ≤2% is hgb F (α2γ2). A
mutation affecting globin chain production Alpha‐thalassemia
or deletion of one of the globin chains leads
to a decreased production of that chain and The alpha‐thalassemias are caused by a
an abnormal globin ratio. The globin that is decrease in the production of alpha‐globin
produced in normal amounts is in excess due to a deletion or mutation of one or more
and forms aggregates or inclusions within of the four alpha‐globin genes located on
the red cells. These aggregates become chromosome 16. Alpha‐gene mapping can be
oxidized and damage the cell membrane
­ obtained to determine the specific mutation.
leading to ineffective erythropoiesis, hemol­ The alpha‐thalassemias are categorized as:
ysis, or both. The quantity and properties silent carrier, alpha‐thalassemia trait, hemo­
of these globin chain aggregates determine globin H disease, hemoglobin H‐constant
the phenotypic characteristics of the spring, and alpha‐thalassemia major. Fre­
thalassemia. quently, the diagnosis of alpha‐thalassemia

Handbook of Pediatric Hematology and Oncology – Children’s Hospital & Research Center Oakland,
Third Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
© 2021 John Wiley & Sons Ltd. Published 2021 by John Wiley & Sons Ltd.
52 Chapter 4

trait in a parent is discovered after the birth trait. Mutations in trans (αα/−) are most
of an affected child. likely in Southeast Asians, therefore these
Silent carrier status is characterized by populations have an increased chance of
three functional genes for α‐globin: (−α/αα). having progeny with hemoglobin H or
There is overlap between the red blood cell alpha‐thalassemia major.
indices of these individuals and normals, Hemoglobin H (−/−α) should be consid­
although the mean corpuscular volume ered in the case of a neonate in whom all of
(MCV) may be slightly lower. This state is the red blood cells are very hypochromic.
deduced when a “normal” individual has a Neonates who have hgb H will also have a
child with hgb H disease or with microcytic high percentage of hgb Bart’s on their new­
anemia consistent with alpha‐thalassemia born screen (~20%). In children, this hemo­
trait. An unusual case of the silent carrier globinopathy is characterized by moderate
state is the individual who carries the hemo­ anemia with a hemoglobin in the 8–10 g/dl
globin constant spring mutation (αcsα/αα). range, hypochromia, microcytosis, red cell
This is an elongated α‐globin due to a termi­ fragmentation, and a fast migrating hemo­
nation codon mutation. Individuals who globin (hgb H) on electrophoresis.
have this mutation have normal red blood Hemoglobin H does not function as a
cell indices but can have children who have normal hemoglobin, and has a high oxygen
hgb H‐constant spring if the other parent has affinity, so the measured hemoglobin in
alpha‐thalassemia trait (αα/−). Generally, these children is misleading. Individuals
these children are more affected clinically who have hgb H generally have a persistent
than children who have hgb H. stable state of anemia that may be accentu­
Individuals who have alpha‐thalassemia ated by increased hemolysis during viral
trait (−α/−α) or (αα/−) are identified by infections and by exposure to oxidant medi­
microcytosis, erythrocytosis, hypochromia, cations such as sulfa drugs, chemicals such
and mild anemia. The diagnosis is made by as benzene, and foods such as fava beans,
genetic studies, ruling out both iron‐defi­ similar to individuals who have G6PD defi­
ciency anemia and beta‐thalassemia trait. In ciency. As the red cells mature, they lose
the neonatal period, when hemoglobin their ability to withstand oxidant stress and
Bart’s (γ4) is present (~5% on newborn hgb H precipitates, leading to hemolysis.
screen), the diagnosis can be suspected These precipitates in the RBC can be seen
(though Bart’s at this quantity is not notated by using a brilliant Cresyl blue (BCB) stain.
on most newborn screens). In children, Therapy for individuals who have hgb H
there are no markers such as elevated hgb disease includes folate, avoidance of oxi­
A2 and hgb F (as seen in beta‐thalassemia dant drugs and foods, genetic counseling,
trait) to make the diagnosis. The diagnosis is education, and frequent medical care.
one of exclusion and is often mistaken for Uncommon occurrences in a child with
iron‐deficiency anemia secondary to the hgb H would be severe anemia, cholelith­
microcytosis. Clues for the diagnosis include iasis, skin ulceration, and splenomegaly
a normal red cell distribution width (RDW) requiring splenectomy. Unlike i­ndividuals
and an increase in red blood cells for the who have beta‐thalassemia, hemosiderosis
level of hemoglobin. During pregnancy, is rare in hgb H disease.
microcytic anemia due to thalassemia can be Children with hemoglobin H‐constant
mistaken for ­anemia of pregnancy (physi­ spring (−/αcsα or αcs−/α–) have a more
ologic or iron ­deficiency), and may be a severe course than children who have hgb
clue to a family history of alpha‐thalassemia H. They have a more severe anemia, with a
Thalassemia 53

steady‐state hemoglobin ranging between 7 ment provided. Studies are underway


and 8 g/dl. They more frequently have sple­ regarding early diagnosis and utilization of
nomegaly and severe anemia with febrile ill­ in utero transplantation.
nesses and viral infections, often requiring
transfusion. If anemia is chronically severe
and the child has splenomegaly, a splenec­ Beta‐thalassemia
tomy may be performed. However, a known
complication of splenectomy in this popula­ Beta‐thalassemia is caused by mutations in
tion is severe post‐splenectomy thrombocy­ the beta‐globin gene. Although there have
tosis with hypercoagulability leading to been hundreds of mutations identified
thrombosis of the splenic vein or hepatic within the beta‐globin gene locus, about 20
veins. This complication has also been different alleles make up about 80% of the
reported as recurrent pulmonary emboli mutations found worldwide. Within each
and clotting diathesis. Children who are geographic population, there are unique
scheduled to have a splenectomy are treated mutations. Individuals with beta‐thalas­
with low‐molecular‐weight heparin, fol­ semia major are usually homozygous for
lowed by low‐dose aspirin, continued one of the common mutations (as well as
indefinitely. having one of the geographically unique
The most severe form of alpha‐thalas­ mutations) that lead to the absence or decre­
semia is alpha‐thalassemia major (−/−). ment of beta‐chain production.
This diagnosis is frequently made in the last The beta‐thalassemia syndromes are
months of pregnancy when fetal ultrasound much more diverse than the alpha‐thalas­
indicates a hydropic fetus. Families who semia syndromes due to the diversity of the
have genetic counseling and appropriate mutations that produce the defects in the
testing may have much earlier diagnosis in beta‐globin gene. Unlike the deletions that
the first trimester. The mother frequently constitute most of the alpha‐thalassemia
exhibits toxemia and can develop severe syndromes, beta‐thalassemias are caused by
postpartum hemorrhage. These infants are mutations on chromosome 11 that affect all
usually stillborn. There can be other con­ aspects of beta‐globin production: tran­
genital anomalies, though none are pathog­ scription, translation, and the stability of the
nomonic for alpha‐thalassemia major. beta‐globin product. Most hematologists
Because of in utero hypoxia, the hemoglob­ feel there are three general categories of
ins found in these infants are hgb Portland beta‐thalassemia: beta‐thalassemia trait,
(ζ2γ2), hgb H (β4), and hgb Bart’s (γ4), and no beta‐thalassemia intermedia, and beta‐
hgb A, A2, or F. These babies often have thalassemia major. However, with the lack of
other complications associated with hydrops genotypic differentiation, there remains
such as heart failure, anasarca, and pulmo­ phenotypic overlap between these three
nary edema. general categories.
If the diagnosis is made early, intrauter­ Splice site mutations also occur and are
ine transfusions can be performed. There of clinical consequence when combined
are reports of survival with chronic transfu­ with a thalassemia mutation. Three splice
sion in these infants, now more recently site mutations occur in exon 1 of the beta‐
replaced by curative hematopoietic stem cell globin gene. These mutations result in three
transplantation. Undoubtedly, more of these different abnormal hemoglobins: Malay, E,
infants could be saved if the diagnosis was and Knossos. Hemoglobin E is a very
anticipated by prenatal diagnosis and treat­ common abnormal hemoglobin in the
­
54 Chapter 4

Southeast Asian population, and when beta‐thalassemia trait. The red blood cell
paired with a β0‐thalassemia mutation indices can be helpful in this differentiation,
can produce severe transfusion‐dependent as the hemoglobin concentration and the
thalassemia. Hemoglobin E is described in red cell count will generally be lower in iron
the section on newborn screening. deficiency. The distinguishing finding in
Individuals who have beta‐thalassemia beta‐thalassemia is a hemoglobin electro­
trait have microcytosis and hypochromia; phoresis with an elevated hgb A2 and F. Both
there may be target cells and elliptocytes will be increased in beta‐thalassemia trait
seen on a peripheral blood smear, though without iron deficiency, and will be normal
some individuals have normal morphology. or decreased in alpha‐thalassemia and iso­
These hematologic features can be accentu­ lated iron‐deficiency anemia. There are sev­
ated in women with trait who are pregnant eral formulas to help in office screening, but
and individuals who are folate or iron defi­ they are based on the assumption that the
cient. If iron deficiency is concurrent with child is not iron deficient. Usually, iron defi­
beta‐thalassemia trait, there may be a rela­ ciency can be ruled out using FEP, transfer­
tively normal hgb A2, potentially con­ rin saturation, or ferritin as a screening test
founding the diagnosis. Free erythrocyte in children who have a hypochromic micro­
protoporphyrin (FEP) is elevated in iron cytic anemia. The least expensive test is a
deficiency and lead poisoning, while normal trial of iron and a repeat hemogram after a
in α‐ and β‐thalassemia, making it a poten­ month. A lead level should be obtained if
tially helpful test in this situation. Iron there is an index of suspicion for lead
deficiency causes decreased hemoglobin
­ toxicity.
production, and folate or vitamin B12 defi­ Diagnostic problems may still arise if
ciency can lead to megaloblastic anemia both alpha‐thalassemia and beta‐thalas­
with increased hgb A2. Both of these defi­ semia coexist, since the changes in hgb A2
ciencies need to be treated prior to evalua­ and F will not be apparent as noted in the
tion for thalassemia trait. In iron‐, B12‐, and preceding text. Family studies and DNA
folate‐replete individuals, the hgb A2 can analysis can be used to make a definitive
be as high as 3.5–8% and the hgb F as high diagnosis.
as 2–5%. Generally, beta‐thalassemia trait is Children who are diagnosed with thalas­
milder in African‐Americans (who fre­ semia intermedia have a homozygous or
quently have a promoter gene mutation), heterozygous beta‐globin mutation that
but has a similar presentation in individuals causes a greater decrease in beta‐chain pro­
of Chinese, Southeast Asian, Greek, Italian, duction than seen in thalassemia minor, but
and Middle Eastern heritage. not to the degree for which chronic transfu­
Infants born in most states in the United sion therapy is required. The phenotype can
States are screened for hemoglobinopathies also occur in children who have a mutation
(all states are required to perform screening that increases production of α‐globin
for sickle cell disease). In states without (alpha‐triplication) leading to increased dis­
newborn screening for hemoglobinopathies symmetry between alpha‐ and beta‐globin
and in recent immigrants to this country, chains. Children who have thalassemia
affected children are frequently found later intermedia are able to maintain a hemo­
than the newborn period, and the evalua­ globin of 7 g/dl or slightly higher with a
tion of their microcytic anemia includes greatly expanded erythron and may mani­
­differentiation between iron deficiency and fest bony deformities, pathological fractures,
Thalassemia 55

and growth retardation. Children who individual patient. Splenectomy has been
have thalassemia intermedia can also have utilized but now is thought to be at least
delayed pubescence, exercise intolerance, partly responsible for the development of
leg ulcers, inflammatory arthritis, and pulmonary hypertension and thrombosis
extramedullary hematopoiesis causing spi­ secondary to chronic hemolysis. Some argue
nal cord compression—a medical emer­ that transfusion therapy is underutilized in
gency requiring radiation therapy and thalassemia intermedia and improved risk
transfusion. They can also have iron over­ stratification is needed.
load due to increased absorption of iron Thalassemia major was first described by
from the gastrointestinal tract and intermit­ a Detroit pediatrician, Thomas Cooley, in
tent transfusion. They are at risk for the 1925. The clinical picture he described is
cardiac and endocrine complications of
­ prevalent today in countries without the
hemosiderosis, but usually at an older age necessary resources to provide patients
than chronically transfused children. with chronic transfusions and iron chelation
Chelation therapy is indicated for increasing therapy. Children who have untreated
ferritin and elevated liver iron. thalassemia major have ineffective erythro­
Children who cannot maintain a hemo­ poiesis, decreased red cell deformability,
globin between 6 and 7 g/dl should have an and enhanced clearance of defective red
alternative diagnosis considered. If thalas­ cells by macrophages. The result is a very
semia is the cause of the anemia, transfusion hypermetabolic bone marrow with throm­
and/or splenectomy should be considered. bocytosis, leukocytosis, and microcytic ane­
Frequently, adolescents and adults are una­ mia in young children prior to enlargement
ble to tolerate the degree of anemia that of the spleen. At presentation, they have
is seen in thalassemia intermedia. Poor almost 100% hgb F (these cells have a longer
growth, hypersplenism, splenic pain, con­ life span due to a balanced globin ratio as γ,
gestive heart failure, severe exercise intoler­ rather than β, globin is present in hgb F).
ance, thrombocytopenia, and leukopenia These children have little or no hgb A2 and
should be considered indications for trans­ a low reticulocyte count. The diagnosis can
fusion and/or splenectomy. be made with certainty by demonstrating
Appropriate clinical management of thalassemia trait in both parents, by globin
thalassemia intermedia patients may be biosynthetic ratios, or by beta‐gene screen­
more difficult than patients with thalas­ ing. Beta‐gene screening identifies the most
semia major requiring chronic transfusion common and some uncommon mutations,
due to phenotypic heterogeneity. Patients but not all mutations. An electrophoresis
with clinically mild disease still may have showing only hgb F, a complete blood count,
serious long‐term complications as and a peripheral blood smear will generally
described in the earlier text due to ineffec­ be diagnostic. In most states, these children
tive erythropoiesis as well as chronic hemol­ will be discovered by newborn screening or
ysis, leading to pulmonary hypertension occasionally by the obstetrician who makes
with resultant congestive heart failure and a diagnosis of thalassemia trait in the mother
thrombosis in addition to cholelithiasis. and obtains a family history of thalassemia
Currently, transfusion therapy is limited to or anemia in both parents prior to the birth
patients with symptomatic anemia or in of the baby.
children with delayed growth and deve­ Children who have untreated thalas­
lopment, and is generally tailored to the semia die in the first decade of life from
56 Chapter 4

a­nemia, septicemia, and pathologic frac­


Hereditary persistence of fetal
tures. When palliative transfusions are
hemoglobin
introduced, children live into their late teens
eventually succumbing to heart failure due
HPFH is also due to deletions in the δ‐ and
to iron overload.
β‐globins, which leads to an increase in hgb
With the introduction of frequent
F (usually 15–30%). Association of HPFH
chronic transfusion therapy with regular
with other β‐hemoglobinopathies can miti­
iron chelation, children are now surviving
gate clinical manifestations. HPFH leads to
into adulthood, adding to the complexity of
pancellular expression of hgb F, which can
the disease. Patients with a matched sibling
distinguish it from delta beta thalassemia.
donor should be offered transplantation;
haploidentical transplant may be an option
within the bounds of a clinical trial. Studies Neonatal screening
utilizing genetic engineering of mononu­ for hemoglobinopathies
clear cells collected by apheresis to produce
hgb A or hgb F which are subsequently Newborn screening identifies patients with
reinfused to the patient after a myeloabla­ beta‐thalassemia major, hemoglobin H, and
tive preparative regimen are underway with sickle cell disease. Patients with beta‐thalas­
promising results to date. The longevity of semia major and sickle cell disease will be
patients who are compliant with their chela­ asymptomatic at birth due to high fetal
tion therapy, or in those who have received hemoglobin levels but will become sympto­
bone marrow transplantation, is not yet matic over the next 2–3 months as normal
known. production of hgb F wanes. Patients with
significant Bart’s hemoglobin (see Table 4.1)
should be considered to have hemoglobin H
Delta beta thalassemia and require alpha‐gene screening to detect
the possibility of hgb H‐constant spring.
Mutations in both δ‐ and β‐globins leads The presence of only hgb F on newborn
to a reduction in hemoglobin A and A2 screen will be interpreted as beta‐thalas­
but with an increase in hemoglobin semia major (see Table 4.2). This diagnosis
F. Delta beta thalassemia trait will lead to must be confirmed with parental electro­
microcytosis (due to beta‐thalassemia trait) phoresis and repeat testing of the infant at
without anemia (due to the increase in 2–3 months after fetal hemoglobin wanes
hemoglobin F). Due to the decrease in A2 with concomitant increase in hemoglobin A
production (similar to iron deficiency), and A2 in the normal patient. The pattern
beta‐thalassemia trait may not be consid­ FA is normal and should not be interpreted
ered, and therefore there must be a high as beta‐thalassemia trait. This is a diagnosis
suspicion for delta beta thalassemia (and in made by the practitioner when microcytic
lieu of hereditary persistence of fetal hemo­ anemia is seen during routine childhood
globin (HPFH) due to the elevated hgb F). screening, and an investigation for thalas­
Homozygous mutations will lead to a semia confirms the diagnosis. All infants
thalassemia intermedia phenotype, while with a suspected hemoglobinopathy should
combination with a second β mutation can be referred to a pediatric hematologist.
lead to thalassemia intermedia or thalas­ Hgb FE will be presumed to be hgb
semia major phenotype. E‐beta‐thalassemia until that diagnosis is
Thalassemia 57

Table 4.1 Classification of the alpha‐thalassemias.

Diagnosis Genetic finding Symptoms Bart’s (%)

Silent carrier α−/αα Hematologically normal 1–3


α‐thalassemia trait α−/α– or αα/− Mild anemia with microcytosis 3–6
and hypochromia
Hemoglobin H α−/− Moderately severe hemolytic 5–20
disease or anemia
Hemoglobin ααCS/− or α−/αCS− Icterus and splenomegaly 5–20
H‐constant spring
α‐thalassemia major −/− Severe anemia not compatible 100
with life; hydrops fetalis
without intrauterine
transfusion

investigated and confirmed or ruled out by exon (exon 1, codon 26: GAG to AAG) that
repeat electrophoresis and family studies. creates an alternate splice site competing
Hemoglobin E is the most common abnor­ with the normal splice site. This results in
mal hemoglobin discovered in the state of abnormal hemoglobin production and mild
California on newborn screening. It is com­ microcytic anemia (hgb ≥ 10 g/dl) in the
mon in Laos, Cambodia, and Thailand. homozygous state. Electrophoresis reveals
Hemoglobin E results from a mutation in an about 90% hgb E with varying amounts of

Table 4.2 Interpretation of common newborn screening results.*

Result Diagnostic possibilities Action required

FA Normal None
β‐thalassemia trait Genetic counseling
F β‐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/β0‐thalassemia Family studies
Referral to hematologist
FSA Hgb S/β+‐thalassemia Referral to hematologist
FSC Sickle cell disease (Hgb SC) Referral to hematologist
FAC Hemoglobin C trait Genetic counseling
FE Hgb E/β‐thalassemia Repeat testing at 2–3 months
Referral to hematologist
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).
58 Chapter 4

hgb F. The heterozygote has a hemoglobin of without any history of recent infection or
about 12 g/dl with microcytosis and an elec­ fever. Beta-thalassemia as well as sickle cell
trophoretic pattern consistent with hgb E anemia should be high in the differential, as
plus hgb A. When combined with other fetal hemoglobin lives 60–90 days and
more severe beta‐thalassemias, hgb E‐beta‐ therefore by 4 months of age the infant will
thalassemia can produce an anemia that is be producing little fetal hemoglobin (α2γ2)
profound, requiring chronic transfusion and should have moved to almost solely
therapy. All children who have hgb E and hemoglobin A (α2β2) unless there is a prob­
hgb F on their state screen require scrutiny lem with the β-globin chain. Additional
for the emergence of a severe thalassemia possibilities include a hemolytic anemia,
syndrome. Individuals who are homozygous transient erythroblastopenia of childhood
for hgb E should not have a significant ane­ (though rarely seen at this young of an age),
mia and do not require special care. Like infant leukemia, and folate and vitamin B12
patients with alpha‐thalassemia, they should deficiency if the baby is being fed goat’s
not be treated with iron for their microcytic milk or the mother is vegan, respectively.
anemia unless they are proven to have con­ b. What additional piece of information
comitant iron deficiency. may be helpful in this young child?
In addition to asking about a family history
of sickle cell anemia and thalassemias, the
Case studies for review provider should look for the results of new­
born screen which could be very helpful in
1. You are seeing an infant that has been this case. If the patient has a newborn
following in your clinic since the newborn screen that is FA, sickle cell anemia and
period. The baby was born full-term with­ β-thalassemia major can be ruled out. More
out any significant issues. You are now see­ concerning newborn screening results
ing the baby back at 4 months for the would include F (β-thalassemia major), FS
well-child check and secondary vaccina­ (hgb SS disease or hgb S/β0-thalassemia),
tions. The parents note that the baby has and FSA (hgb S/β+-thalassemia).
been more listless, not eating as well, and β0-thalassemia and β+-thalassemia are dif­
looking paler. There are no recent infections ferentiated based on the presence (β+) or
or fevers. absence (β0) of hemoglobin A, although
a. Assuming this baby is anemic, what phenotypically the patient can have an
would your differential diagnosis be at extremely variable clinical presentation, in
this point? part due to the persistence of fetal hemo­
Causes of anemia are quite broad at this globin. If the patient had sickle cell trait,
point. Complete blood count with differen­ their newborn screen result would be FAS,
tial, MCV, and reticulocyte count would be as the percentage of hemoglobin A should
helpful to narrow the differential. Iron defi­ be greater than the percentage of hemo­
ciency is unlikely before 6 months of age globin S. If necessary, results can be con­
due to maternal iron stores unless the baby firmed at 4 to 6 months of age after fetal
was born prematurely (and would have been hemoglobin levels should be significantly
routinely put on iron therapy), if the mother decreased or, if possible, the parents can be
had severe anemia during pregnancy, or if tested. In this case, the newborn screen
there was acute or occult blood loss. Viral results are F only.
suppression or aplasia from infections such c. How might repeat hemoglobin elec­
as parvovirus is possible, but less likely trophoresis be different at this age?
Thalassemia 59

The patient that has no hemoglobin A will medical issues. The mother did not note any
be on the extreme end of the clinical spec­ significant issues during the pregnancy.
trum with definitive β-thalassemia major. a. Assuming this may be a thalassemia-
For those patients with some β-globin pro­ related condition, what are the most
duction, the clinical severity of disease at likely diagnoses?
this point cannot be determined on a molec­ Given the lack of frequent transfusion,
ular level. Patients who are solely fetal β-thalassemia major is highly unlikely in a
hemoglobin (α2γ2) on newborn screen will generally well 7-year-old. Also given the
likely continue to make some amount of intermittent periods of symptomatic hemol­
fetal hemoglobin in addition to hemoglobin ysis, β-thalassemia and α-thalassemia trait
A2 (α2δ2). Their repeat hemoglobin electro­ are not possible diagnoses. The most likely
phoresis will represent this with variable diagnosis would be hemoglobin H or
amounts of hemoglobin A2 and F and no H-constant spring given the intermittent
hemoglobin A. periods of jaundice and minimal transfu­
d. What would you expect the parents’ sion needs. β-thalassemia intermedia is also
hemoglobin electrophoresis to show? possible, though less likely. Splenomegaly
The parents likely both have β-thalassemia on exam could be diagnostic of both condi­
trait. If they were tested as newborns, they tions, though a more prominent spleen
would be FA. As adults, after fetal hemo­ would be most consistent with hemoglobin
globin production has decreased, they H or H-constant spring.
would have increased amounts of both A2 b. What would be the best next testing?
(>3.5%) and F (>2%) in addition to hemo­ Basic labs including a CBC and complete
globin A. metabolic panel should always be under­
e. What other clinical signs might be taken. With the assumption of an underly­
apparent in the infant? ing thalassemia condition, hemoglobin
One would expect to see increasing signs of electrophoresis should be done in addition
extramedullary hematopoiesis with increas­ to genetic testing for α-thalassemia muta­
ing age and decreasing fetal hemoglobin tional deletions. BCB prep will be useful for
production. Some of the early signs could be inclusion bodies (Heinz bodies) that would
frontal bossing as well as maxillary and be present with hemoglobin H or H-constant
mandibular prominence. The infant may spring. In both β-thalassemia intermedia as
have failure to thrive as well as the develop­ well as hemoglobin H there will be a micro­
ment of hepatosplenomegaly. cytic anemia with potential elevation in the
indirect bilirubin secondary to chronic
2. You are seeing a 7-year-old male in your hemolysis. Hemoglobin electrophoresis will
primary care clinic who recently emigrated show a decrement in hemoglobin A and
from Southeast Asia. The family notes that increase in A2 and F with β-thalassemia
he has generally been well, though has peri­ intermedia while should show elevated
ods of jaundice from which he has recovered hemoglobin Bart’s (β4 tetramers) with
without significant issues. He has not hemoglobin H or H-constant spring. The
received frequent medical care in the past, confirmatory diagnosis for hemoglobin H
though the family does note that he had one or H-constant spring will be genetic testing
period of jaundice which required a blood which would report three deletional or non-
transfusion. He has no siblings. The parents functional (constant spring) mutations.
also have not received frequent medical c. What counseling would you give the
care, though do not note any significant family based on the underlying diagnosis?
60 Chapter 4

Although the hemoglobin level can reveal the amount of hemoglobin A is decreased
moderate anemia with hemoglobin H or but not absent, this will also present with a
H-constant spring, the general picture is normal newborn screen of FA as would be
similar to the patient’s history, with infre­ seen in β‐thalassemia trait and β+‐thalas­
quent transfusion requirements and periods semia. α‐thalassemias including hemo­
of jaundice and splenomegaly. The spleen globin H and α‐thalassemia major will
may need to be removed in the future, present with Bart’s hemoglobin (γ4 tetram­
depending on the symptoms and the size of ers) on newborn screen whereas α‐thalas­
the spleen. In the case of β-thalassemia semia trait will have a normal newborn
intermedia, the clinical course is quite vari­ screen. The answer is c.
able, with some patients benefiting from
chronic transfusions, especially if there is 2. You are seeing an 8-month-old male infant
growth delay in the younger patient. On the who recently immigrated from Southeast
other hand, some patients with β-thalassemia Asia with no previous medical care. He had
intermedia may not require or benefit from a sibling that died at a young age of unknown
chronic transfusion due to the lack of sig­ causes. His parents are unaware of any
nificant symptomatology and therefore known medical history. On exam he appears
have an unjustifiable risk of iron overload pale with frontal bossing and a large spleen.
from chronic transfusion. Hemoglobin electrophoresis is most likely
to show which of the following patterns:
a. 98% Hgb A, 1% A2, 1% F
Multiple choice questions b. 93% Hgb A, 4% A2, 3% F
c. 60% Hgb A, 40% Hgb S
1. You are following up on newborn screen d. 98% Hgb F, 2% Hgb A2
results on a healthy 2-week-old male infant e. 100% Hgb S
of Southeast Asian descent with a strong
family history of microcytic anemia. You 3. Match the below with the correct
note that the infant has an FA pattern on condition:
newborn screening. All of the following are a. 9 8% Hgb A, Sickle cell trait
potential diagnoses, EXCEPT: 1% A2, 1% F
a. Normal newborn b. 93% Hgb A, Hgb SS
b. β‐thalassemia trait 4% A2, 3% F
c. β‐thalassemia major c. 6 0% Hgb A, β‐thal trait
d. α‐thalassemia trait 40% Hgb S
e. β+‐thalassemia d. 98% Hgb F, β‐thal major
Explanation: Although not all hemoglo­ 2% Hgb A2
binopathies can be diagnosed at birth, e. 100% Hgb S Normal
β‐thalassemia major will present on new­ Explanation: The percentage of F, A2 and S
born screen with F hemoglobin only. An FA distinguishes between trait conditions and
pattern is usually normal, as the normal disease. Patients with β‐thalassemia major
newborn will produce more fetal hemo­ will not be able to produce any hemoglobin
globin and less adult hemoglobin A. The FA A so in the newborn period they will have
pattern does not characterize how much Hgb F alone (answer d). After birth they may
hemoglobin A is being produced except produce a small amount of Hgb A2. Patients
being less than hemoglobin F. If there is an with β‐thalassemia trait will have increased
abnormality in the β‐globin locus such that production of Hgb F (normally <2%) and Hgb
Thalassemia 61

A2 (normally 2%‐3.5%) after birth (answer Lal, A., Goldrich, M.L., Haines, D.A. et al. (2011).
b). Patients with sickle cell trait will have an Heterogeneity of hemoglobin H disease in
S% of 40% or less, which can also be seen in childhood. N. Engl. J. Med. 364: 710–718.
the frequently transfused patient with sickle Richardson, M. (2007). Microcytic anemia.
Pediatr. Rev. 28: 5–14.
cell anemia (answer c). The patient with Hgb
Taher, A.T., Musallam, K.M., Karimi, M. et al.
SS will produce only Hgb S after the newborn
(2010). Overview on practices in thalassemia
period (answer e). A normal hemoglobin intermedia management aiming for lowering
electrophoresis is represented by answer a. complication rates across a region of ende­
micity: the OPTIMAL CARE study. Blood
115: 1886–1892.
Suggested reading Vichinsky, E., Cohen, A., Thompson, A.A. et al.
(2018). Epidemiology and clinical characteris­
Hoppe, C.C. (2009). Newborn screening for non‐ tics of nontransfusion‐dependent thalassemia
sickling hemoglobinopathies. Hematology in the United States. Pediatr. Blood Cancer 65:
Am. Soc. Hematol. Educ. Program 1: 19–25. e27067.
5 Transfusion Medicine

Transfusion of blood products continues to in previously untransfused patients to elimi­


be an important and necessary part of nate the risk of clerical error.
­therapy in children with hematologic and Directed donation from first‐degree rela­
oncologic diagnoses. Many complications tives (especially maternal) should be dis­
can result from transfusion, both infectious couraged for patients who may be candidates
and noninfectious. With continued improve­ for an allogeneic bone marrow transplant
ments in donor screening, and better testing due to the possibility of antigen sensitiza­
techniques, infections from known entities tion. If it cannot be avoided, the blood
have become a rarity. However, as long as should be irradiated to prevent graft‐versus‐
blood component therapy is derived from host disease (GVHD). Studies have shown
human blood donation, the risk will persist. that blood from directed donation is not
Donor selection criteria are designed to safer than the general donation pool due to
screen out potential donors with increased current sophisticated screening and testing
risk of infection with HIV‐1/2 (human inm­ methods.
munodeficiency virus types 1 and 2), These risks must be carefully considered
HTLV‐I/II (human T‐cell leukemia virus and weighed against expected benefit
types I and II), and hepatitis B and C, as well each time a transfusion is contemplated.
as other infectious pathogens. Despite rigor­ Informed consent should be obtained prior
ous screening and testing for these infec­ to every nonemergent transfusion. In
tions, the risk of transmitting these viruses is California, the Gann Act must be renewed
not totally eliminated. on a yearly basis for those patients undergo­
Clerical errors and misidentification are ing frequent transfusion due to an underly­
major risks for transfusion and can result in ing hematologic or oncologic condition.
serious consequences, including death. It is
essential that all blood samples drawn for a
type and crossmatch be clearly labeled with Packed red blood cell
the patient’s identification. Before adminis­ transfusion
tration of the blood product, the order
should be checked, patient identification Transfusion decisions are made on the basis
reviewed (patient’s identification band), and of clinical context, rather than the level of
blood type verified. Many institutions have hemoglobin alone (see Figure 5.1). Prior
moved toward testing two separate blood to transfusion, it is necessary to assess the
type specimens for nonemergent ­transfusion mechanism responsible for anemia (i.e., bone

Handbook of Pediatric Hematology and Oncology – Children’s Hospital & Research Center Oakland,
Third Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
© 2021 John Wiley & Sons Ltd. Published 2021 by John Wiley & Sons Ltd.
64 Chapter 5

Symptomatic anemic patient

Chronically transfused patient?

Yes No

Give leukopoor, phenotypically Acute blood loss?


matched blood unless emergent Yes
condition does not allow time No

Patient with concern for Oncology patient or neonate?


autoimmune hemolytic anemia?

Review transfusion history Yes No


Yes No
Give 10–15 mL/kg over 4 hours
in majority of cases
Ensure leukopoor, Ensure leukopoor
Start Transfuse to irradiated blood blood
immunosuppressive normal
therapy hemoglobin level
For sickle cell patients:
Give sickledex negative blood Give CMV negative
Avoid Hgb > 12 g/dL blood if CMV
Consider exchange transfusion seronegative in the
(see Chapter 3 for details) If patient Transfuse for peri-transplant
symptomatic give continued blood period*
phenotypically losses
matched blood

Review transfusion history


Give 10 –15 mL/kg over 4 hours
in majority of cases

Figure 5.1 Blood transfusion guidelines (*refer to institutional guidelines, as transfusion of CMV‐seronegative PRBCs is controversial).
Transfusion Medicine 65

marrow infiltration, ineffective erythro­ and alloimmunization from PRBC transfu­


poiesis of chronic disease, occult or obvious sion. Although not universal as yet in the
blood loss, transfusion or drug‐related United States (it is estimated that approxi­
­suppression of normal hematopoiesis, and mately 85% of RBCs distributed to United
nutritional deficiency), the severity of the States hospitals by their blood providers are
signs and symptoms, and the likelihood of prestorage leukoreduced), l­eukoreduction is
resumption of normal hematopoiesis. becoming standard of care and is universal in
Whole blood is rarely, if ever, utilized in Canada and much of Europe.
standard practice, though may occasion­ Irradiation of blood products should
ally be utilized for massive transfusion be utilized for all patients who are immu­
after ­military trauma. Packed red blood nocompromised in order to prevent
cells (PRBCs) are depleted of the majority ­transfusion‐associated graft‐versus‐host
of platelets and white blood cells and need disease (TA‐GVHD). This should include
to be ABO and RhD compatible, at the all oncology patients as well as neonates.
­minimum. Phenotypically, and now more Cytomegalovirus (CMV)‐negative blood
recently genotypically, matched blood for should be considered in patients in the
minor antigens should be utilized for peri‐transplant period who are noted to be
patients undergoing chronic transfusion CMV‐seronegative in addition to neonates
(e.g., sickle cell disease). One unit of born to CMV‐negative mothers and patients
PRBCs is approximately 250–350 ml with a with immunodeficiencies. Institutional
hematocrit of 55–60% when prepared with guidelines vary and should be consulted in
adsol. Based on the hematocrit of the determining the need for CMV‐negative
stored PRBCs, one can calculate the pre­ blood secondary to the limited supply of
sumed rate of rise from blood transfusion. this product.
For adsol‐preserved units, the transfusion Finally, washing of PRBCs is rarely indi­
factor is approximately 5 (assuming no cated and decreases the red cell mass by
hemolysis of transfused blood or blood approximately 20%. Washing is indicated
loss). The estimated hemoglobin (hgb) for severe allergic transfusion reactions,
increase can be derived utilizing this trans­ hyperkalemia, large‐volume transfusions
fusion formula: (due to risk of hyperkalemia), and selective
IgA deficiency.
Estimated hemoglobin rise g/dl
Volume of PRBC transfusion
n ml/kg
Transfusion factor Indications for PRBC
transfusion
For example, for a transfusion of 15 ml/kg,
the estimated hemoglobin increase should Neonates
be 3 g/dl. Of note, waiting a certain period of Guidelines for transfusion in neonates and
time for a posttransfusion “reequilibration” infants vary widely in the literature. Clinical
prior to rechecking the hemoglobin is not correlation is required in addition to the
necessary. ­following basic guidelines:
Removal of the vast majority of remaining
white blood cells, or leukoreduction, is an ●● Transfusion of leukopoor, irradiated
important step in reducing transfusion‐related blood in premature infants and low‐birth‐
risks including risk of infection, febrile non­ weight neonates (CMV‐negative if CMV
hemolytic transfusion reactions (FNHTRs), status of mother is negative or unknown).
66 Chapter 5

●● High oxygen requirement or severe car­ should be transfused slowly with judicious
diac disease, keep hgb ≥ 11–15 g/dl. usage of diuretics, and exchange transfusion
●● Mild‐to‐moderate respiratory distress or should be considered in those with heart
perioperative care, keep hgb ≥ 10 g/dl. failure.
●● Stable infants who become symptomatic

from anemia, keep hgb > 7–8 g/dl (based on Oncology patients
degree of symptoms and growth). In general, a hemoglobin of 7 g/dl is used as
the threshold for transfusion in pediatric
Acute blood loss oncology patients. This may be altered in
and nonimmune hemolytic cases where the patient is asymptomatic
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 com­ count). Infants with effects on growth and
pensation, patients with acute blood loss or development due to anemia should be main­
nonimmune hemolytic anemia may be tained at a higher hemoglobin; similarly,
symptomatic at much higher hemoglobin adolescents may complain of headache and
levels and should be transfused accordingly. fatigue and be less symptomatic with hemo­
In situations of continuing blood loss, globin in the 8–10 g/dl range. Those with
patients should be transfused to achieve an cardiopulmonary dysfunction and those
expected hemoglobin level in addition to requiring procedural sedation with antici­
receiving blood to compensate for ongoing pated blood loss should be kept in the
losses. In an emergent situation, transfusion 8–10 g/dl range as well.
consent is not required, and if there is no
time for a crossmatch, 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 chronic with a continuing drop in hemoglobin or
anemia (i.e., hgb < 5 g/dl), followed by viral resultant significant cardiopulmonary dys­
suppression, transient erythroblastopenia function, phenotypically matched blood
of childhood, aplastic anemia, and newly may be given with close observation know­
diagnosed leukemia. Pediatric patients can ing that hemolysis is likely to continue and
present with an extremely low hemoglobin may even be augmented.
(i.e., 2–3 g/dl) and yet appear to be relatively
well‐compensated. Patients should be assessed Chronically transfused patients
closely for subtle signs of congestive heart Patients with β‐thalassemia major as well as
failure (cardiomegaly on chest radiograph, some with β‐thalassemia intermedia, hgb
increased diastolic blood pressures, hepato­ SS, and hgb S/β0‐thalassemia require chronic
megaly, oliguria, and periorbital edema) transfusion therapy, as outlined in previous
prior to transfusion. Slow transfusion (i.e., chapters. These patients should have a red
1 ml/kg/h) has been recommended in the cell phenotype prior to initiating transfu­
past, although studies have shown that sions and, based on institutional practice,
2–3 ml/kg/h is safe in patients with normal will usually receive blood phenotypically
underlying cardiopulmonary function. Those matched to a small panel of antigens (at our
with signs of cardiopulmonary dysfunction institution, other Rh antigens including C/c
Transfusion Medicine 67

and E/e as well as Kell). If the patient devel­ Exchange transfusion/


ops antibodies to other minor red cell anti­ erythrocytapheresis
gens (e.g., MNS, Duffy [Fya/Fyb], Kidd [Jka/ Exchange transfusion or erythrocytaphere­
Jkb], and Lewis [Lea/Leb]), more extensive sis may be indicated in the severely anemic
phenotype matching is performed in order patient with congestive heart failure, acute
to decrease the continued risk for alloim­ sickle cell events, hyperbilirubinemia, or in
munization. Genotyping for a wider range an anemic patient treated with severe fluid
of antigens is becoming more universally restriction (e.g., increased intracranial pres­
available and will likely become the future sure, ICP). In sickle cell disease, exchange
standard of care. Sickle cell patients in transfusion quickly reduces the concentra­
addition should receive sickledex‐negative tion of sickle cells without increasing the
blood, as blood donors are not screened for hematocrit or whole blood viscosity. In
sickle cell trait. Transfusion is given on a addition, red cell exchange transfusion
regular basis (i.e., every 3–4 weeks) in order reduces iron accumulation, since an equal
to suppress ineffective erythropoiesis. volume of red cells and iron are removed
as infused. Therefore, for sickle cell dis­
Dosing of PRBC transfusion ease, erythrocytapheresis is preferred over
The desired incremental increase in hemo­ straight transfusion in patients requiring
globin should be considered when deter­ chronic transfusion in addition to those
mining the amount of blood to be transfused. patients with acute events. Automated
In general, 10–20 ml/kg over 4 hours is given erythrocytapheresis can be done rapidly and
in order to increase the hemoglobin by safely in most situations. Limitations of this
2–4 g/dl, typically using a maximum of 3 technique include increased red cell utili­
units in consideration of ideal body weight zation, venous access, local availability, and
and total blood volume. Care should be increased cost.
taken to not waste blood; therefore, orders
should be rounded to the nearest unit or half
unit, as feasible. Remaining blood may be
sterilely aliquoted and used later in the same Platelets
patient in order to decrease exposure to
multiple donors. Slow transfusion should Platelet transfusions are indicated for
be given in patients with signs of volume thrombocytopenic patients with bleeding
overload or cardiopulmonary dysfunction. due to severely decreased platelet produc­
Patients with heart failure should be tion or for patients with bleeding secondary
exchange transfused. Care should be taken to functionally abnormal platelets.
to not increase the hemoglobin above 12 g/dl Transfusion is not indicated for those with
in patients with sickle cell d
­ isease second­ rapid destruction (e.g., immune thrombo­
ary to hyperviscosity and increased risk cytopenic purpura and neonatal alloim­
of central nervous system (CNS) events. mune thrombocytopenia) unless there is
Similarly, in patients with leukemia and life‐threatening hemorrhage; however,
hyperleukocytosis at diagnosis (i.e., white transfusion may be useful in the bleeding
blood cell count >100 × 109/l), the hemo­ patient with rapid consumption (e.g., dis­
globin should preferably be kept below seminated intravascular coagulation [DIC])
10 g/dl and transfusion avoided if possible or dilutional thrombocytopenia (massive
due to the same risks of increasing viscosity transfusion or exchange). Platelets are fre­
and the propensity for leukostasis. quently needed in the patient receiving
68 Chapter 5

chemotherapy or are thrombocytopenic e. Bleeding patient with normal coagu­


secondary to a marrow infiltrative process. lation studies, platelets <50 × 109/l; with
abnormal coagulation studies, platelet
Indications for platelet count <100 × 109/l.
transfusion f. Patient requiring a moderately inva­
1. Premature or sick infants sive surgical procedure, with a platelet
a. Stable infant with platelet count count <50–100 × 109/l (requiring discus­
<50 × 109/l. sion with surgical team secondary to lack
b. Distressed infant (or requiring surgical of evidence‐based guidelines). Most sur­
procedure) with platelet count <100 × 109/l. geons would like platelet count to remain
2. Children >50–75 × 109/l for 48–72 hours after the
a. Platelet count <10 × 109/l; higher if surgical procedure (this is extended to
febrile, septic, or with active bleeding. during chemotherapy after incomplete
b. Platelet count <20–50 × 109/l with a resection of brain tumors due to contin­
minor invasive procedure such as: ued risk of bleeding).
i. Lumbar puncture. g. Intramuscular injection (i.e., Erwinia
ii. Extracorporeal membrane oxy­ asparaginase) with platelet count
genation (ECMO) or cardiovascular <20 × 109/l.
(CV) bypass. h. Rapid consumption associated with
iii. Other minor procedures such as sepsis, DIC.
central line placement.
c. Invasive procedure in a patient with a Dosing of platelet transfusion
qualitative platelet defect. One unit of random platelets (~40 ml)
d. More invasive procedures will require per 10 kg will increase the platelet count
discussion with the surgical team by 40–50 × 109/l if there is no active con­
regarding platelet transfusion threshold, sumptive process (e.g., fever, immune
although recommendations are generally thrombocytopenic purpura, sepsis, alloim­
not evidence‐based. munization, or DIC) or sequestration. This
3. Patients undergoing therapy for is equivalent to an increment in platelet
malignancy count of 10 × 109/l/ml/kg of transfused
a. Platelet count <10 × 109/l; higher if platelets (i.e., in a 10‐kg child, 10 ml/kg or
febrile, septic, or with active bleeding. 100 ml of transfused plates should raise the
b. Induction chemotherapy: platelet count by 100 × 109/l). The platelet
i. Acute lymphoblastic leukemia count should be checked 1–2 hours after
(ALL) <10 × 109/l. infusion to identify refractory patients.
ii. Acute myeloid leukemia (AML) A patient is refractory if 1 hour after trans­
<10–20 × 109/l. fusion the platelet increment is less than
c. Children undergoing intensive ther­ 5–10 × 109/l per unit transfused for two sep­
apy with active mucositis should have arate transfusions.
their platelet count maintained at A patient may have platelet refractori­
20–50 × 109/l (i.e., Head Start protocol ness secondary to alloantibodies (immune‐
for brain tumors, postallogeneic myeloa­ mediated). Nonimmune causes of platelet
blative transplantation). refractoriness are common and include
d. Lumbar puncture with platelet count splenomegaly, fever, infection, DIC, veno‐
<20–50 × 109/l; potentially <50–100 × 109/l occlusive disease posttransplant, and use of
with diagnostic lumbar puncture. amphotericin B.
Transfusion Medicine 69

For refractory patients, a trial of cross­ treatment of stable clotting factor deficiencies
matched platelets should be given. Other pos­ in which no concentrate is available (i.e., not
sibilities for treatment should this fail include for factor VIII or IX). By definition, each mil­
leukocyte‐depleted, human leukocyte antigen liliter of undiluted plasma contains 1 interna­
(HLA)‐matched platelets, intravenous immu­ tional unit (IU) of each coagulation factor.
noglobulin (IVIG) with HLA‐matched plate­ Plasma consists of the anticoagulated
lets, or massive transfusion with random clear portion of blood separated by centrifu­
donor platelets (to overwhelm the antibody). gation. FFP is collected from single donors,
Most institutions now utilize pheresed with each unit being removed from a unit of
platelets that are harvested from a single whole blood and frozen within 6 to 8 hours of
donor and generally contain greater than collection. FFP should not be used when the
30 × 109/l of platelets, equivalent to approxi­ coagulopathy can be corrected more effec­
mately six to eight units of random platelets. tively with specific treatment such as vitamin
The volume is usually 250–350 ml. Secondary K, cryoprecipitate, or factor concentrate.
to the apheresis, these platelet products are Due to the high concentration of plasma
considered to be leukoreduced. In addition, antibodies, FFP is often the cause in cases of
although controversial, most believe that transfusion‐related acute lung injury (TRALI)
apheresis is sufficient to reduce the risk of and therefore should be used judiciously. The
CMV transmission. Plasma ABO compati­ direct antiglobulin test (DAT; Coombs test)
bility should generally be utilized, especially may also be positive for this reason.
with increasing volume of transfusion. Type‐
specific platelets can be given in cases of Indications for FFP
refractoriness and should be given to patients ●● Bleeding or invasive procedure with docu­

in the peri‐transplant period to eliminate the mented clotting factor deficiency and
production of any potential platelet antibod­ appropriate factor not available.
ies. As with PRBC transfusion, platelet irra­ ●● Treatment of protein C or S deficiency,

diation is required in oncology patients, factor XI deficiency (hemophilia C).


premature or low‐birth‐weight newborns, ●● Bleeding during massive transfusion, not

and patients with immunodeficiencies. from thrombocytopenia.


Dosage of platelet transfusion is gener­
ally 10–15 ml/kg and can be given over Dosing of FFP
30 minutes to 1 hour. Due to the expected The usual dosage of FFP is 10–15 ml/kg over
increase in platelet count and short life span 1 hour, which is expected to increase the con­
of platelets, one unit of pheresed platelets is centration of coagulation factors by 25–50%.
usually sufficient for transfusion in patients Due to the presence of isohemagglutinins,
over 30 kg. As with PRBCs, pheresed platelet FFP should be ABO‐matched. White blood
units should be rounded to the nearest half cells are killed or made nonfunctional during
and full unit as feasible to decrease waste the freezing process; therefore, leukoreduc­
and sterilely aliquot as needed. tion and irradiation are unnecessary.

Fresh frozen plasma Cryoprecipitate

Fresh frozen plasma (FFP) is a source of Cryoprecipitate is prepared by thawing FFP


plasma proteins, including nonlabile clotting and recovering the cold precipitate. Each
factors, such as fibrinogen. It is used for the bag contains >80 U factor VIII coagulant
70 Chapter 5

activity and >150 mg fibrinogen in approxi­


Granulocyte transfusion
mately 15 ml of plasma. In addition, cryo­
precipitate contains factor XIII and von
Patients with profound and prolonged neu­
Willebrand factor. Bags must usually be
tropenia are at increased risk for serious life‐
pooled to achieve an adequate dose.
threatening fungal and bacterial infections.
The beneficial effect of granulocyte transfu­
Indications for cryoprecipitate
sion in this population with known persis­
●● Bleeding or invasive procedure with factor
tent infection, especially with Gram‐negative
VIII deficiency or von Willebrand disease
organisms and fungus, is yet to be proven
where specific factor concentrate is not
by randomized controlled trials although
available.
observational studies are available. Donor
●● Bleeding or invasive procedure with hypofi­
mobilization with granulocyte colony‐stim­
brinogenemia or factor XIII deficiency.
ulating factor (G‐CSF) has increased granu­
locyte yields and therefore the therapeutic
Dosing of cryoprecipitate
benefit of transfusion, as the dose of granu­
The usual dosage for cryoprecipitate is
locytes is the most important factor in suc­
1 unit/5 kg. For hypofibrinogenemia with
cess of this treatment modality.
coagulopathy, the goal is to maintain fibrin­
Granulocytes migrate toward, phagocyt­
ogen >100 mg/dl. Specific factor or coagula­
ize, and kill bacteria. When given a granulo­
tion protein levels need to be determined in
cyte transfusion, the cells migrate to the foci
addition to assessing clinical status to
of infection, though there is rarely a measur­
decide on frequency of transfusion. As with
able increase in the peripheral granulocyte
FFP, cryoprecipitate is preferably ABO com­
count. This is likely from sequestration at
patible and leukoreduction and irradiation
the site of infection, prior immunization to
are not required.
leukocyte antigens, or a consumptive pro­
cess secondary to the infection. Side effects
of granulocyte transfusion include the risk
Antithrombin III
of CMV infection, TA‐GVHD, respiratory
distress with pulmonary infiltrates (due to
Antithrombin III (ATIII) concentrate is
TRALI, concurrent administration of
available for use in patients with inherited
amphotericin B, or secondary to granulo­
or acquired ATIII deficiency (i.e., sepsis,
cyte sequestration), and alloimmunization.
throm­bosis, and medication‐induced). It
may also be needed in patients receiving
heparin therapy who have a low ATIII Preparation of granulocyte
level. Patients with thrombosis after aspar­ concentrates
aginase therapy should have an ATIII level Donor mobilization is recommended with
checked and repleted as necessary. Dosing G‐CSF at 5 mcg/kg (maximum 300 mcg
of ATIII is based on the baseline and IV/SC) and dexamethasone 8 mg orally,
desired level using the following calcula­ 12–24 hours prior to collection. Granulocytes
tion: units desired (IU) = (desired ATIII% are then collected by apheresis and ideally
level − baseline ATIII%) × body weight should be transfused within 8–12 hours of
(kg). The target level is 80–120%. Vials are collection. RBC contamination requires
typically 500 IU and the dose can be ABO compatibility. Due to risks of TA‐
adjusted ±10% (i.e., rounded to a full vial GVHD and CMV infection, irradiation
to avoid wastage). and CMV seronegativity are required (in
Transfusion Medicine 71

those that are CMV‐negative), respectively. check of all labels, forms, and patient identifi­
Amphotericin B and concomitant granulo­ cation should be done, in addition to notify­
cyte transfusion is potentially associated with ing the blood bank. Transfusion reactions are
severe pulmonary reactions, although the summarized in the following text and include
evidence is controversial; however, it is rea­ FNHTRs, allergic reactions, immune‐medi­
sonable to space these therapies apart by at ated hemolysis, TRALI, transfusion‐associated
least 4 hours. HLA‐matched granulocytes circulatory overload (TACO), TA‐GVHD,
should be given in patients with known allo­ and acute infection.
immunization. Since granulocyte half‐life is
only 7 hours, daily collection and transfusion Hemolytic transfusion reactions
for several days are likely required for benefit. Hemolytic transfusion reactions can either
be acute or delayed. Acute hemolytic trans­
Indications for granulocyte fusion reactions (AHTRs) are usually due to
transfusion clerical error resulting in the transfusion of
With a lack of randomized controlled trials, an ABO incompatible unit. AHTRs classi­
there are no evidence‐based guidelines for cally present with fever, chills, nausea, and
granulocyte transfusion. Limited data in vomiting in addition to dyspnea, hypoten­
neonates with sepsis have failed to show a sion, shock, hemoglobinuria, and DIC.
significant benefit. After weighing the Bacterial contamination must be considered
potential risks and benefits, as well as deter­ in the differential for an AHTR. Delayed
mining the daily availability of an eligible hemolytic transfusion reactions (DHTRs)
donor and collection site, granulocyte trans­ present 2–14 days after transfusion with
fusion can be considered in severely neu­ milder symptoms including low‐grade fever,
tropenic patients with a refractory or jaundice, and a posttransfusion hemoglobin
progressive bacterial or fungal infection on increment less than expected due to minor
appropriate, aggressive therapy with neutro­ antigen incompatibility (alloimmunization)
penia that is expected to continue for, at the from prior transfusion.
least, several days. Evaluation of a potential AHTR should
include workup of all recently transfused
blood products (see Figure 5.2). A bedside
Transfusion reactions check of labeling should occur followed by
laboratory evaluation including repeat cross­
Approximately 4% of transfusions are asso­ matching and a DAT. The DAT may not
ciated with some form of adverse reaction, always be positive if all the antibodies have
ranging from brief episodes of fever to life‐ been destroyed during the hemolytic crisis.
threatening episodes of hemolysis and Other labs that should be sent to rule out
shock. Fortunately, the majority of reactions intravascular hemolysis include indirect bili­
are short‐term, specifically FNHTRs and rubin, lactate dehydrogenase (LDH), plasma
allergic reactions, and easily managed. Life‐ free hemoglobin (and/or haptoglobin), and
threatening reactions are nearly always due urinalysis to check for hemoglobinuria.
to clerical error resulting in transfusion of DHTRs lead to extravascular hemolysis
an ABO incompatible unit. The challenge and should be evaluated in a patient with
for the clinician is to promptly recognize clinical or laboratory symptoms after the first
potential serious complications that may 24 hours. Labs to follow include a posttrans­
present with common symptoms such as fusion hemoglobin level and reticulocyte
fever. For any transfusion reaction, a ­bedside count, indirect bilirubin, LDH, and DAT.
72 Chapter 5

Fever (≥38.0°C or 100.4°F)


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 ≥1–2°C ↑ in temperature


Initiate supportive care (IVF, compared to past 24 hours?*
antibiotics)
Contact Intensive Care
Yes No

Stop transfusion Continue 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 bilirubin, LDH, UA, DAT, plasma free hgb
Consider CXR if respiratory symptoms or new hypoxia

Positive workup?

Yes No

Provide appropriate care based on Restart transfusion


likely diagnosis and clinical symptoms Monitor clinical exam closely
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).
*see text for more detail

If an AHTR is suspected, emergent man­ FNHTRs were significantly more com­


agement is vital. The transfusion should be mon prior to near universal leukoreduction
immediately stopped, and IV fluid resuscita­ and occur due to pyrogenic cytokines
tion commenced. Inotropic support may be released by leukocytes during blood compo­
required for hypotension and shock. Renal nent storage. FNHTR is defined as a tem­
perfusion and urine output should be fol­ perature increase of 1 °C (1.8 °F) without any
lowed closely, and additional blood product other attributable cause during or within
support may be required. DHTRs usually 4 hours of transfusion. More serious causes
do not require intervention, although may of fever including AHTR and bacterial con­
rarely cause profound anemia. tamination must be ruled out prior to the
Transfusion Medicine 73

diagnosis of FNHTR being made. Trans­ Patients with multiple episodes may benefit
fusion must therefore be discontinued until from premedication with an antihistamine
an AHTR can be ruled out (see Figure 5.2). and, if not effective, potentially corticoster­
Fever due to FNHTR is usually self‐limited oids. If ­allergic reactions continue, washed
and resolves with antipyretic usage and blood products should be utilized.
stopping the transfusion. Chills, rigors, and
discomfort can occur, mimicking an AHTR. Transfusion‐related acute lung
Transfusion can potentially be restarted injury
after AHTR has been ruled out based on the TRALI is becoming increasingly recognized
clinical status of the patient and urgency of as an important cause of morbidity after
the transfusion. Although patients with a transfusion with plasma‐containing blood
history of FNHTR often receive premedica­ components. Signs and symptoms include
tion with acetaminophen with future trans­ hypoxia, chest infiltrates (without volume
fusions, this practice is not evidence‐based. overload), dyspnea/tachypnea, fever, and
Antipyretics should be c­onsidered for the hypotension. TRALI generally occurs within
patient with multiple FNHTRs; if acetami­ 6 hours of transfusion completion. A major­
nophen is not effective, a trial of washed ity of patients with TRALI will recover in a
blood components can be considered. Of 2‐ to 4‐day period although respiratory
note, transfusion can be given to the febrile support, and in some cases mechanical ven­
patient if urgently necessary, as the criteria tilation, is often required. Pressor support
for workup (beyond a close and continued may be necessary, and treatment with cor­
clinical assessment) is an increase of 1–2 °C ticosteroids may be helpful.
in fever (dependent on institutional stand­
ard) compared to the fever curve from the Transfusion‐associated
previous 24 hours. circulatory overload
TACO also is becoming more widely recog­
Allergic transfusion reactions nized, but is likely rare in the pediatric popu­
Allergic reactions are typically type I hyper­ lation. TACO occurs due to cardiogenic
sensitivity reactions to plasma in blood edema from too rapid or too large a volume
components. Allergic reactions are common, of transfusion. Clinical signs such as dysp­
occurring in 1–5% of transfusions, and nea/tachypnea and hypoxia may be confused
usually with mild symptoms such as urti­ with TRALI. Differentiating features include
caria, although anaphylaxis can occur. For hypertension rather than hypotension due to
the majority of allergic reactions, an antihis­ pulmonary over‐circulation with a positive
tamine such as diphenhydramine is sufficient fluid balance. Chest radiograph should be
to alleviate symptoms and the transfusion more consistent with pulmonary edema/
can likely be completed in the allotted time. effusion rather than infiltrates. Aggressive
For more severe symptoms, the transfusion diuresis should be utilized.
should be stopped. Steroids, an H2 blocker
(e.g., famotidine), epinephrine, volume Transfusion‐associated graft‐
expansion, β2 agonists (e.g., albuterol), and versus‐host disease
oxygen may be required. Patients with a TA‐GVHD can occur in immunocompro­
severe allergic reaction should be tested for mised patients who receive nonirradiated
IgA deficiency. As with FNHTRs, premedica­ blood or platelet transfusion due to donor
tion (with an antihistamine) remains contro­ T‐lymphocytes that cannot be rejected by
versial in patients with a history of a reaction. the host. TA‐GVHD has also been reported
74 Chapter 5

in immunologically normal patients with an care unit (PICU) with septic shock. He is
HLA‐compatible donor such as in homoge­ secondarily found to be anemic and throm­
neous populations or in cases where the bocytopenic with coagulopathy and hypofi­
donor is a close relative (directed donation). brinogenemia in DIC.
Clinical symptoms are equivalent to those After initial stabilization of the patient
with transplant GVHD including fever, ano­ with intravenous fluids, antibiotics, mech­
rexia, vomiting/diarrhea, and skin rash. anical ventilation, and vaso pressors, the
Hepatic dysfunction and pancytopenia can PICU team is determining their transfu­
similarly manifest. sion plan. Current hemoglobin is 7.0 g/dl,
platelets are 34 × 109/l, PT (prothrombin
Bacterial infections time) is 27.2 seconds, PTT (partial throm­
Blood components may be contaminated boplastin time) is 74.6 seconds, and
with bacteria at the time of collection or fibrinogen is 76 mg/dl. There is no noted
during processing. Infection rates are higher active bleeding. 1:1 mixing studies correct
after platelet transfusion, since platelets are both the PT and PTT, implying a factor
stored at room temperature, although clini­ deficiency rather than an antibody. The
cal signs and symptoms are often more patient does appear to have some amount of
severe after transfusion of contaminated hemolysis with an elevated indirect biliru­
PRBCs. Due to the increased risk of infec­ bin and LDH, but has a negative direct
tion with platelets, we utilize a temperature Coombs test.
increase of 1 °C (39 °C in the previously afe­ a. What blood products should be
brile patient) over the past 24 hours versus transfused?
increase of 2 °C (40 °C in the previously afe­ b. What amount of PRBCs should be
brile patient) for PRBCs in the stable‐ given?
appearing patient prior to commencing an c. Are there any special considerations
infection workup (i.e., blood culture of the for the type of blood product required?
patient and bag, ceftriaxone, AHTR workup d. What are some specific risks with this
for PRBCs). Infection is associated with a patient presentation?
rapid onset of symptoms and a high rate of First, the patient should be given PRBCs,
morbidity and mortality, especially from pheresed platelets (if possible), and FFP.
Gram‐negative organisms. The patient may Cryoprecipitate may or may not be required
present acutely septic with fever/chills, dependent on the increase in fibrinogen
hypotension, tachycardia, and shock. If bac­ with FFP. If the patient is clinically worsen­
terial contamination is suspected, the trans­ ing (e.g., problems oxygenating, dropping
fusion should be stopped immediately. blood pressures), consideration should be
Aggressive supportive treatment including given to transfusing O-negative PRBCs
IV fluid resuscitation, initiation of broad‐ while awaiting crossmatching.
spectrum antibiotics, and potential therapy Second, one can utilize the transfusion
for renal failure, shock, and/or DIC should factor to estimate the increment in hemo­
be started emergently. globin with transfusion. In this case, the
transfusion factor will likely overestimate
the bump as the patient is actively septic and
Case studies for review therefore hemolyzing PRBCs and consum­
ing platelets. Still, estimating will help deter­
1. You are following a 12-year-old, 60-kg mine a reasonable plan. Assuming the
boy admitted to the pediatric intensive decision is made to give 15 ml/kg (3 units),
Transfusion Medicine 75

one would expect at most an increase in ­transfusion-associated microchimerism.


hemoglobin of 3 g/dl: Microchimerism is the presence of donor
lymphocytes in the recipient’s circulation.
Transfusion amount (ml/kg)/transfusion The significance of transfusion-associated
factor = hgb increase (g/dl) microchimerism is unknown. Patients
Therefore, in this case (15 ml/kg)/5 = 3 g/dl. receiving large amounts of plasma are at risk
for developing TRALI that must be consid­
In general, platelet and FFP transfusion will ered with worsening respiratory symptoms.
also be 10–15 ml/kg. Platelet transfusion
should initially be a maximum of one 2. You are following a 10-year-old, 40-kg
­pheresed unit (approximately 250–350 ml). female on the oncology ward that was
Posttransfusion hemoglobin, platelets, and recently diagnosed with leukemia. She is
coagulation studies can help determine the due for a lumbar puncture with chemother­
need for additional transfusion as well as the apy under anesthesia. She has had persistent
likely frequency of necessary transfusion fevers to 39°C, has a central line, but has had
(directly dependent on the clinical status of negative blood cultures to date. The assump­
the patient). Time for “equilibration” after the tion is that her fevers are secondary to her
completion of transfusion is unnecessary. underlying leukemia. Her hemoglobin is
Third, in this case, no significant special 6.2 g/dl and her platelets are 8 × 109/l. She
arrangements need to initially be made in has not had any active bleeding and she is
regard to the transfusions. The patient does not symptomatic secondary to her anemia.
not need sickledex-negative blood unless Her CMV status is pending.
they have concomitant sickle cell disease. a. What would you recommend in
Also, phenotypically matched blood is regard to transfusion?
unnecessary. As the patient does not have a Although she is asymptomatic, strict trans­
known underlying reason to be immuno­ fusion thresholds should be followed for
compromised, irradiated blood and platelets oncology patients to minimize symptoms,
are not required. Similarly, CMV-negative specifically maintenance of the hemoglobin
blood is unnecessary. ≥7 g/dl and platelets ≥10 × 109/l. Additionally,
Finally, specific risks exist for this patient most anesthetic procedures require hemo­
and must be considered during transfusion. globin around 7 g/dl. Thresholds for lumbar
Due to the likelihood of continued transfu­ punctures vary, though a minimum of
sions as well as the presence of sepsis, the 20–30 × 109/l should be utilized for all but
patient may develop volume overload and the first therapeutic lumbar puncture where
potentially TACO; therefore, the judicious guidelines generally agree upon maintaining
use of diuretics with transfusion should a higher threshold (i.e., ≥75–100 × 109/l).
be considered. In addition, a significant In regard to PRBC transfusion, she should
volume of PRBC transfusion can have a
­ receive 10–15 ml/kg while minimizing allo­
dilutional effect on platelets and coagulation immunization risk by transfusing multiple
factors, thus increasing transfusion require­ units from one donor. In this situation,
ments for these blood products. The patient 15 ml/kg is approximately 2 units of PRBCs
is also at risk for hyperkalemia with and therefore would be the ideal choice. The
increasing PRBC transfusion. If hyper­ PRBCs should be irradiated, since she has
kalemia begins to occur, washed PRBCs started chemotherapy and is immunosup­
should be given. Massive PRBC or whole pressed to eliminate donor T-cells and risk
blood transfusion has been shown to induce of transfusion-associated graft-versus-host
76 Chapter 5

disease. Since her CMV status is pending, highest temperature in the last 24 hours. In
consideration can be given for CMV- the case of platelet transfusion, a more strin­
seronegative PRBCs, based on institutional gent threshold is utilized to rule out a septic
standards. Platelets should also be irradiated, unit secondary to platelets being kept agi­
though generally do not need to be CMV- tated at room temperature. Our institutional
seronegative (again based on institutional standard is to work up a platelet unit with
standard) due to minimization of WBCs any temperature that is 1 °C greater than the
through the platelet pheresis collection pro­ highest temperature in the last 24 hours or
cedure. Platelets are also given at 10–15 ml/ with a new fever in the afebrile patient.
kg, though are generally maximized to 1 unit Patients again are most likely to be having a
due to their short life (i.e., 3–4 days for trans­ febrile nonhemolytic reaction or allergic
fused platelets), unless the clinical scenario reaction rather than sepsis, but the worst-
dictates the need for additional platelets (i.e., case scenario must be considered and ruled
ongoing bleeding, significant platelet out. In the case of a platelet transfusion, the
consumption). patient must first be evaluated and stabilized
b. Can the patient be transfused even as needed. The platelet transfusion should
though she is febrile? be stopped and the patient cultured and
This is a common question that occurs on started on antibiotics (if not already on anti­
the oncology unit. Although in the ideal sce­ biotics). The platelet bag should be returned
nario the patient would be afebrile prior to to the blood bank where it should be worked
transfusion, this is often not the case. up for a transfusion reaction, which in this
Acetaminophen can be trialed prior to situation would include a culture of the
transfusion if there is time to see if this platelet bag. An acute hemolytic reaction
allows the patient to defervesce. The febrile with pheresed platelets is highly unlikely
patient should be evaluated prior to transfu­ given the small amount of RBC contamina­
sion, and if stable should be okayed for tion, though workup for hemolysis is gener­
transfusion. ally part of the transfusion-reaction workup,
c. The nurse calls that the patient is including a DAT (Coombs), retyping of the
spiking a fever to 40.1°C during the bag and patient, CBC/retic, LDH, and
PRBC transfusion. What are the next urinalysis.
steps? What if the patient spiked a
40.1°C fever during the platelet
transfusion? Multiple choice questions
The patient should first be clinically evalu­
ated to ensure she is stable. Although an 1. You are on the oncology service following
acute hemolytic reaction due to ABO mis­ a patient with acute myelogenous leukemia.
match is highly improbable, it must be first The patient is CMV seropositive. The patient
ruled out in any PRBC transfusion. The received myelosuppresive chemotherapy
most likely diagnosis is a febrile nonhemo­ 10 days prior and now has a hgb of 6.1 g/dl.
lytic transfusion reaction, though an allergic Why should the patient receive irradiated
reaction and sepsis from a tainted unit PRBCs?
should also be considered and may initially a. Prevention of acute hemolytic trans­
present with fever. In the stable patient fusion reaction
­without signs of allergy or sepsis, the PRBC b. Prevention of allergic reaction
transfusion can be continued, since the tem­ c. Prevention of febrile non‐hemolytic
perature is less than 2°C greater than the transfusion reaction
Transfusion Medicine 77

d. Prevention of transfusion‐associated breakdown due to antibody formation to


graft‐versus‐host disease minor antigens in the chronically transfused
e. Prevention of transfusion‐associated patient. The estimated rise in hgb can be cal­
acute lung injury (TRALI) culated by determining the amount of blood
Explanation: Passive transfusion of donor transfused per kg (in this case 650 ml
T‐cells in PRBCs can lead to a T‐cell donor divided by 45 kg or 14.4 ml/kg) and then
vs. recipient response in the severely immu­ dividing that amount by 5. Therefore the
nocompromised host leading to aplasia. expected rise in hgb in this case is 14.4
Irradiation kills these donor T‐cells and thus divided by 5 or 2.9. The answer is d.
prevents TA‐GVHD. Acute hemolytic trans­
fusion reactions occur due to clerical error 3. You are following a 12-year-old male
and ABO mismatch between donor and inpatient with sickle cell anemia with a hgb of
recipient. Prevention of allergic reaction can 5.3 g/dl (baseline hgb 7‐8 g/dl) and reticulo­
occur through the use of washed products. cytopenia due to presumed viral suppression.
Prevention of febrile non‐hemolytic trans­ Studies for parvoviral infection are pending.
fusion reactions occurs through the use of Based on the hgb and reticulocytopenia
leukofiltration which eliminates the major­ you decide correctly to transfuse the patient.
ity of WBCs and is standard of practice at Which of the below are the correct parame­
most blood banks in the United States. ters for PRBC transfusion in this patient?
TRALI can occur due to plasma compo­ a. Phenotypically matched
nents, especially from fresh frozen plasma b. Phenotypically matched, sickledex
(FFP) but also possibly platelets and PRBCs negative
and can be prevented through judicious use c. Phenotypically matched, sickledex
of blood products. The answer is d. negative, irradiated
d. Phenotypically matched, sickledex
2. You are on the oncology service follow­ negative, CMV negative
ing a patient with acute myelogenous leuke­ e. There are no requirements for this
mia. The patient is CMV seropositive. The urgently required PRBC unit
patient received myelosuppresive chemo­ Explanation: Patients that are chronically
therapy 10 days prior and now has a hgb of transfused (i.e., sickle cell anemia, thalas­
6.1 g/dl. The patient weighs 45 kg and you semia) should receive phenotypically
give 2 units of irradiated PRBCs. The total matched PRBCs to prevent alloimmuniza­
volume of the 2 units is 650 ml. Assuming tion and refractoriness over time. As people
no ongoing blood loss or hemolytic reaction with sickle cell trait are not precluded from
due to transfusion, what is the presumed donating blood, it is important to ensure
post‐transfusion hemoglobin? that any PRBC transfusion does not contain
a. 6.8 g/dl sickle cells (i.e., sickledex negative).
b. 7.5 g/dl Irradiated blood is limited to immunocom­
c. 8.1 g/dl promised patients, as is CMV seronegative
d. 9.0 g/dl blood for patients that may require hemat­
e. 9.8 g/dl opoietic stem cell transplant and are immu­
Explanation: For PRBCs preserved in Adsol, nocompromised and CMV seronegative. In
the transfusion factor is estimated to be 5 an emergent situation if no phenotypically
and can be used to calculate the estimated matched blood is available, e. could be con­
rise in hemoglobin assuming no ongoing sidered but this patient is relatively stable.
losses from blood loss or loss due to red cell The answer is b.
78 Chapter 5

4. You are following a 10-year-old female marrow aspiration and lumbar puncture in the
patient with aplastic anemia admitted to the morning. You have been instructed to trans­
hospital for routine transfusions for anemia fuse platelets to reach a goal platelet count of
and thrombocytopenia. She has a history of 30 × 109/l and to maintain a hgb ≥7.0 g/dl for
febrile non‐hemolytic and allergic reactions anesthesia. The patient is 24 kg. Evening labs
to both blood products so she receives pre‐ result with a hgb of 6.0 and platelets of 14 ×
medications for both PRBCs and platelets. 109/l. The patient is CMV seropositive. Which
About one hour into the PRBC transfusion is of the below is the best answer:
she spikes a fever to 102°F. You are called to a. Give 1/2 unit platelets and PRBCs
assess the patient and note that she is newly b. Give 1/2 unit irradiated platelets and
hypotensive. All of the following are appro­ 1/2 unit irradiated PRBCs
priate steps EXCEPT: c. Give 1 unit platelets and PRBCs
a. Order a transfusion reaction work up d. Give 1 unit irradiated platelets and
including blood culture, urine, Coombs, 1 unit PRBCs
bilirubin e. Give 1 unit irradiated platelets, 1 unit
b. Start empiric ceftriaxone irradiated PRBCs
c. Give an IVF bolus Explanation: One unit of pheresed platelets
d. Ask for a culture of the PRBC bag and and one unit of PRBCs is generally 250–350
clerical check ml and the practitioner should aim to
e. Continue with the PRBC transfusion transfuse in the range of half‐units so that
Explanation: Acute transfusion reactions the blood bank can sterilely aliquot the
can include febrile non‐hemolytic transfu­ remainder. Transfusion should be given
sion reactions (FNHTR), acute hemolytic with the goal of approximately 10‐15 ml/kg,
reaction, allergic reaction and acute infec­ with a maximum transfusion of 1 unit plate­
tion. A new fever or a fever 1‐2°C higher lets and generally 2 units PRBCs. Although
than the previous highest temperature in the the patient has not yet received chemother­
last 24 hours should prompt a transfusion apy, they are likely still immunosuppressed
reaction work up. ABO incompatibility is secondary to their underlying disease so it is
highly unlikely and would present within safest to irradiate the blood products to pre­
the first few minutes of transfusion with vent any potential risk of TA‐GVHD. The
hemolysis but should be considered even answer is e.
beyond this point if with typical symptoms.
Fever is most likely secondary to a FNHTR
but allergic reaction and infection should be Suggested reading
ruled out especially with a high fever.
Although PRBC transfusion may be able to Delaney, M., Wendel, S., Bercovitz, R.S. et al.
be resumed, this should not occur until the (2016). Transfusion reactions: prevention,
patient improves and the immediate work‐ diagnosis, and treatment. Lancet 388:
2825–2836.
up is negative. For the hypotensive patient,
Doctor, A., Cholette, J.M., Remy, K.E. et al. (2018).
given the potential concern for bacterial Recommendations on Red Blood Cell
contamination, the transfusion should not Transfusion in General Critically Ill Children
be resumed. The answer is e. Based on Hemoglobin and/or Physiologic
Thresholds from the Pediatric Critical Care
5. You are following a new diagnosis patient Transfusion and Anemia Expertise Initiative.
with ALL who is due for diagnostic bone Pediatr. Crit. Care Med. 19: S98–S113.
6 Chelation Therapy

proton magnetic resonance imaging (R2


Transfusional iron overload
MRI) is becoming widely utilized as an
accurate and noninvasive measure of organ
Iron overload may occur in any patient
iron content, and T2* MRI is becoming the
receiving intermittent transfusions for acute
standard for measuring cardiac iron stores.
illness (e.g., sickle cell disease), chronic
Institution of chelation therapy relative
transfusion therapy (e.g., thalassemia, sickle
to transfusion frequency is based on institu-
cell disease, red cell aplasia, bone marrow
tional practice. Patients that will require
failure syndromes), or in those having
chronic transfusion therapy should be che-
received an intensive period of frequent
lated before their liver iron reaches a high
transfusion (e.g., myelosuppressive chemo-
level, measured by liver biopsy, MRI quanti-
therapy/radiation for treatment of malig-
fication, or based on the number of transfu-
nancy and after hematopoietic stem cell
sions received (i.e., chelation initiated after
transplantation). Each milliliter of blood
receiving 10–20 transfusions, or approxi-
contains 1 mg of iron and normal iron stores
mately 3–6 g of transfusional iron in a 30‐kg
are approximately 3 g, with 2 g in the blood
patient). Thalassemia patients tend to have a
and 1 g in the liver. Therefore, patients
higher iron burden than sickle cell patients,
receiving frequent transfusion likely will
as the chronic inflammatory state in sickle
have received a large amount of transfused
cell disease limits gastrointestinal iron
iron and should be monitored for evidence
absorption due to upregulation of hepcidin.
of iron accumulation.
Thalassemia patients should be counseled to
There is no simple test for quantifying
be on a low‐iron diet.
total iron burden. Serial serum ferritin levels
Patients receiving intermittent transfu-
are helpful in determining hepatic iron
sion or those receiving frequent transfusion
stores, but values are altered in states of
therapy in a short period of time (e.g., with
inflammation. Liver biopsy remains the gold
intensive leukemia therapies) may have a
standard for quantification of total body
more insidious development of iron over-
iron, but is an invasive procedure. Liver iron
load. Patients with leukemia should have
levels ≥7 mg/g dry weight liver are indicative
serum ferritin levels checked at the end of
of iron overload. The superconducting
therapy. Those with ferritin levels >1000 ng/
quantum interference device (SQUID) is
ml should have serial checks every 3 months;
accepted as a noninvasive method to quanti-
if ferritin remains above this threshold after
tate total body iron, but its limited availabil-
6–12 months, the patient should undergo
ity does not allow for routine use. Currently,

Handbook of Pediatric Hematology and Oncology – Children’s Hospital & Research Center Oakland,
Third Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
© 2021 John Wiley & Sons Ltd. Published 2021 by John Wiley & Sons Ltd.
80 Chapter 6

liver biopsy or R2 MRI for more accurate presumed increased compliance. Exjade and
iron quantification. These patients, espe- Jadenu may not be effective in all patients,
cially male adolescents, may benefit from which must be weighed against the risks of
monthly phlebotomy which can be discon- noncompliance with Desferal. Deferiprone
tinued when the ferritin and liver iron (Ferriprox or Kelfer) is also available. Usage
concentration reach normal levels (i.e.,
­ of two agents concomitantly is currently
<3 mg/g dry liver weight). Younger children undergoing clinical trials and may have par-
and menstruating females have been shown ticular benefit in chelation of cardiac iron.
to have reversible iron overload in most Concomitant administration of ascorbic
such cases. acid (vitamin C) increases the excretion of
Current treatment strategies for iron iron when given with chelation. Iron chela-
overload and potential side effects of iron tors should not be administered when there is
chelators are summarized in Table 6.1. concern for a bacterial infection. By mobiliz-
Desferoxamine (Desferal) has the longest ing free iron, chelators promote bacterial
treatment record but requires parenteral growth, in particular Yersinia enterocolitica.
administration and has been widely replaced For febrile patients on chelator therapy, chela-
by deferasirox (Exjade/Jadenu) due to the tion should be stopped until blood c­ ultures
convenience of oral intake and therefore are definitively negative. Broad‐spectrum

Table 6.1 Iron chelators.

Name Desferoxamine (Desferal) Deferasirox (Exjade/ Deferiprone


Jadenu) (Ferriprox/Kelfer/L1)

Dose (mg/kg/d) 25–50 20–40 Exjade 75–100


14–28 Jadenu
Administration SC/IV, given as continuous PO (oral), daily PO, TID
infusion over 8–24 h,
5–7 d/wk
Side effects Irritation at infusion site, GI disturbance, rash, Agranulocytosis,
ototoxicity (tinnitus, renal and hepatic neutropenia, GI
transient hearing loss), impairment, GI disturbance,
ocular disturbance hemorrhage transaminitis,
(decreased night vision), arthropathy,
allergic reactions, progression of
growth failure, skeletal hepatic fibrosis
disturbance, pulmonary
hypersensitivity
Potential Highly effective but Long‐term data Long‐term data
therapeutic compliance may be an lacking; may not be lacking
issues issue due to route of effective in all
administration patients; has not
been studied in
combination with
other agents

Abbreviations: SC, subcutaneous; IV, intravenous; TID, three times a day; GI, gastrointestinal;
FDA, Food and Drug Administration.
Chelation Therapy 81

antibiotic prophylaxis should be initiated in particular are at risk due to normal develop-
the ill‐appearing or splenectomized patient mental behaviors such as putting objects
and include coverage for this unusual and toys in the mouth and chewing on unu-
organism. sual surfaces (e.g., windowsills).
Lead entering the intravascular space
rapidly attaches to the red blood cell, with
Lead toxicity minimal amounts (3%) detected in the
plasma. The half‐life in the blood is approxi-
Lead poisoning is an environmental disease mately 21–30 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 (65–90%). Lead can
­universal efforts to decrease environmental enter any cell and toxicity may occur in any
lead contamination, with a resultant decrease tissue or organ. Classically, in severe lead
in measured blood lead levels in children intoxication, gastrointestinal and central
over the past two to three decades. Sources of nervous system toxicities are the most clini-
lead have included gasoline additives, food‐ cally apparent. Gastrointestinal symptoms
can soldering, lead‐based paints, ceramic include anorexia, nausea, vomiting, abdom-
glazes, certain toys, drinking water systems inal pain, and constipation. The blood lead
(lead pipes), and folk remedies. The use of level is typically ≥50 mcg/dl when these
these products has been markedly reduced symptoms are present. Lead poisoning was a
as the result of federal guidelines and the lethal disease in the United States decades
development of cost‐effective alternatives. ago, primarily related to neurotoxic effects.
Lead in gasoline and paint is at extremely At levels ≥100 mcg/dl, children may show
low levels and has been eliminated altogether evidence of encephalopathy, including a
from food‐can soldering. Housing built marked change in mentation or activity,
prior to 1960 is likely to have been painted ataxia, seizures, and coma. Increased intrac-
with high‐content lead‐based paint, and ranial pressure may be present on examina-
since lead isotopes are very stable, environ- tion. These effects are usually permanent
mental exposure presents an ongoing risk. with long‐term sequelae of retardation, pal-
High‐risk populations have a greater likeli- sies, and growth failure.
hood of living in older housing, which has Most children who have elevated blood
not had lead abatement. Risk factors for lead levels have subclinical disease. Fewer
excessive lead exposure include poverty, age than 5% of children present with overt
younger than 6 years, African‐American symptoms of lead toxicity. An elevated
ethnicity, and urban housing. blood lead level, suggesting excessive envi-
Lead may enter the body through direct ronmental exposure, is defined as 10 mcg/
ingestion, inhalation, or via skin absorption. dl. Many studies have shown associations
The most common pathway among young between blood lead levels and impaired
children is through the mouth. Lead absorp- neurocognitive function. These results have
tion is enhanced in the presence of other provided the primary impetus for current
dietary mineral deficiencies, such as cal- public health efforts. Of note, no lead level is
cium and iron, due to competitive biochem- normal and even lead levels below 10 mcg/dl
ical pathways. Pica behavior enhances the are thought to lead to subtle neurocognitive
likelihood of direct ingestion. Toddlers in dysfunction.
82 Chapter 6

Screening for lead toxicity Pediatrics (AAP) recommend venous blood


Prevention and treatment of lead toxicity lead sampling for children identified to be at
remains a major public health concern, and high risk (living in housing built prior to
efforts for screening have primarily focused 1960, indigent, urban, and minority chil-
on high‐risk populations. All children dren) at 1 and 2 years of age. Venous blood
should have a screen of potential environ- samples should be used to assess blood lead
mental exposures by their primary health levels, as capillary samples may give falsely
care provider starting at 1 year of age and low results. If the screening test confirms an
repeated when the child is mobile and elevated blood lead level, specific manage-
attains hand‐to‐mouth behavior. The ment guidelines have been developed by the
Centers for Disease Control and Prevention CDC and AAP and are summarized in
(CDC) and the American Academy of Table 6.2. The vast majority of children with

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 mentioned 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, dimercaptosuccinic acid) 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 (also called British anti‐Lewisite, BAL) 25 mg/kg/d IM, divided q4
h 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: TID, three times a day; BID, twice a day; CNS, central nervous system; EDTA,
ethylenediaminetetraacetic acid; IV, intravenous; IM, intramuscular.
Chelation Therapy 83

elevated lead levels are not candidates for using the same criteria specified in
chelation therapy with currently available Table 6.2. Ongoing efforts should be made
drugs. Children with low levels (<20 mcg/ to provide the family with education in
dl) are asymptomatic and unlikely to have order to prevent exposure in the child’s
significant increase in lead excretion with environment in addition to assuring ade-
chelation. These children benefit primarily quate nutrition. Family members and
from decreased exposure. ­siblings should be screened as well.
Blood lead levels measure the blood con-
centration at a point in time and may not be
able to accurately predict bone stores. Bone Case study for review
lead content may be assessed noninvasively
using a radiographic technique called X‐ray 1. You are working at a primary pediatric
fluorescence. Measurement of the heme clinic in an urban setting and have instituted
precursor, free erythrocyte protoporphyrin
­ routine lead screening at 1 year of age.
(FEP), may also provide a useful clue about A routine screening result returns at
the duration of exposure and the degree of 17 mcg/dl for a 14-month-old male whom
lead accumulation. Excessively high FEP lev- you screened at his first visit with you.
els classically are seen with severe lead toxic- a. What are the initial steps?
ity. Other causes of elevated FEP levels include The family should be contacted to return to
iron deficiency, inflammatory disorders (due see you. We generally recommend repeating
to decreased iron absorption), increased fetal a venous sample to ensure an accurate result
hemoglobin, and rarely, porphyria. prior to contacting the public health depart-
ment and instituting any treatment (if
Management of lead toxicity required). An environmental screen should
The primary aims of management are pre- be conducted and the patient ruled out for
vention of future lead exposure and result- any pica behavior. Concomitant iron defi-
ant absorption, as well as enhancement of ciency should be ruled out as pica behavior
excretion. The steps to accomplish these leading to ingestion of lead may be due to
goals include: underlying iron deficiency.
1. Assessment of the environment to elimi- b. A repeat venous lead level comes
nate the sources of exposure or removal of the back at 24 mcg/dl. The patient is also
child from the contaminated environment. noted to have a microcytic anemia with
2. Modifying the child’s behavior to hemoglobin of 9.4 g/dl and mean cor-
decrease hand‐to‐mouth activity. puscular volume (MCV) of 69 fl. What
3. Ensuring adequate nutrition, especially are the next steps?
minerals, to limit lead absorption, including The confirmatory testing shows that the
evaluation for concomitant iron deficiency. patient has lead toxicity. The public health
4. Administering medications (chelators) department must be contacted to do an
in children with very high lead levels to environmental assessment. Since the child
increase lead excretion. has concomitant iron deficiency, there is a
chance that he ingested lead and therefore
After chelation therapy, a period of re‐ an abdominal radiograph should be under-
equilibration for 10–14 days should be taken. Any siblings of the patient should
allowed, prior to repeat assessment of the also be screened for lead toxicity with venous
blood lead concentration. Subsequent samples and treated as appropriate based on
treatments should be based on these levels, their levels. The mild iron deficiency anemia
84 Chapter 6

should be concomitantly treated with iron at Brittenham, G.M. (2011). Iron‐chelating therapy
a dose of 3–4 mg/kg elemental iron daily to for transfusional iron overload. N. Engl. J.
prevent pica behavior. The family should be Med. 364: 146–156.
counseled on appropriate nutrition as well Chandra, L. and Cataldo, R. (2010). Lead poison-
ing: basics and new developments. Pediatr.
as methods to avoid lead. Potential unusual
Rev. 31: 399–406.
sources of lead such as toys or candies from
Gracia, R.C. and Snodgrass, W.R. (2007). Lead
international countries should be consid- toxicity and chelation therapy. Am. J. Health
ered and ruled out. After these interven- Syst. Pharm. 64: 45–53.
tions, the patient should be retested in 2 Kwiatkowski, J.L. (2011). Real‐world use of iron
weeks to ensure improvement in lead level, chelators. Hematology Am. Soc. Hematol.
and treatment as appropriate based on the Educ. Program 1: 451–458.
subsequent level. Marsella, M. and Borgna‐Pignatti, C. (2014).
Transfusional iron overload and iron chela-
tion therapy in thalassemia major and sickle
Suggested reading cell disease. Hematol. Oncol. Clin. North Am.
28: 703–727.
Bellinger, D.C. (2008). Very low lead exposures
and children’s neurodevelopment. Curr. Opin.
Pediatr. 20: 172–177.
7 Approach to the
Bleeding Child
Hemostasis is a critical protective response abrasions, or abnormal intraoperative bleed-
of the body to reverse a loss of vascular ing. Aberrations in secondary hemostasis are
integrity and prevent excessive blood loss. It characterized by bleeding from large vessels
requires a coordinated interaction between with subcutaneous, palpable hematomas,
platelets, vascular endothelial cells, and hemarthroses, or intramuscular hematomas.
plasma clotting factors. The first and pri- The hemophilias are examples of disorders in
mary stage of hemostasis is the formation of this category and are reviewed in Chapter 9.
a platelet plug, which involves a complex
interaction between circulating platelets and
the exposed vascular subendothelial layer. Evaluation of the bleeding
The steps in the process include platelet child
adhesion, mediated by an interaction
between von Willebrand factor (VWF) and There are three critical questions that must
platelet surface glycoprotein (Gp) Ib; and be addressed when faced with a child who is
platelet activation, mediated by platelet sur- actively bleeding or by history has experi-
face glycoprotein IIb/IIIa and leading to enced a major hemorrhage. The first two
release of platelet contents. Gp IIb/IIIa inter- questions are, “is the patient continuing to
acts with VWF and fibrinogen, leading to bleed?” and “is the patient hemodynami-
platelet aggregation and enlargement of the cally stable?” These questions should be
platelet plug. This lays the foundation for answered quickly and simultaneously.
the formation of a fibrin clot, the secondary Patients who are actively bleeding but are
stage of hemostasis caused by the activation hemodynamically stable should have efforts
of the coagulation cascade. directed at controlling the bleeding. While
Clinical disorders associated with abnor- therapies that are specific for a particular
malities of primary hemostasis include bleeding disorder should not be provided
­vascular abnormalities, qualitative abnormal- before a diagnosis is made, more general
ities of the platelets, quantitative abnormal- strategies such as ice, pressure, and elevation
ities of the platelets, and von Willebrand can be used. Recombinant factor VIIa is
disease (VWD). These aberrations in primary being increasingly used to control severe or
hemostasis are characterized by ­bleeding of life‐threatening bleeding caused by a variety
the mucous membranes, epistaxis, and super- of mechanisms. Patients who do not appear
ficial ecchymoses. Typical manifestations are to be actively bleeding but are hemodynami-
prolonged oozing from minor wounds or cally unstable require rapid initiation of

Handbook of Pediatric Hematology and Oncology – Children’s Hospital & Research Center Oakland,
Third Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
© 2021 John Wiley & Sons Ltd. Published 2021 by John Wiley & Sons Ltd.
86 Chapter 7

v­ascular re‐expansion and a search for A family history and pedigree are cru-
occult bleeding. cial, as many bleeding disorders are heredi-
Once the patient is stabilized and bleed- tary. The history should also address the use
ing is controlled, the third critical question is of over‐the‐counter and prescription drugs
whether the child presenting with bleeding that can induce bleeding. Common offend-
warrants an evaluation for a bleeding disor- ers include aspirin, ibuprofen, naproxen,
der. Examples of excessive bleeding include and other nonsteroidal anti‐inflammatory
epistaxis lasting more than 15 minutes drugs (NSAIDs). It is important to ask the
despite the appropriate application of pres- patient specifically about the use of medica-
sure to the side of the nose, significant blood tions for colds, sinus trouble, muscle aches,
loss following a dental procedure lasting or headaches, which may contain these
more than 24 hours or requiring blood medications. It is also important to ask
transfusion, bruising that seems excessive whether the patient has access to oral anti-
following trauma, or menorrhagia (heavy coagulant medications prescribed to other
menstrual bleeding lasting for 7 days or loss family members, such as warfarin, clopi-
of more than 80 ml of blood per cycle). When dogrel, prasugrel, or ticagrelor. Some anti-
evaluating a child for “excessive bruising,” it biotics, penicillins in particular, can affect
is essential to determine whether the bruis- platelet function or be associated with
ing could be the result of nonaccidental ­specific inhibitors of clotting, while others
trauma. Bruising involving the scalp, back, such as vancomycin can be associated with
or chest or having a pattern suggestive of thrombocytopenia through an immune‐
common instruments of abuse such as belts, mediated process. Anticonvulsants can
cords, or wire should be reported. Excessive cause thrombocytopenia, and procainamide
bruising caused by underlying bleeding dis- has been associated with an acquired lupus
orders occurs on areas more commonly anticoagulant.
involved in falls or trauma such as shins and
bony prominences. Physical examination
In addition to the routine examination, the
History skin should be scrutinized carefully for
Assessment of the child with a suspected or petechiae, purpura, and venous telangiecta-
known bleeding diathesis begins with a sias. The joints should be examined for
complete history. The nature of the bleeding swelling or chronic changes such as contrac-
should be explored with particular attention tures or distorted appearance with asymme-
to location, duration, frequency, and the try related to repeated bleeding episodes.
measures necessary to stop it. The time of Mucosal surfaces, such as the gingiva and
the patient’s first episode of bleeding should nares, should be examined for bleeding.
be documented and a careful history of
bruising during the toddler age is impor- Initial laboratory evaluation
tant. A previous history of bleeding asso- The purpose of the initial laboratory
ciated with trauma or surgery, dental ­evaluation is to screen for the presence of a
extraction, circumcision, or tonsillectomy is bleeding disorder, hopefully categorize the
also important, as is a history of petechial disorder as primary or secondary, and direct
rash, arthritis with hemarthroses, or blood further evaluation. Appropriate screening
transfusion. In females, the duration and tests include a CBC, peripheral blood smear
severity of menstrual bleeding should be (PBS), prothrombin time (PT), and acti-
documented. vated partial thromboplastin time (APTT,
Approach to the Bleeding Child 87

hereafter PTT). The international normal- The PT measures the integrity of the
ized ratio (INR) corrects differences in PT extrinsic system of coagulation (extrinsic
due to variable thromboplastin potency secondary to the necessity of activation by
across laboratories. In certain circum- tissue factor). Factor VII is the only coagula-
stances, this list will also include a fibrino- tion factor measured by the PT that is not
gen and a thrombin time (TT). measured by the PTT. Thus, in isolated fac-
A CBC provides several pieces of useful tor VII deficiency, the PT is prolonged with
information. It provides the platelet count, a normal PTT. Factor VII is vitamin–K‐
identifying in most cases the presence or dependent and has a very short half‐life, so
absence of thrombocytopenia. However, it is the PT is one of the most sensitive measures
important to review an actual blood smear, of oral anticoagulant therapy with vitamin K
as a small number of patients with a low antagonists such as warfarin. The INR is
measured platelet count will have pseudo- used primarily to evaluate the adequacy of
thrombocytopenia, a condition caused by warfarin therapy; the PT should be used to
platelet clumping in the presence of the anti- look for the presence of a clotting factor
coagulant EDTA. The CBC also provides deficiency.
the hemoglobin and mean corpuscular vol- The PTT assesses the integrity of the
ume (MCV), which can provide clues intrinsic system of coagulation (intrinsic
regarding the duration and severity of the secondary to direct endothelial activation).
patient’s bleeding. The presence of anemia Depending on the laboratory methods, the
indicates a clinically significant bleeding PTT will be normal when the activity of all
disorder; the concomitant presence of measured coagulation factors is at least 30%
microcytosis suggests the bleeding has been of normal. It is prolonged in patients with
prolonged and has led to iron deficiency. A hemophilia, and in some patients with
normocytic anemia suggests the bleeding VWD due to decreased concentration of
has been more recent and likely more severe, factor VIII in the plasma. However, the PTT
as the blood loss has likely been more sig- is much more susceptible than the PT to
nificant. Abnormalities in more than one spurious abnormalities caused by errors in
cell line (anemia, thrombocytopenia, or collection or processing of the specimen.
neutropenia) suggest the presence of a bone These are outlined in Table 7.1.
marrow failure state such as aplastic anemia The most commonly encountered inhib-
or marrow infiltration as is seen in leuke- itors detected in children are so called lupus
mia. Most analyzers provide a measure of anticoagulants. These are IgG antibodies
the mean platelet volume (MPV) that can be directed against phospholipids, and while
useful in evaluating the thrombocytopenic they are commonly identified in adults with
patient, however visualization of the plate- autoimmune disease, they occur frequently
lets on the smear is often more accurate, as in children as a postinfectious phenomenon
automated MPV may be challenging with that is short‐lived. As phospholipid is an
severe thrombocytopenia or clumping. essential cofactor in the PTT assay, antiphos-
Large platelets are often seen in consump- pholipid antibodies will commonly cause
tive thrombocytopenia such as active prolongation of the test. Addition of normal
bleeding or immune thrombocytopenia
­ plasma (with additional phospholipid) as is
purpura (ITP); normal‐sized platelets in done in mixing studies will often at least
hypoproliferative thrombocytopenia; and partially normalize the test, but it will most
small platelets in inherited conditions such often remain abnormal. These antibodies
as Wiskott–Aldrich syndrome. have no effect on clotting in vivo, and
88 Chapter 7

Table 7.1 Factors affecting the validity of the PTT test.

Aspect Problem Remarks


Sample Clots Clots caused by slow blood flow or delay in
collection transferring sample will cause abnormal results.
Plasma volume Volume of anticoagulant must be corrected for
plasma volume. If tube not completely filled, or
patient 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
values period (immaturity of the coagulation system).
Inhibitor vs. factor A number of agents can interfere with the PTT
deficiency assay and cause a factitious prolongation of
the test. If the concentration is high, they can
lead to a mild prolongation 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 in the text).
Abbreviations: PTT, partial thromboplastin time; PT, prothrombin time.

patients with lupus anticoagulants do not almost always associated with heparin or
have clinical bleeding; in fact, their most an inhibitor.
common coagulation problem is thrombo- Abnormalities in one or more of the
philia rather than bleeding. abovementioned screening tests will be
The TT is useful in evaluating the ter- noted with most bleeding disorders. An
minal steps of coagulation and identifying approach to identifying the most likely
anticoagulants present in plasma. The TT coagulation disorders given the results of
is abnormal when the plasma concentra- these tests is presented in Table 7.2.
tion of fibrinogen is decreased (hypofibrin- Once the diagnosis is made, specific
ogenemia or afibrinogenemia), when the treatment can be provided based on the rec-
fibrinogen present is dysfunctional (dysfi- ommendations provided in the following
brinogenemia), or when there are circulating chapters. One exception is the management
anticoagulants (heparin) or fibrin degrada- of epistaxis. Though epistaxis is caused by a
tion products. As dysfibrinogenemia is variety of conditions, in most situations an
most often asymptomatic, a prolonged TT initial uniform management plan is appro-
in the face of a normal fibrinogen level is priate and therefore presented here.
Approach to the Bleeding Child 89

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 (FXIII screening;
do FXIII assay if abnormal)
Euglobulin clot lysis (replaced by
TEG in most places); can also
measure specific functional assays
Alpha‐2‐antiplasmin, PAI‐1, TPA
PT normal PTT inhibitor PTT mixing study
PTT prolonged von Willebrand disease Factor assays (i.e., VIII, IX, XI)
Platelet count Hemophilia A (FVIII deficiency) von Willebrand studies
normal or B (FIX deficiency) Thrombin time/reptilase time
Heparin contamination
PT prolonged PT inhibitor PT mixing study
PTT normal Vitamin K deficiency Factor assays (i.e., II, VII, IX, X)
Platelet count Warfarin
normal Factor VII deficiency
Liver dysfunction
PT prolonged Circulating inhibitor PT/PTT mixing studies
PTT prolonged Liver dysfunction TT
Platelet count Vitamin K deficiency Reptilase time (unaffected by
normal Factor deficiency (II, V, X, or heparin)
fibrinogen) Fibrinogen
Dysfibrinogenemia Factor assays
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 or Tests for Helicobacter pylori
immune deficiency (i.e., HIV, hepatitis panel
common variable Anticardiolipin antibodies
immunodeficiency) Direct antiglobulin test
Von Willebrand disease Serum immunoglobulin levels
Early stage of bone marrow failure Serum complement levels
syndrome von Willebrand factor multimer
analysis (type IIB)
Bone marrow aspirate
Marrow chromosomal analysis

Abbreviations: PT, prothrombin time; PTT, partial thromboplastin time; PFA‐100, platelet
function analyzer‐100; TEG, thromboelastography; PAI‐1, plasminogen activator inhibitor‐1;
TPA, tissue plasminogen activator; TT, thrombin time; DIC, disseminated intravascular
coagulation; ITP, immune thrombocytopenic purpura; HIV, human immunodeficiency virus.
90 Chapter 7

strictor) applied to the inserted end of the


Management of epistaxis
sponge can provide additional hemostasis.
Anterior packs may be left in place for
1. Place patient in a sitting position to
1‐5 days, though should be removed
decrease venous pressure or, if the patient is
within 24 hours in an immune‐compro-
recumbent in bed, turn head to the side.
mised patient due to the risk of infection.
Keep the head higher than the level of the
Humidification and nasal saline spray can
heart. Do not allow patient to lie flat.
help prevent drying and crusting of the oral
2. Flex neck anteriorly, with the chin touch-
mucous membranes as a result of mouth
ing the chest.
bleeding. Broad‐spectrum antibiotics, to
3. With the thumb and index finger, pinch
cover skin flora as well, should be consid-
the soft parts of the nose. Hold pressure
ered in patients who are immunosuppressed
firmly over the lower half of the nose. Avoid
with a nasal pack in place. Vaseline gauze
compressing the upper half of the nose.
packing, when placed correctly and snugly,
4. Hold pressure for 20 minutes with the
is a reliable means of packing. Packing may
head in a flexed position. If manual pres-
be soaked in a solution of 4% lidocaine and
sure is stopped momentarily to examine or
topical epinephrine (1:1000) to provide
change dressings, the 20‐minute digital
local anesthesia and vasoconstriction.
pressure will likely need to start again.
Pressure and time allow for clot formation
to occur. If bleeding continues, reassess Case study for review
location of digital pressure and reapply. If
bleeding stops and recurs, repeat manual A 3‐year‐old female presents to the emer-
pressure for 20 minutes. If bleeding contin- gency department (ED) with petechiae and
ues and clot(s) can be visualized, remove repeated epistaxis. Two weeks earlier she
clot(s) and reapply digital pressure for had been admitted to an outside hospital
20 minutes. with epistaxis accompanied by two episodes
5. Advise patient not to blow nose for at of vomiting dark red blood. Results of the
least 12 hours to avoid dislodging the clot. laboratory evaluation included: WBC count
6. Nasal packing may be indicated if the 8.2 × 109/l; hemoglobin 10.7 g/dl; platelet
source of bleeding is not well‐visualized or count 142 × 109/l, PT, 11.5 seconds (normal);
bleeding is profuse. Types of packing include APTT 31.2 seconds (normal); and fibrino-
compressed sponge, Vaseline gauze packing, gen, 2.5 g/l (RI, 2.0–4.0 g/l). The patient was
Gelfoam, or topical thrombin packing. discharged in good condition after inser-
Compressed sponges are compressed when tion of nasal packs. One day prior to the
dry and expand when wet. The expansion current presentation, the patient again had
produces active and passive absorption and prolonged epistaxis, this time accompanied
places gentle pressure on the mucosa. The by four episodes of hematemesis and a mel-
nasal sponge should fit snugly through the anotic stool. Her parents denied fever or
naris and be placed along the floor of the diarrhea. She is not currently receiving any
nasal cavity. Sponges are easy to insert and medications. The patient had a history of
can be removed with little discomfort. easy bruising and recurrent gingival bleed-
Neosporin can be applied to the sponge for ing, but no history of hemarthrosis. There
ease of insertion and act as an antimicrobial was no family history of abnormal
agent. Topical thrombin powder (a vasocon- bleeding.
Approach to the Bleeding Child 91

On examination, she appeared pale. t­hrombasthenia, an inherited platelet func-


Her vital signs were normal. Her skin had tion abnormality discussed more fully in a
scattered petechiae. Crusted blood was subsequent chapter.
­present in both nares. The physical exami-
nation was otherwise unremarkable.
a. What would you do next? Multiple choice questions
It appears that at present the patient is not
actively bleeding and is hemodynamically 1. You are seeing a 12-year-old male patient
stable, so efforts can be directed to deter- in the emergency department with recurrent
mining if a bleeding disorder is present. epistaxis. All of the following are reassuring
The patient’s PT and PTT were normal that there is no underlying bleeding diathesis
initially, making a coagulopathy less EXCEPT:
likely. Also, epistaxis is more commonly a. Normal coagulation studies
associated with VWD, thrombocytope- b. No family history of bleeding
nia, or platelet function abnormalities. problems
The platelet count was normal, ruling out c. Patient has a mild microcytic anemia
thrombocytopenia. d. No personal history of easy bleeding
b. In order to identify a possible bleed- or bruising beyond the epistaxis
ing disorder, what other laboratory stud- e. No noted blood in the urine or stool
ies might be helpful? Explanation: Risk of bleeding can often be
A repeat CBC would be indicated given the determined by obtaining a thorough per-
additional bleeding that has occurred. The sonal and family history. Important ques-
WBC count was 21 × 109/l, hemoglobin 6.1 tions to ask include a family history of
g/dl, MCV 84.7, platelet count 384 × bleeding which required blood transfu-
109/l,and reticulocyte count 6.8%. These sions, which lead to difficulty with surger-
results indicate that despite a healthy bone ies, and with special consideration to male
marrow response the anemia is worsening, members of the maternal side when
consistent with significant blood loss. The ­considering hemophilia A or B in a male
platelet count is normal, again ruling out patient. History of menorrhagia and
thrombocytopenia as a cause of the bleed- ­problems of bleeding during delivery are
ing. The MCV is normal, suggesting that the also important questions to ask. Personal
anemia is due to acute blood loss rather than history should concentrate on the type of
to iron deficiency from chronic, ongoing bleeding, frequency of symptoms, duration
blood loss. of symptoms and previous need for inter-
Repeat PT is 10.3 seconds, PTT is 25.5 ventions. Utilization of medications
seconds; both are normal. VWF antigen is including aspirin and NSAIDs should be
212%, factor VIII is 182%, ristocetin cofac- obtained. Finally, baseline screening evalu-
tor activity is 160%. These results rule out ation to look for anemia, thrombocytope-
VWD. nia and coagulopathy should be ascertained
Platelet aggregation studies reveal an through a CBC and coagulation panel.
absent response to ADP, epinephrine, ara- Microcytic anemia may be due to chronic
chidonic acid, and collagen, but a normal occult blood loss and may point to an
response to ristocetin. This pattern of underlying bleeding diathesis in this case.
response is diagnostic of Glanzmann The answer is c.
92 Chapter 7

2. You are seeing a 14-year-old female patient d. Hemophilia B


who notes heavy periods as compared to her e. Hemophilia C
older sister requiring >6 heavy pads per day. Explanation: Disorders in the intrinsic path-
She notes that her mother has a similar com- way include factor XI (hemophilia C), factor
plaint and suffers from anemia but has had IX (hemophilia B) and factor VIII (hemo-
no further work up or diagnosis. She eats a philia A), especially in the male patient. For
varied diet and has no other medical com- prolonged PTT, heparin contamination can
plaints. She does not take any other medica- be ruled out by repeating the test or check-
tions. On screening labs you note she has a ing a thrombin time which is very sensitive
microcytic anemia with normal coagulation to heparin (i.e., normal thrombin time rules
studies. Of the following the most likely out heparin contamination). A 1:1 mix will
diagnosis is: help differentiate between a factor defi-
a. Vitamin K deficiency ciency in which addition of normal plasma
b. Hemophilia A will correct the PT or PTT prolongation or
c. Immune thrombocytopenic purpura an antibody, in which case the 1:1 mix will
d. Von Willebrand disease not correct. The answer is a.
e. Dysfibrinogenemia
Explanation: The most likely diagnosis in 4. You are following a patient who has been
this case is dysfunctional uterine bleeding in the PICU for quite some time recovering
which is common but given the above from sepsis. There is concern for underlying
choices, von Willebrand disease (VWD) is organ dysfunction and the patient has been
most likely and affects as much as 1% of the on total parenteral nutrition (TPN) for quite
population. Symptoms can be non‐specific some time. A coagulation panel is checked
and there may or may not be a family his- and is notable for a prolonged PT with a
tory. PTT may be prolonged in certain normal PTT. Of the following, all are possi-
cases due to concomitant deficiency of ble diagnoses EXCEPT:
FVIIIc. Similarly iron deficiency anemia a. Vitamin K deficiency
and/or thrombocytopenia may be present. b. Liver disease
Screening for VWD should include meas- c. FVII deficiency
urement of von Willebrand factor antigen, d. Presence of an inhibitor
ristocetin cofactor (von Willebrand factor e. Heparin contamination
activity) and activated FVIII. The answer Explanation: Isolated prolongation of the
is d. PT, or the extrinsic pathway can occur for
multiple reasons. Vitamin K deficiency
3. You are seeing a 4-year-old male patient affects multiple coagulation factors includ-
in the hospital admitted with vomiting and ing FII, FVII, FIX and FX in addition to pro-
altered mental status eventually found to have tein C and protein S. As FVII has the shortest
bilateral subdural hematomas on CT head half‐life, mild vitamin K deficiency may
imaging. Non‐accidental trauma is possible affect the extrinsic pathway prior to the
and a hematology work up is commenced. intrinsic pathway. This is similarly the case
The PT is normal while the PTT is prolonged. in liver disease as the majority of coagula-
The PTT corrects on 1:1 mix. Of the follow- tion factors are produced in the liver. FVII
ing, all are possible diagnoses EXCEPT: deficiency is extremely rare but would also
a. Vitamin K deficiency lead to isolated prolongation of the PT. As
b. Heparin contamination no 1:1 mix was noted, the PT prolongation
c. Hemophilia A could also be due to an antibody which
Approach to the Bleeding Child 93

would lead to the PT remaining prolonged i­ liopsoas bleed, the first step is to give factor
on 1:1 mix. Heparin specifically affects the replacement, ideally with the patient’s home
intrinsic pathway and would only affect the medication supply. CT head is not indicated
PT if there was significant heparinization, in this case unless the exam or history are
but this would occur in conjunction with a concerning. The other steps are all impor-
very high PTT. The answer is e. tant but can be done after factor is given.
Factor levels will be important to follow to
5. You are seeing a 12-year-old male with ensure the patient is receiving therapeutic
severe hemophilia A. He has been poorly dosing, with the goal of achieving 100%
compliant with his prophylactic factor infu- ­factor levels. In the case of hemophilia A,
sions and presents to the ED complaining of the patient should receive a dose of 50 units
severe hip pain and inability to fully rotate x weight (kg) to achieve 100% factor levels,
the hip. Of the following the most important assuming there is no concomitant inhibitor
initial consideration is: (and rounded up to the nearest vial size).
a. Checking factor levels The answer is b.
b. Administering recombinant FVIII
c. Performing a CT to rule out an occult Suggesting reading
head bleed
d. Performing a CT to rule out an iliop- Allen, G.A. and Glader, B. (2002). Approach to
soas bleed the bleeding child. Pediatr. Clin. N. Am. 49:
e. Determining his full medical history 1239–1256.
Explanation: Severe hemophilia implies a Bansal, D., Oberoi, S., Marwaha, R.K., and Singhi,
factor level of <1%, with moderate hemo- S.C. (2013). Approach to a child with bleeding
philia has a factor level of 1%‐5% and mild in the emergency room. Indian J. Pediatr. 80:
411–420.
hemophilia a factor level of 5%‐50%.
Khair, K. and Liesner, R. (2006). Bruising and
Patients with severe hemophilia are main- bleeding in infants and children – a practical
tained on prophylactic factor to prevent approach. Br. J. Haematol. 133: 221–223.
spontaneous bleeding episodes, one of the Monagle, P., Ignjatovic, V., and Savoia, H. (2010).
most severe being an iliopsoas bleed. Hemostasis in neonates and children: pitfalls
Although it is important to rule out an and dilemmas. Blood Rev. 24: 63–68.
8 Von Willebrand
Disease
Von Willebrand disease (VWD) is the most VWD is classified into three types: Type
common inherited bleeding disorder, affect- 1, found in up to 85% of cases, is charac-
ing as much as 1% of the general population, terized by partial deficiency of normally
and equally affects both sexes as well as all functioning VWF. Type 2 is characterized
races and ethnicities. The actual prevalence by functional defects in VWF. It is broken
is difficult to determine, as many affected down into four subtypes. Type 2A is marked
individuals are either asymptomatic or have by defects in multimerization and a con-
such mild symptoms that they do not seek comitant decrease in platelet adhesion. The
medical attention. While most patients with measured amount of VWF:Ag (antigen) is
VWD have the inherited form, an acquired normal or only slightly decreased, but the
form also occurs. The three characteristic VWF:RCo (ristocetin cofactor) is much
clinical features of the inherited form are lower. Type 2B is caused by mutations that
excessive mucocutaneous bleeding, abnormal pathologically increase platelet–VWF bind-
von Willebrand factor (VWF) laboratory ing, which leads to the proteolytic degrada-
studies, and a family ­history of abnormal tion and depletion of large, functional VWF
bleeding. multimers. Circulating platelets are also
VWF is a large multimeric glycoprotein coated with abnormal VWF, which may
that is synthesized and stored in megakary- prevent the platelets from adhering at sites
ocytes and endothelial cells. VWD is usually of injury. The VWF‐coated platelets often
inherited in an autosomal dominant man- become sequestered in the microcircula-
ner, but a rare autosomal recessive form and tion, leading to thrombocytopenia. Type
an X‐linked recessive form have also been 2M is characterized by defects in ligand
described. VWD results from either a binding between VWF and platelet GPIb or
­deficiency or a defect in the VWF protein. connective tissue. The ratio of VWF:RCo
This protein plays two critical roles in hemo- to VWF:Ag is low as in Type 2A, but the
stasis: the large multimers bind to platelet multimer panel appears normal. Type 2N
­glycoprotein Ib (GPIb) causing platelet acti- is marked by decreased binding between
vation and adherence to damaged endothe- VWF and FVIII, resulting in a phenotype
lium, and it is the carrier protein for factor of autosomal recessive hemophilia. Type 3
VIII (FVIII), stabilizing it and protecting VWD is a rare condition affecting about
it from degradation and clearance from 1 per one million individuals, and is charac-
plasma. This accounts for the prolonged terized by the almost complete absence
APTT seen in some patients. of VWF.

Handbook of Pediatric Hematology and Oncology – Children’s Hospital & Research Center Oakland,
Third Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
© 2021 John Wiley & Sons Ltd. Published 2021 by John Wiley & Sons Ltd.
96 Chapter 8

The majority of affected patients experi- ­ leeding questionnaire (PBQ) has also been
b
ence mucocutaneous bleeding such as shown to be useful as a screening tool
epistaxis, easy bruising, and menorrhagia for VWD in the pediatric population.
in women. Gastrointestinal bleeding is a Unfortunately, while its negative predictive
particular problem for patients with Type value is high, its positive predictive value is
2A VWD. All VWD patients may also low due to the wide range of normal bleed-
experience post‐traumatic or postsurgical
­ ing symptoms seen in normal individuals.
bleeding. Patients with severe VWD may Often, once the child is diagnosed with
experience bleeding into muscles and joints VWD, a parent or siblings are found to have
as well. VWD should be suspected in the the disorder after screening.
patient with platelet‐type bleeding and a In patients with mild VWD, the stress
family history of a bleeding diathesis. associated with serious operations or child-
birth may prevent symptoms. VWF is an
acute phase reactant and the level will fre-
Clinical presentation quently increase into the normal range fol-
lowing surgery, in pregnancy, or in patients
The clinical features of VWD can be quite with active liver disease or collagen vascular
variable. In most cases of VWD, the bleed- disease. It can be reduced in hypothyroid-
ing is mild. Mucosal bleeding such as ism. Even the stress of phlebotomy can
epistaxis, gingival bleeding with tooth increase the level of VWF, which can make it
brushing, ecchymoses, or menorrhagia are difficult to confirm the diagnosis even with
classic. The initial presentation, however, repeat testing. Neonates have elevated VWF
may be postoperative bleeding, such as fol- levels following vaginal delivery, making it
lowing a tonsillectomy and adenoidectomy difficult to diagnose in the neonatal period.
or dental extraction. In retrospect, the child VWF levels can be up to 25% lower in indi-
who was thought to have normal childhood viduals who are blood type O, increasing the
complaints (bruising and epistaxis) is real- difficulty in distinguishing between VWD
ized to actually have a bleeding disorder. and normal individuals with a low VWF
Recurrent or severe epistaxis, particularly level. VWF levels also increase physiologi-
in the older child or adult, is unusual and cally throughout life; patients diagnosed
warrants investigation. Since other affected with VWD in childhood may no longer
family members are often unaware they meet the VWF level criterion when they get
have the condition, simply inquiring about a older. However, many of them do still appear
history of bleeding problems often yields to be at risk for bleeding.
negative results. Because the bleeding his-
tory is so important for the diagnosis of
VWD, many attempts have been made to Diagnosis
objectively quantify reported bleeding
symptoms by developing bleeding assess- Screening tests are of little value in making
ment tools. The International Society on the diagnosis of VWD. The CBC may dem-
Thrombosis and Haemostasis (ISTH) has onstrate iron‐deficiency anemia if the
developed and performed initial validation patient’s blood loss has been significant, but
of a bleeding assessment tool for use in this is nonspecific. The rare patient with
screening patients for VWD. Normal ranges Type 2B VWD may have mild thrombocy-
have been established for children, adult topenia. If the level of VWF is sufficiently
males, and adult females. A pediatric low, the APTT may be prolonged due to
Von Willebrand Disease 97

decreased concentration of factor VIII, but The most common type of VWD (Type 1)
this is neither sensitive nor specific. The has a normal multimeric analysis.
platelet function analyzer (PFA‐100) is a
more useful screening test for VWD. It The classification of patients with VWD
measures the time required for blood, drawn based on the results of these tests is pre-
through a fine capillary, to block a mem- sented in Table 8.1.
brane coated with collagen and epinephrine Recent advances in diagnostic testing
(CEPI) or collagen and adenosine diphos- include the use of the VWF:GPIbM assay in
phate (CADP). Its sensitivity in identifying place of measuring VWF:RCo due to several
patients with VWD is fairly high, but its issues with the latter that limit its usefulness.
specificity is low. Four specific tests have The assay introduces gain‐of‐function
traditionally been used to diagnose VWD: mutations into GPIbα, allowing it to bind
1. Factor VIII coagulant (FVIIIc): functional spontaneously to VWF without ristocetin.
measurement of FVIII coagulant activity, At the present time access to this test is lim-
which is carried in the circulation by VWF. ited in the United States, but it is widely
2. Von Willebrand factor antigen (VWF: available in Europe and Canada. VWF also
Ag): VWF protein as measured by protein functions to bind collagen exposed follow-
assays; does not imply functional ability. ing tissue injury. The nature of this interac-
Immunologic quantitation of VWF by tion is complex, but is very dependent on
either the quantitative immunoelectropho- the presence of high‐molecular‐weight mul-
retic assay (Laurell assay) or the enzyme‐ timers. Patients with increased bleeding
linked immunosorbent assay (ELISA). scores and unexplained bleeding symptoms
3. Von Willebrand factor activity (VWF: may benefit from collagen‐binding testing
RCo): ristocetin induces the binding of to explore the possibility of an undiagnosed
VWF to the GPIbα receptor on formalin‐ collagen‐binding defect in VWF. The avail-
fixed platelets. The slope of the platelet ability of this testing is unfortunately also
agglutination curve correlates with the level limited at the present time.
of plasma VWF. Leebeek and Eikenboom (see Suggested
4. Multimeric analysis: an agarose gel readings) outline a system for diagnosing
­electrophoretic study used to identify quanti- VWD and identifying the subtype based
tative or qualitative multimer abnormalities. on available laboratory testing. Patients with

Table 8.1 Classification of VWD.

VWD VWF:Ag VWF:RCo FVIII:C RCo:Ag ratio Multimer pattern


subtype

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 ristocetin cofactor activity; FVIII:C, Factor VIII coagulant; N, normal.
98 Chapter 8

undetectable (or <5 IU/dl) levels of VWF:Ag


Treatment
have Type 3 VWD. However, some patients
with severe Type 1 VWD also have antigen
The goal of treatment in VWD is to control
levels <5 IU/dl. Measurement of VWF pro-
or prevent serious or life‐threatening bleed-
peptide, produced during synthesis of VWF,
ing. There are no effective therapies to limit
allows distinction between these two enti-
the chronic bruising that many active chil-
ties. Patients with Type 3 disease have absent
dren experience, and parents need to be
or very low levels of VWF propeptide, while
given assistance to accept that reality. Many
patients with severe Type 1 have minimally
children and adults with Type 1 VWD,
reduced or normal levels.
especially males, never require therapy.
­
If patients have measurable levels of
Individuals with Type 2 or Type 3 VWD are
VWF:Ag, the next step is to compare the
much more likely to need treatment at
Ag level with the measured level of activity.
­various times throughout life.
Currently this is most commonly measured
There are three strategies used to treat
as VWF:RCo activity. Patients with a pro-
patients with VWD: (i) increase the plasma
portional decrease in activity (VWF:RCo/
concentration of VWF by releasing endoge-
VWF:Ag >0.6) have Type 1 VWD, while
nous VWF stores through stimulation of
patients with a disproportionate decrease
endothelial cells with desmopressin; (ii)
in activity have Type 2 disease. Further
replace VWF by infusing exogenous coagu-
subtyping testing such as VWF multimers
lation factors (high‐purity VWF concen-
and ristocetin‐induced platelet aggregation
trate or low‐purity factor VIII‐VWF
(RIPA) are then used to distinguish
concentrate); and (iii) use agents that pro-
between Types 2A, 2B, and 2M disease.
mote hemostasis and wound healing, but do
RIPA is normal in Type 2N and can be
not substantially alter the plasma concentra-
decreased in Types 2A and 2M. A ratio of
tion of VWF. These strategies are not mutu-
FVIIIc to VWF:Ag of ≤0.6 may indicate
ally exclusive; different bleeding episodes
Type 2N VWD; this can be confirmed by
may require a combination of two or even all
measuring the binding of factor VIII to
three of these strategies to be effective.
VWF. Normal binding confirms the diag-
Desmopressin (DDAVP) is the treatment
nosis of mild hemophilia A.
of choice in mild Type 1 disease (up to 85%
Using this algorithm, the diagnosis of
of all cases) and may be of benefit with some
Type 2 or Type 3 VWD is very straightfor-
of the other variants. It stimulates endoge-
ward. However, it remains difficult to deter-
nous release of VWF from endothelial cells.
mine an unambiguous level of VWF:Ag to
A good response is defined as an increase in
use to define Type 1 disease. There is a linear
VWF level of at least threefold, and to a
relationship between VWF:Ag level and
hemostatic level. The approximate half‐life
bleeding phenotype, inheritance pattern,
of these released factors is 8–10 hours, but
and identification of VWF mutations.
patients with VWF clearance defects will
Several current guidelines recommend that
have an accelerated decline in VWF level
patients with bleeding tendency and a
after treatment, limiting the utility of
VWF:Ag level between 30 and 50 IU/dl be
DDAVP in treating severe bleeds. An intra-
classified as having “low von Willebrand
nasal form of DDAVP (Stimate® 1500 mcg/
factor” or “possible Type 1 disease,” but they
ml) is available to treat bleeding in VWD.
are not classified as having VWD.
Typical dosing is one spray for patients
Nonetheless, they may require treatment
<50 kg and two sprays for patients ≥50 kg.
before surgery or with bleeding.
Von Willebrand Disease 99

The peak effect is observed in 30–60 minutes. needed to promote healing and achieve
A therapeutic trial of DDAVP should be hemostasis. Preinfusion and postinfusion
given to determine individual responsive- measurements of VWF:Ag or VWF:RCo
ness to this therapy with measurement of can be done to establish continued clinical
factor VIII and VWF levels before and after responsiveness. However, it may be difficult
administration. After baseline levels are to obtain the results of these tests quickly.
obtained, a standard dose of Stimate is Patients with Type 1 VWD who have
administered, and levels are measured again severe bleeding manifestations unrespon-
60 minutes after administration. The peak sive to DDAVP, or those patients with desm-
effect is typically seen 60–90 minutes after opressin‐unresponsive Type 2 or 3 disease,
infusion. A positive trial is defined as at require treatment with exogenous VWF.
least a threefold increase in the VWF:Ag The Food and Drug Administration (FDA)
and VWF:RCo activities. Ninety percent has approved three plasma‐derived factor
of patients with Type 1 VWD demonstrate a VIII/VWF products for the treatment of
positive response to IV DDAVP. Levels can VWD, Humate‐P®, Wilate®, and Alphanate®.
be checked at later time points to confirm Koate‐DVI is another combination product,
a normal half‐life of the released VWF. If but it has not been studied extensively in
the patient fails a Stimate challenge, an IV VWD and its use is not FDA approved. In
DDAVP challenge can be performed by addition, in 2015, the FDA approved the
administering 0.3 mcg/kg of DDAVP intra- first recombinant VWF product, Vonvendi®,
venously in 30–50 ml of normal saline over for treatment of severe VWD in patients
15–30 minutes, and measuring levels 18 years of age and older. It is important to
60 minutes after infusion. remember that patients with severe VWD
Stimate is most useful for individuals have decreased levels of both VWF and
with more frequent episodes of bleeding FVIII, and both factors typically require
requiring therapy, such as women with replacement. The available agents have dif-
menorrhagia not controlled using other fering ratios of FVIII to VWF, so should not
measures. Given its high cost and relatively be considered interchangeable. Humate‐P
short shelf life (6 months after the bottle is contains 50–100 IU/ml VWF:RCo activity
opened), it is less suitable for individuals and 20–40 IU/ml FVIII activity. It has the
with very infrequent bleeding episodes. highest ratio of VWF:RCo to FVIII activity
Though effective, there is some variability in and the highest concentration of high‐
response due to inconsistent absorption. molecular‐weight multimers. Alphanate
Therefore, patients should be tested for contains 40–180 IU/ml FVIII activity and at
responsiveness prior to prescribing. least 16 IU/ml VWF:RCo activity. Wilate
Patients undergoing surgical procedures has essentially equal amounts of VWF:RCo
such as tonsillectomy or dental extractions and FVIII activity. It has demonstrated both
may need extended therapy to maintain safety and efficacy in patients with VWD.
increased levels of VWF and factor VIII There are no head‐to‐head studies compar-
until healing has occurred. Studies have ing the efficacy of any of these products.
demonstrated that DDAVP can be repeated Wilate’s packaging promotes more frequent
daily for up to 4 days with little loss of effi- dosing with lower doses of product, which
cacy as measured by VWF levels or PFA‐100. the company states is more physiologic and
Tachyphylaxis with repeated dosing should more cost‐effective. Vonvendi contains no
be considered following a major surgery or FVIII activity. After administration, FVIII
when prolonged elevated levels of VWF are levels increase due to Vonvendi’s binding of
100 Chapter 8

FVIII as it is released, but it takes 6–8 hours f­amilies must be instructed to adhere to the
to achieve hemostatic levels in severely every 6 hour schedule. The dose of tranexamic
affected patients. If used for severe bleeding acid is 10 mg/kg/dose IV prior to a procedure
or prior to urgent surgery, factor VIII and then every 6–12 hours for patients with
replacement is required as well. hemophilia. In VWD‐associated menorrha-
The goal of VWF replacement therapy is gia, it is recommended in an oral form
to reach a VWF:RCo level of 100 IU/ml and ­(available in 650 mg tablets to be dosed twice a
nadir of >50 IU/ml. Recommended starting day) to be taken at the initiation of menses for
doses are 40–60 IU/kg of VWF:RCo activity, up to 5 days per month. Both drugs are avail-
followed by 20–40 IU/kg every 8–24 hours able in oral and intravenous forms. These
as needed to maintain desired levels. Major drugs are particularly effective in minimizing
surgical procedures should only be per- bleeding from mucosal surfaces due to the
formed at centers that have the capability of increased level of fibrinolytic activity present
measuring VWF activity levels in‐house on in these areas.
a daily basis. To decrease the risk of throm- Patients with Type 3 VWD have plasma
botic events, VWF:RCo levels should be VWF and FVIII levels that are extremely low
kept <200 IU/ml, and FVIII levels should be (typically less than 5 IU/dl). Desmopressin is
<250–300 IU/ml. usually ineffective in these cases, but a thera-
Other adjuvant therapies are available that peutic trial of desmopressin may be given, as
are often helpful in managing bleeding in occasionally patients will respond appropri-
patients with VWD. Direct pressure is very ately. These patients often have significant
effective in managing epistaxis and bleeding bleeding including profound epistaxis or
from tooth sockets or lacerations. Family hemarthroses similar to that seen in patients
members must be taught to maintain continu- with hemophilia due to their low levels of
ous pressure “without peeking” for at least both FVIII and VWF. With bleeding, such
20 minutes for best results. If the bleeding patients should be managed similarly to
remains uncontrolled, a single dose of desmo- patients with hemophilia, using the previ-
pressin is usually effective to stop bleeding in ously mentioned FVIII concentrates that con-
patients who have shown responsiveness. Low tain adequate levels of VWF and FVIII. Six to
estrogen oral contraceptives have been very ten percent of Type 3 patients will develop
helpful in controlling menorrhagia, as they alloantibodies to VWF replacement products.
minimize the amount of endometrial develop- As with hemophilia patients who develop
ment that takes place. Minor surgical proce- inhibitors, the most common manifestation is
dures, such as laceration repairs or dental an impaired response to infused concentrates.
extractions, can frequently be managed with a However, some patients will develop anaphy-
single treatment of desmopressin or factor lactic reactions as well. Management of bleed-
concentrate followed by antifibrinolytic ther- ing in these patients is complicated and may
apy for 7–10 days. Two antifibrinolytic agents involve use of recombinant FVIII product,
are available, epsilon aminocaproic acid recombinant FVIIa, and/or activated pro-
(Amicar®) and tranexamic acid (Lysteda®); thrombin complex. Although the use of these
Amicar is the more widely used agent. The products seems reasonable, experience is
oral dose of aminocaproic acid is 100–200 quite limited, and caution is warranted.
mg/kg (maximum dose, 10 g) as the initial Patients with Type 2A VWD have abnor-
dose, followed by 100 mg/kg (maximum dose, mally small VWF multimers and desmo-
5 g) every 6 hours. To maintain e­ ffectiveness, pressin, though frequently effective, often
Von Willebrand Disease 101

has only a transient effect. Serious bleeding by one of three mechanisms: autoimmune
should be treated with plasma‐derived clearance or inhibition of VWF, increased
FVIII/VWF concentrates. These patients shear‐induced proteolysis of VWF, or
may do well with desmopressin for the treat- increased binding of VWF to platelets
ment of minor bleeds or dental extractions. or other cell surfaces. Autoimmune causes
Type 2B VWD is often associated with of AVWS in children include postviral anti-
mild thrombocytopenia due to excessive body production similar to immune throm-
platelet‐binding to an abnormal VWF mole- bocytopenia purpura, lymphoproliferative
cule and rapid clearance. Stress can exacer- diseases, systemic lupus erythematosus, and
bate the thrombocytopenia. Desmopressin is other autoimmune disorders, and some can-
usually contraindicated due to the potential cers, most commonly Wilms tumor. It can
for worsening thrombocytopenia and failure also be seen related to certain drugs or
to demonstrate a therapeutic response. hypothyroidism. Pathological increases in
However, in mild bleeding, desmopressin fluid shear stress can occur with cardiovas-
may be effective. For internal bleeding or sur- cular lesions such as ventricular septal
gery, patients should receive FVIII/VWF defect and aortic stenosis, or with primary
concentrate. If profound thrombocytopenia pulmonary hypertension. This leads to
exists, concomitant administration of plate- increased destruction of VWF, particularly
lets may be necessary. the larger multimers; the multimer panel
Patients with 2M VWD will demonstrate often resembles that seen in Type 2A VWD.
an increase in VWF:Ag but not in VWF:RCo Its occurrence in Wilms tumor and other
in response to DDAVP, due to the defective malignancies is believed to be due to expres-
binding of their VWF to GPIb. These sion of platelet GPIb on the tumor cell
patients routinely require replacement with ­surface, leading to binding of VWF to the
FVIII/VWF concentrates for bleeding epi- tumor with subsequent degradation.
sodes or for surgery. Patients with Type 2N Hyaluronic acid secretion by nephroblas-
VWD similarly require factor replacement; toma cells is another potential mechanism
although they produce normal amounts of in Wilms tumor patients leading to
FVIII, it is destroyed prematurely due to the decreased efficacy of VWF. Patients may
inability of their VWF to bind to the FVIII present with classic signs and symptoms of
molecule and protect it. Replacement with VWD and their levels of VWF, VWF:Ag,
functional VWF will typically normalize and FVIII are typically quite low.
their FVIII levels even if the relative concen- Desmopressin may induce a transient rise
tration of FVIII in the product is low, since in VWF in AVWS. One small study demon-
they are able to produce normal amounts. strated that IVIG at a dose of 1 g/kg/day for
2 days was beneficial in improving VWF
­levels and decreasing clinical bleeding, but
Acquired von Willebrand this is only beneficial in immune‐mediated
syndrome AVWS. Plasmapheresis, corticosteroids, and
immunosuppressive agents have also been
Acquired von Willebrand syndrome successful in these patients. For serious
(AVWS) refers to defects in VWF concen- bleeding, treatment with factor concentrate
tration, structure, or function that are not (high VWF content) should be given. The
inherited directly but are consequences of antibodies typically disappear with control
other medical disorders. It is usually caused or resolution of the underlying disease.
102 Chapter 8

Other considerations Suggested reading

As a general precaution, all children with Castaman, G. and Linari, S. (2016). Human von
bleeding disorders should be immunized Willebrand factor/factor VIII concentrates in
with the hepatitis B vaccine, with boosters the management of pediatric patients with
given at appropriate time intervals, since von Willebrand disease/hemophilia A. Ther.
Clin. Risk Manag. 12: 1029–1037.
these children have a lifelong potential for
Leebeek, F.W. and Eikenboom, J.C. (2016). Von
receiving blood products. Patients and fami-
Willebrand’s disease. N. Eng. J. Med. 375:
lies should be counseled to avoid aspirin, 2067–2080.
nonsteroidal anti‐inflammatory drugs Sharma, R. and Flood, V.H. (2017). Advances in
(NSAIDs), and other platelet‐inhibiting the diagnosis and treatment of Von Willebrand
drugs. Also, it is generally recommended disease. Blood 130: 2386–2391.
that children wear medical bracelets and
have information readily available (at
school, home, doctor’s office) specifying the
diagnosis and treatment for bleeding.
9 Hemophilia

Hemophilia is defined as an inherited factor activity. Boys with severe hemophilia


­bleeding disorder caused by low levels, or bleed with minimal trauma and bleeding
absence, of a blood protein that is essential symptoms are usually apparent by the time
for clotting. The congenital hemophilias are the infant begins to crawl or walk, whereas
uncommon disorders, with a total incidence those with mild hemophilia usually bleed
of 10–20 per 100 000 births. The most com- only with significant trauma or surgery and
mon form is factor VIII (FVIII) deficiency may have a delayed diagnosis.
(hemophilia A or classic hemophilia), with Several other congenital factor deficien-
an estimated incidence of 1 in 5000 males. cies have been described as well, but are all
The second most common form is factor IX very uncommon. They are described in
(FIX) deficiency (hemophilia B), with an Table 9.1.
incidence of approximately 1 in 25 000 males. Afibrinogenemia is associated with fre-
Both the FVIII and FIX genes are carried on quent and serious bleeding. Spontaneous
the X chromosome, thus both hemophilia bleeding is rare if the fibrinogen level is
A and B are X‐linked recessive disorders. >50 mg/dl. The thrombin time (TT) is the
Approximately 80% of hemophilia patients most sensitive test to diagnose this disorder.
have hemophilia A and 20% have hemo- Hypoprothrombinemia is an autosomal
philia B. Both disorders are found through- recessive disorder associated with mild
out the world and do not appear to have any bleeding. All patients have some prothrom-
ethnic or racial predisposition. bin activity. Prothrombin deficiency also
Hemophilia A and hemophilia B are occurs in patients with vitamin K deficiency,
clinically indistinguishable, since both FVIII liver disease, and warfarin use. Factor V
and FIX are essential factors in the intrinsic (FV) deficiency is often characterized by
clotting pathway for activating factor X bleeding from mucous membranes includ-
(FX). Factor levels are measured in compar- ing epistaxis, menorrhagia, and bleeding
ison to a reference standard that is assumed after dental procedures. Congenital defi-
to have a factor level of 100% (1.0 IU/ml). In ciency of both FV and FVIII can occur as a
the normal individual, FVIII and FIX levels result of a mutation in the ERGIC‐53 gene,
range from 50–200% (0.50–2.0 IU/ml). which acts as a chaperone for intracellular
Disease severity is defined as severe (<1%), transport of these factors through the endo-
moderate (1–5%), and mild (6–50%). The plasmic reticulum. Patients with factor VII
frequency of bleeding symptoms generally deficiency who have levels less than 1% have
correlates well with the measured residual bleeding symptoms similar to that of classic

Handbook of Pediatric Hematology and Oncology – Children’s Hospital & Research Center Oakland,
Third Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
© 2021 John Wiley & Sons Ltd. Published 2021 by John Wiley & Sons Ltd.
104 Chapter 9

Table 9.1 Rare coagulation factor deficiencies.

Deficiency Estimated Source of replacement Biological Gene on


prevalence 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 or 24 h 13
plasma‐derived
concentrate
Factor XI 1/1 000 000 FFP 40 h 4
Factor XIII 1/2 000 000 Recombinant or 3–5 d 1 and 6
plasma‐derived
concentrate

Abbreviations: PCC, prothrombin complex concentrate; FFP, fresh frozen plasma; rFVIIa,
recombinant factor VII.

hemophilia with hemarthrosis, intramus- with α2‐antiplasmin deficiency are unable


cular hemorrhage, and intracranial hemor- to effectively inhibit plasmin, and therefore
rhage (ICH). Heterozygotes are typically lyse clots abnormally quickly leading to
asymptomatic. Two‐thirds of patients with abnormal bleeding. As clots are formed nor-
factor X (FX) deficiency have hemarthroses mally, all the usual screening coagulation
and hematomas. The proportion of patients tests are normal except for the euglobulin
with FX deficiency who require treatment is and urea clot lysis tests. Factor XII (FXII)
higher than that of the other rare coagula- deficiency, prekallikrein deficiency, and
tion deficiencies. Factor XI (FXI) deficiency high‐molecular‐weight kininogen deficiency
is characterized by mucocutaneous and all cause prolongation of the PTT but are
­genitourinary tract bleeding; the likelihood not associated with clinical hemorrhage.
of bleeding does not correlate well with
measured FXI levels. Only individuals with
factor XIII (FXIII) deficiency whose levels Clinical presentation
are <1% are symptomatic. Bleeding mani-
festations include soft‐tissue hemorrhages, Boys with hemophilia may develop symp-
hemarthroses, and hematomas, and patients toms very early in life, experiencing a
undergoing surgery or trauma may experi- ­hemorrhage after circumcision or with sep-
ence poor wound healing. aration of the umbilical cord. However, up
Alpha2‐antiplasmin deficiency, factor XII to 50% of affected male infants have no dif-
deficiency, prekallikrein (Fletcher’s factor) ficulty during the neonatal period, and a
deficiency, and high‐molecular‐weight kini- negative history of bleeding after circum-
nogen (Fitzgerald’s factor) deficiency have cision does not rule out the diagnosis of
also been identified using appropriate clot- hemophilia. Mild hemophilia may go unsus-
ting measurements. Alpha2‐antiplasmin is pected for years until the patient experi-
the primary inhibitor of plasmin, a key com- ences trauma or has a surgical procedure.
ponent of the fibrinolytic pathway. Patients Conversely, patients with severe hemophilia
Hemophilia 105

often experience spontaneous hemorrhages, affected as the infant begins crawling, followed
both externally and internally, into the by shoulders, wrists, hips, knees, and ankles.
head, joints, muscles, and retroperitoneum. The hands and spine are rarely involved. The
Bleeding from the mouth or frenulum often initial signs of a hemarthrosis are vague and
occurs during infancy, and minimal lacera- difficult to detect, especially in the nonverbal
tions may cause very prolonged bleeding. infant. Early, aggressive treatment of an acute
Intramuscular injections (as with immuni- hemarthrosis is recommended to relieve
zations) often result in large hematomas. symptoms and hopefully prevent recurrent
Joint hematomas (hemarthroses) can result bleeding into the same joint, setting up the
in secondary joint degeneration. Life‐ situation of a ­“target” joint.
threatening blood loss can occur with intra- Bleeding into muscle is characterized by
muscular bleeds. ICH occurs in 2–14% of pain and limitation of mobility. They can be
hemophilia patients and is associated with a difficult to evaluate, particularly if they
4–34% mortality. Although ICH is much occur in deep muscles such as the iliopsoas
more common in severe hemophilia, it has muscle (see the following text). They can
been reported in patients with mild hemo- result in long‐term complications such as
philia as well. permanent muscle contractures. It is impor-
ICH is the most serious complication in tant to exclude any potential neurovascular
hemophilia. A history of trauma is elicited compromise that can result from compres-
in only 20–25% of patients with central sion of 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 intrac- sequences and to the large volume of blood
erebral, subdural, subarachnoid, or epidural. loss that can occur acutely into this large
Patients who survive these episodes fre- muscle bed and into the retroperitoneal
quently experience complications such as space. The presenting signs of groin or lower
mental retardation, seizure disorders, or abdominal/upper thigh pain can be difficult
motor impairment. Any history of head to differentiate from a hip bleed. With an ili-
trauma should be considered emergent. A opsoas bleed, examination reveals inability
nonfocal neurologic examination does not to extend the hip, with normal internal and
exclude a diagnosis of an intracranial bleed. external hip rotation. Radiologic confirma-
Any suspicion of a significant intracranial tion by CT scan is recommended.
injury should prompt a computerized Soft tissue hemorrhages occur very com-
tomography (CT) study of the head and monly and do not require therapy unless
close neurological observation. Immediate their location is near vital structures. For
treatment to achieve a factor level of instance, a retropharyngeal bleed with
80–100% should be provided and main- potential airway compromise should be
tained until an intracranial bleed can be aggressively treated. Vigorous examination
fully excluded. An intraspinal hemorrhage of the oropharynx with a tongue blade or
should be excluded with any back trauma or other manipulations should be avoided to
symptoms of a peripheral neuropathy. A prevent injury with subsequent hemor-
lumbar puncture should never be performed rhage. A “straddle” injury resulting in a
in a hemophilia patient without factor ther- hematoma of the perineal/perirectal area
apy to avoid this complication. should be closely evaluated for neurologic
Hemarthroses appear in young children as compromise as evidenced by either bladder
they become mobile. The elbows are often first or bowel dysfunction.
106 Chapter 9

About 70% of hemophilia patients will bleeding. A prolonged PTT in conjunction


have at least one episode of hematuria. with a normal PT and platelet count is
Usually, these episodes are painless, thus if ­suggestive of the diagnosis; this can be con-
there is severe pain associated with hematu- firmed by specifically measuring levels of
ria, a thrombus in the renal pelvis should FVIII and FIX.
be excluded. Increased fluid intake and bed Newborn males with a possible or con-
rest is recommended. Factor replacement firmed diagnosis of hemophilia should be
may be warranted with persistent symp- carefully evaluated for an intracranial hem-
toms. Treatment with antifibrinolytic drugs orrhage, especially if delivered via assisted
is contraindicated given the risk of upper vaginal delivery (vacuum extraction or
urinary tract obstruction caused by clot. forceps). However, infants delivered via
­
Epistaxis occurs in hemophilia patients, cesarean section are also at risk for severe
most commonly in those with severe dis- bleeding. The initial PTT can be performed
ease. Gastrointestinal bleeding can also on cord blood, but it must be remembered
occur but is atypical; an underlying gastro- that the upper limit of normal for newborns
intestinal lesion should be excluded. is longer than for adults. The normal range
Intramural bleeding into the bowel wall can for FVIII levels in the newborn is the same
occur, with signs of severe pain and possible as for adults, but FIX levels are lower until
intestinal obstruction. Appropriate therapy the concentrations of factors produced by
may avoid an unnecessary laparotomy. the liver mature to adult levels (approxi-
Prolonged gingival oozing is common mately 6 months of age). Therefore, the
after shedding of deciduous teeth, eruption diagnosis of hemophilia B may be more dif-
of new teeth, or instrumentation. Treatment ficult initially. Confirmatory studies should
with an antifibrinolytic medication (i.e., be performed on a sample obtained by veni-
tranexamic or aminocaproic acid) is recom- puncture if a cord blood sample was ini-
mended for prolonged symptoms. Routine tially obtained. Arterial as well as femoral
dental care is particularly important in the or jugular venous sites to obtain plasma
patient with a bleeding disorder to prevent samples should be avoided if possible.
the need for restorative care in the future Prenatal testing for both conditions is avail-
where extensive factor replacement therapy able to pregnant women who are known
is often indicated. Regional block anesthesia carriers.
is discouraged, even with factor coverage,
given the risk of hemorrhage that can lead to
nerve damage and extension into the neck Treatment
causing airway compromise.
After the newborn period, affected infants
rarely require treatment until they become
Diagnosis more mobile with increasing age. Treatment
recommendations for the hemophilia
The diagnosis of FVIII deficiency should be patient depend on several factors: the type
suspected in males presenting with a charac- and severity of disease (mild, moderate, or
teristic bleeding history, especially if there is severe; FVIII or FIX deficiency); the site of
a family history of males with abnormal the bleed (proximity to vital structures) with
bleeding. However, 30% of patients with consideration of long‐term debilitating con-
hemophilia A and B have sporadic muta- sequences; and therapy response (factor
tions and no family history of ­ abnormal recovery/survival and inhibitor status).
Hemophilia 107

Table 9.2 Guidelines for factor replacement in hemophilia A and B.

Site of hemorrhage Optimal factor Dose (units/kg body Minimum


level (%) weight) duration of
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.

Treatment of patients with hemophilia and the challenge of adequate vascular


and bleeding is by replacement of factor in access, particularly in young patients.
a concentrated form. Many high‐purity When providing on‐demand therapy, the
plasma‐derived preparations are currently duration, frequency, and doses of factor pro-
available, as are recombinant FVIII and FIX vided depend on the severity of the hemo-
products. World Federation of Hemophilia philia and bleeding episode. Factor vials
guidelines do not recommend one type of vary in the number of units of factor activity
product versus another and each has their they contain. The required factor dose
pros and cons. Plasma‐derived factor should be calculated, and vials should then
appears to have less development of inhibi- be selected that come closest to the calcu-
tor than recombinant products, but requires lated target dose. Doses are calculated using
a larger volume than recombinant products the following formula:
and has a theoretical risk of acquiring new
infections not currently screened. Dose units of FVIII = Weight kg
There are two treatment strategies cur- desired % rise of FVIIII level 0.5
rently used for patients with hemophilia: (i)
episodic or on‐demand treatment, provid- For FIX, multiply result by 2, for recom-
ing treatment at the time a bleeding episode binant FIX, multiply by 2.4 (due to lower
occurs; and (ii) prophylactic therapy, pro- recovery rate of unclear etiology).
viding periodic infusion of factor replace- Round up to the whole vial, since factor
ment to prevent such bleeding episodes. A is expensive and not uniformly dispersed in
number of single institutional case series suspension. Table 9.2 outlines the dose and
have demonstrated the superiority of proph- duration of therapy for commonly encoun-
ylaxis regimens in the prevention of joint tered bleeding episodes in hemophilia
disease, but widespread adoption of this patients. More serious bleeding episodes
strategy for all patients has been limited by should be treated using doses at the higher
the high cost of factor replacement therapy end of the range.
108 Chapter 9

Other important interventions for a


Other considerations
hemarthrosis include initial immobilization
and application of ice to the area or use of a
In addition to aggressive factor replacement
Cryo/Cuff. For particularly critical joints
when bleeding has occurred and the use of
such as the shoulder or hip, where long‐
physical therapy programs to prevent or
term consequences of pressure‐induced
minimize chronic joint disease, comprehen-
avascular necrosis can be debilitating, a
sive medical care must be provided to all
longer treatment course is recommended.
patients with hemophilia. All immunizations
Iliopsoas bleeds should be treated as retrop-
including hepatitis B vaccine should be
eritoneal rather than as muscle bleeds.
given on schedule (subcutaneous rather than
intramuscular). Regular dental care is essen-
tial to prevent excessive decay and gingival
Inhibitors
disease that may predispose to complications
and need for more aggressive surgical man-
The development of an inhibitor, an antibody
agement. Psychological support is essential
that inactivates the coagulant function of
to assist patients and families cope with the
replacement factor, is one of the most serious
emotional and social burden imposed by the
complications of hemophilia treatment. FVIII
disease. A comprehensive program of patient
inhibitors develop in 20–30% of boys with
and family education will prevent complica-
severe hemophilia A, whereas FIX inhibitors
tions caused by failure to recognize or under-
develop in 3–5% with severe hemophilia B.
stand warning signs of hemorrhage as well as
These usually develop within the first 50
empower family members and eventually
exposures to factor replacement. Risk factors
the patient to take an active role in the man-
for inhibitor development include the nature
agement of the disease. Education regarding
of the underlying mutation, with those lead-
in‐home treatment is vital and should be
ing to substantial loss of coding information
used whenever possible given the benefit
representing greater risk, high‐intensity prod-
of prophylactic factor administration for
uct exposure, CNS bleeding, and African‐
patients with severe disease as well as in the
American race. Inhibitors are quantitated by
rapid recognition and treatment of bleeding,
Bethesda titers: one Bethesda unit (BU) is the
especially hemarthroses, which directly
amount of inhibitor that inactivates 50% of
relates to ­minimizing long‐term complica-
FVIII function in a one‐stage clotting assay.
tions of the disease.
Low‐titer inhibitors (<5 BU) can be managed
by using larger doses of factor replacement;
higher‐titer inhibitors require the use of alter-
native hemostatic agents. Alternative agents Novel agents
include prothrombin complex concentrates
(PCCs), activated PCCs (aPCCs), recombi- Over the last several years, multiple
nant factor VIIa (rFVIIa), and a newer extended half‐life agents for both factor VIII
bypassing agent emicizumab (see in the fol- and factor IX deficiencies have been studied
lowing text). Immune tolerance induction and approved. These products have been
with high, frequent doses of FVIII or FIX has most successful for factor IX deficiency,
also been successful. Management of patients increasing the half‐life threefold to sixfold
with inhibitors is difficult and complex and and thus decreasing prophylactic replace-
should be performed only by experienced ment therapy to once weekly (from twice
hematologists. weekly) in young patients and potentially
Hemophilia 109

once every other week in the well‐controlled an AAV‐induced liver‐induced immune


older patient. Due to the interaction with response of unclear etiology, those with anti-
von Willebrand factor, the half‐life exten- bodies to AAV above the cutoff, and patients
sion of factor VIII products is significantly with factor VIII or IX inhibitors.
less, only 1.5‐fold to twofold, thus decreas-
ing the need for prophylactic dosing from
thrice weekly to twice weekly. Though a Case study for review
smaller effect, for a young patient this can
still have a significant benefit for both You have been following a patient with
patient and family. severe hemophilia A since soon after birth
Emicizumab, a bispecific monoclonal and his mother has questions about plans
antibody, which bypasses factor VIII and for an upcoming second pregnancy. She is
instead engages factor IX and factor X, has worried about the chance of having another
shown benefit in studies for patients with child with hemophilia.
factor VIII inhibitors. More recent studies a. What are some initial important
have shown the benefit in pediatric and points that you should discuss?
adult patients with factor VIII deficiency It is first important to ascertain if there is a
without inhibitors. As a bypassing agent, family history (i.e., maternal uncles, other
emicizumab does not lead to inhibitor males on the maternal side) making this an
development. Additionally, it has the benefit inherited trait rather than a sporadic muta-
of being given subcutaneously and with a tion which can occur in up to 30% of
long half‐life allowing for dosing once every patients. Even without a relevant (or known)
4 weeks (after initial 4 weekly loading family history, this is more likely to be
doses). Emicizumab was FDA approved for inherited, and therefore genetic testing can
all patients with hemophilia A, with or with- be performed to determine if the mother is
out inhibitors, in 2018. an obligate carrier especially if there is no
known family history.
b. Assuming the mother is a carrier,
Gene therapy what are the next important discussion
points?
Gene therapy has been studied for decades in Given the mother is a carrier, she will
hemophilia and remains a long‐term cura- have a 50% chance of passing this on to
tive opportunity that is continuing to be either a symptomatic male or carrier female
refined. Most recently, adeno‐associated (unless symptomatic due to lyonization
viral vectors (AAVs) have been utilized, which is rare). It should be discussed that
though there remains potential to utilize prenatal diagnosis can be made through
gene editing in future through zinc‐finger chorionic villus sampling (CVS) or amnio-
nucleases (ZFN) or clustered regularly inter- centesis, if desired (the risks of which
spaced short palindromic repeat (CRISPR). should also be discussed). If the family is
Current challenges with gene therapy include utilizing in vitro fertilization, prenatal
differing manufacturing processes and a genetic diagnosis (PGD) can also be
wide range of AAV doses leading to variable ­utilized to ensure a an unaffected male or a
target clotting factory activity. Additionally, non‐carrier female.
there have been many patient groups c. Based on this information, the
excluded to date including pediatric patients, mother decides to move forward with a
those with underlying hepatitis secondary to second pregnancy without any further
110 Chapter 9

testing. What information is it important should be referred to hematology, although


to pass on to her and her OB/Gyn? routine factor prophylaxis is not typically
Given the potential for a (male) child with required at this age.
hemophilia, it is vital that the treating
team know of this risk and utilize the least
invasive methods during the delivery.
­ Suggested reading
Spontaneous vaginal delivery is not con-
traindicated but vacuum suction and for- Doshi, B.S. and Arruda, V.R. (2018). Gene ther-
ceps should be avoided. After birth, the apy for hemophilia: what does the future
umbilical cord blood can be tested to see if hold? Ther. Adv. Hematol. 9: 273–293.
there is prolongation of the PTT, knowing Graf, L. (2018). Extended half‐life factor VIII
the normal range for a newborn is different and factor IX preparations. Transfus. Med.
(i.e., more prolonged) than for an adult. Hemother. 45: 86–91.
Arterial, femoral, and jugular punctures Pipe, S.W., Shima, M., Lehle, M. et al. (2019).
should be avoided in the new baby, and Efficacy, safety, and pharmacokinetics of emici-
zumab prophylaxis given every 4 weeks in
intramuscular injection should be avoided
people with haemophilia A (HAVEN 4): a mul-
until a diagnosis is made. Head ultrasound ticenter, open‐label, non‐randomised phase 3
should be done immediately after birth study. Lancet Hematol. 6: PE295–PE305.
unless hemophilia is definitively ruled out. Srivastava, A., Brewer, A.K., Mauser‐Bunschoten,
Similarly, circumcision should be delayed E.P. et al. (2013). Guidelines for the management
if the diagnosis is unclear of if the child has of hemophilia. World Federation of Hemophilia
hemophilia. The child with hemophilia Guidelines. Haemophilia 19: e1–e47.
10 Thrombophilia

Virchow’s triad describes risk factors for


Evaluation of thrombosis
thrombus (blood clot) formation: (i) venous
stasis; (ii) endothelial injury; and (iii) 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 thrombo-
acquired risk factor for VTE is the presence
sis secondary to aberrations in the coagula-
of a central venous catheter (i.e., endothelial
tion system. Thrombophilia is becoming
injury). Other acquired risk factors include
increasingly recognized in pediatric patients
trauma, surgery, infection, nephrotic syn-
and can refer to multiple potential condi-
drome, inflammatory syndromes, diabetes,
tions: (i) spontaneous thrombus formation;
complex congenital heart disease, liver dis-
(ii) recurrent thrombosis; (iii) thrombosis
ease, and malignancy (e.g., acute lympho-
out of proportion to the underlying risk fac-
blastic leukemia in association with the use
tor; and (iv) thrombosis at a young age.
of asparaginase or solid tumors with tumor
Optimal prevention and treatment in
thrombus). Asymptomatic patients may be
pediatric patients with thrombosis differs
found to have an incidental clot on imaging,
from adult patients for several reasons.
or the patient may be symptomatic, most
These include physiologic age‐dependent
often with swelling, pain, or erythema in an
differences in the hemostatic system that
extremity. Venous ultrasonography with
influence the risk for venous thromboem-
Doppler flow remains the cornerstone of
bolism (VTE), differing underlying etiolo-
evaluation for thrombosis, as in adults. Lack
gies and location of clots, and differing
of venous compression or Doppler flow is
responses to antithrombotic agents. Little
diagnostic for thrombus. This imaging
evidence exists as how best to manage pedi-
modality is limited to the extremities, neck,
atric patients with thrombosis, both from a
distal subclavian veins, and, potentially, dis-
diagnostic and from a treatment standpoint.
tal iliac veins. Echocardiography can be uti-
Much of the currently utilized data are based
lized to evaluate for atrial clots and the
on adult studies, and here we summarize the
proximal superior and inferior vena cava.
generally accepted recommendations in lieu
Imaging of the abdomen and pelvis as well
of evidence‐based pediatric guidelines (see
as the upper venous system requires CT or
Figure 10.1).

Handbook of Pediatric Hematology and Oncology – Children’s Hospital & Research Center Oakland,
Third Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
© 2021 John Wiley & Sons Ltd. Published 2021 by John Wiley & Sons Ltd.
112 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 embolism,
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)
Antiphospholipid 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)
thrombophilia Yes

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, pro-
thrombin time; PTT, partial thromboplastin time; CBC, complete blood count; CNS, central nervous
system; OCP, oral contraceptive pill; ATIII, antithrombin III; ALL, acute lymphoblastic leukemia).

magnetic resonance venography (MRV). patient with a known acquired risk factor
Similarly, sinus venous thrombosis must be and secondary thrombosis, it is unclear that
evaluated by magnetic resonance imaging/ testing for thrombophilia will aid in the
arteriography (MRI/MRA). Pulmonary management of the patient (e.g., asympto-
emboli or thrombosis can be identified with matic catheter‐related thrombosis). Patients
a helical CT scan. might have a single‐trait thrombophilia on
testing, but this alone may not contribute
significantly to thrombus development.
Testing for thrombophilia Patients with a consumptive process (e.g.,
disseminated intravascular coagulation and
Although often undertaken, clear guidelines sepsis) and secondary thrombosis such as
are lacking as to the utility of testing for an with deficiencies in protein C and S and
underlying hypercoagulable state. In the antithrombin III (ATIII) could benefit from
Thrombophilia 113

Table 10.1 Primary thrombophilia traits.

Thrombophilia trait Description

Factor V Leiden G1691A polymorphism, present in ~5% of Caucasian population,


leads to inherited activated protein C resistance
Prothrombin gene PT G20210A polymorphism, present in ~2% of Caucasian population,
mutation leads to elevated prothrombin levels
Antithrombin III Can be seen in consumptive process (sepsis, DIC, active clot), liver
deficiency disease, heparin therapy, complex congenital heart disease,
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 against cell membrane phospholipid, called lupus
antibodies anticoagulant, paradoxically leads to elevated PTT without correction
on 1:1 mix. Secondary confirmatory testing includes anticardiolipin
antibodies and β2 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.

testing and factor replacement if found to be counseling. The family must be advised that
deficient in the acute illness. Patients with a positive screen for a single thrombophilia
deficiencies should be re‐tested in the steady trait will not result in a change in manage-
state to determine if the patient is indeed ment and may lead to unnecessary anxiety.
baseline deficient (heterozygous state) or Similarly, negative testing may give false
was low due to consumption. For patients reassurance and does not completely abro-
with spontaneous thrombosis, often found gate the potential for thrombosis. The pri-
in adolescents (e.g., adolescent female start- mary thrombophilia traits are listed in
ing oral contraceptives) and in neonates Table 10.1.
(secondary to stroke or sinus venous throm-
bosis), testing should be considered. For
patients with a symptomatic catheter‐related Management of thrombosis
thrombosis or for an asymptomatic patient
with a positive family history, the decision to Management of pediatric thrombosis is
test for thrombophilia must be made on an again based on adult studies and expert
individual basis after appropriate family guidelines. Removal of the precipitating
114 Chapter 10

agent should be done as possible in cases TPA as suggested by Manco‐Johnson (see


with an acquired risk factor. Treatment of suggested further reading) at a dose of 0.03–
thrombosis is undertaken to prevent clot 0.06 mg/kg/h with a maximum of 2 mg/h for
propagation, pulmonary embolus, and post- 24–48 hours to decrease the risk of signifi-
thrombotic syndrome (PTS). Although cant hemorrhage that is seen with bolus
reported frequently in adults, PTS is poorly TPA, unless there is a life‐threatening clot
understood in children but can potentially (e.g., massive pulmonary embolism).
lead to chronic pain, swelling, skin changes, Arterial clots may benefit from a higher sys-
and collateral venous formation. Neonatal temic dose for a shorter time period. Also, if
thromboembolism guidelines are generally possible, TPA should be instilled distal to
similar to pediatric guidelines, though a the clot in order to decrease first‐pass liver
more conservative approach may be consid- metabolism, although this is mitigated with
ered given the potential increased risk of the use of systemic TPA, as it is expected
hemorrhage in this population. The deci- that much of the TPA will bypass the
sion to initially utilize a thrombolytic (tissue obstruction through collateral circulation.
plasminogen activator, TPA) versus an anti- Patients receiving systemic low‐dose TPA
coagulant (unfractionated heparin, UH, therapy should have close monitoring of
low‐molecular‐weight heparin, LMWH, or coagulation studies (prothrombin time
a direct oral anticoagulant, DOAC) must [PT], partial thromboplastin time [PTT],
be made in consultation with a pediatric and fibrinogen), platelets, and plasminogen
hematologist. In rare cases, thrombectomy to ensure hemostatic and fibrinolytic poten-
or thrombolysis by intervention radiology tial and the need for potential repletion with
should be considered. fresh frozen plasma (FFP).

Thrombolysis Anticoagulation
In general, thrombolysis is not routinely If TPA is thought to pose significant risk, or
recommended except in situations with
­ if the patient has a nonocclusive thrombus
hemodynamic compromise or which are or transient risk factors, UH or LMWH (or,
life‐threatening. Data are lacking in pediat- potentially, a DOAC) may be started. UH is
ric patients and expert consensus guidelines a natural anticoagulant and works by com-
have very low certainty based on the existing plexing to the physiologic inhibitor ATIII,
evidence (see suggested further reading). In accelerating the inhibition of thrombin and
consultation with hematology, a thrombo- other coagulant proteins. It is therefore
lytic can be considered in patients with an important to ensure adequate levels of ATIII
occlusive thrombus without significant risk when administering heparin. Many poten-
factors for bleeding (recent surgery, central tial issues are present with UH usage in chil-
nervous system hemorrhage). Patients with dren: (i) rapid clearance; (ii) low ATIII levels
a long‐standing occlusive thrombus (i.e., in the first few months of life; (iii) greater
>14 days) or without evidence of improve- variability in dosing compared to adults;
ment on imaging studies after 24–48 hours and (iv) lack of evidence assessing the opti-
of TPA therapy may benefit from thrombec- mal target PTT range for the prevention and
tomy or thrombolysis by interventional treatment of VTE. The patient must also be
radiology, though this is no longer routinely monitored for the development of heparin
recommended. Although not evidence‐based, antibodies that can most notably lead to
we typically treat with low‐dose systemic heparin‐induced thrombocytopenia (HIT).
Thrombophilia 115

Long‐term usage can also lead to osteoporo- and is recommended at a dose of 0.1 mg/kg
sis. LMWH is more widely utilized now due SC once daily. Specific fondaparinux levels
to a narrower dosing range and wide thera- should be obtained different from fraction-
peutic window secondary to direct targeting ated anti‐Xa levels and dosing adjusted
of factor Xa as well as reduced incidence of accordingly.
HIT and osteoporosis. Concomitant use of TPA and UH or
In general, we utilize LMWH unless LMWH may also be beneficial in clot lysis
there is potential need for immediate rever- without significantly increasing hemor-
sal of anticoagulation. Although protamine rhagic risk, although again this recommen-
can be utilized with LMWH, the reversal dation is not evidence‐based in pediatric
effect is not complete (thought to be about patients and must be determined on a per
two‐thirds effective), and therefore a risk patient basis in consultation with a pediat-
of bleeding can still be present if, for ric hematologist, weighing the potential
instance, surgical intervention is required risks of hemorrhage and potential benefits
emergently. In these cases, UH should be of clot lysis (and is not recommended in
utilized due to its short half‐life and ability combination in the most recent consensus
to be fully reversed. UH is generally given guidelines). Therapeutic efficacy can sim-
at a loading dose of 75 U/kg followed by a ply be followed with repeat imaging,
maintenance dose of 20 U/kg/h with a goal although this can be difficult with central
PTT of two to three times the upper limit venous clots that may require multiple CT
of the reference range or, preferably, unfrac- or MRI with resultant increased radiation
tionated anti‐Xa levels of 0.3–0.7 U/ml, if exposure and cost, respectively. Other
available in a timely manner. Currently, the potential markers to follow for clot lysis
only LMWH approved by the FDA for include d‐dimers or fibrin‐split products
pediatric patients is enoxaparin, with a which should increase with lysis, although
half‐life of 4 hours and subcutaneous (SC) marker elevation may be confounded by
dosing (though extended half‐life daily SC continuing inflammation.
fondaparinux is being increasingly uti-
lized). For treatment of thrombosis, enoxa- Duration of therapy
parin must be given every 12 hours, usually Duration of therapy depends on multiple
starting at 1.0–1.25 mg/kg (see Formulary factors including the time to removal of
and Manco‐Johnson for more specific inciting agents such as central venous cath-
dosing guidelines). Treatment effective- eters, presence of thrombophilia traits, and
ness is followed by fractionated anti‐Xa time to clot resolution. Patients initially
levels, with goal levels of 0.5–1.0 U/ml treated with TPA or UH therapy should be
(checked 4 hours after the second or subse- transitioned to LMWH or warfarin therapy
quent dose). For prophylaxis, 0.5 mg/kg is for long‐term maintenance. Due to lack of
typically given twice daily. Anti‐Xa levels pediatric data, DOACs are not currently
do not need to be checked with prophylac- recommended in consensus guidelines but
tic dosing. In practice, some hematologists are being increasingly utilized. Because of
utilize 1 mg/kg once daily for prophylaxis, the time required to reach an appropriate
although available data suggest this may therapeutic level, warfarin therapy (if uti-
undertreat a group of patients, especially lized) should begin at least 48 hours prior
those <5 years of age. Fondaparinux obvi- to discontinuation of heparin therapy.
ates the need for more than daily dosing Maintenance with LMWH is frequently
116 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 analog 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
Apixaban Like rivaroxaban except twice daily dosing
Endoxaban Can be used in adults at a reduced dose with CrCl 15–50 ml/
min, though has unclear safety in CrCl >95 ml/min due to
increased stroke compared with 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
fibrillation, in the United Kingdom as an alternative to
warfarin for thromboprophylaxis

Abbreviations: ATIII, antithrombin III; CrCl, creatinine clearance; HIT, heparin‐induced


thrombocytopenia; SC, subcutaneous; IV, intravenous.

used due to the difficulty in maintaining a


New agents
therapeutic international normalized ratio
(INR) with warfarin therapy.
As described, multiple new agents will very
For patients with clot resolution, antico-
likely be FDA approved in the near future
agulation should be continued at prophylac-
for pediatric patients and are summarized in
tic dosing for 6 weeks to 3 months after
Table 10.2 (many of which are increasingly
removal of the inciting agent (e.g., central
being used off‐label).
venous catheter) due to the likelihood of
continued endothelial injury. For patients
without clot resolution (potentially after
ineffective interventional thrombectomy Case study for review
and/or thrombolysis), anticoagulation
should be continued for 6–12 months to You are seeing a 17-year-old female in clinic
potentially decrease or resolve the clot and who is deciding on whether to start oral
prevent the development of PTS. Patients contraceptive pills (OCPs). She has an aunt
that have a spontaneous or recurrent that had a “blood clot” when she was in her
thrombus and are found to have multitrait fifties. Your patient does not know any fur-
thrombophilia (≥3 traits) or antiphospho- ther details about the incident but does
lipid syndrome should continue lifelong know about the potential increased risk of
anticoagulation. blood clots with OCPs.
Thrombophilia 117

a. What advice would you give the patient? an underlying risk factor in the aunt’s
All patients who are starting OCPs should thromboembolism, testing her and/or your
be made aware of the increased risk of blood patient would not be advised. In the situa-
clots with estrogen-containing contracep- tion where thrombophilia testing of your
tives (about a three-times increased risk). patient is not advisable (the most likely
This risk is thought to be about 2–3 per 10 scenario in general), you should counsel
000 per year on OCPs, or about 0.02–0.03% your patient closely on the risks and benefits
incidences per year (baseline risk being of OCPs and the potential lower-risk alter-
approximately 0.008% per year). Single-trait native therapies such as progesterone-only
thrombophilias, most commonly factor V preparations (e.g., Depo-Provera).
Leiden mutation, increase this risk signifi-
cantly. Specifically, for factor V Leiden, the Suggested reading
risk for a heterozygous carrier increases the
risk approximately 30 times to a yearly inci- Manco‐Johnson, M.J. (2006). How I treat venous
dence of 0.6–0.9%. This incidence is also thromboembolism in children. Blood 107:
highest in the first year of OCP usage. 21–29.
b. She asks about testing for clotting risk Monagle, P., AKC, C., Goldenberg, N.A. et al.
factors, what do you advise her? (2012). Antithrombotic therapy in neonates
Thrombophilia testing can be considered on and children. Antithrombotic Therapy and
an individual basis for the patient wishing to Prevention of Thrombosis, 9th ed.: American
start OCPs who has a family member with a College of Chest Physicians Evidence‐Based
Clinical Practice Guidelines. Chest 141:
known thrombophilia. Additionally, the
e737s–e801s.
patient should be offered alternative lower
Monagle, P., Cuello, C.A., Augustine, C. et al.
risk forms of contraception. In this case, the (2018). American Society of Hematology
practitioner should try to get more informa- 2018 Guidelines for management of venous
tion regarding the episode of thromboembo- thromboembolism: treatment of pediatric
lism in the aunt. Were there acquired risk venous thromboembolism. Blood Adv. 2:
factors such as trauma, surgery, or immobil- 3292–3316.
ity? Was the clot thought to be spontaneous? Monagle, P., AWA, L., Thelen, K. et al. (2019;
If so, was the aunt tested for thrombophilia? https: //doi. org/1 0 . 1 0 1 6 /S2 3 5 2 ‐3 0 2 6 ) .
Current recommendations would not Bodyweight‐adjusted rivaroxaban for chil-
advise testing your patient without further dren with venous thromboembolism
(EINSTEIN‐Jr): results from three multi-
information. If the aunt had a spontaneous
center, single‐arm, phase 2 studies. Lancet
thrombus, one could advise that she should Hematol. 10: 30161–30169.
be tested for thrombophilia, and if found Raffini, L. (2008). Thrombophilia in children:
to be positive your patient could be tested who to test, how, when, and why? Hematology
as well. If it was determined that there was Am. Soc. Hematol. Educ. Program: 1: 228–235.
11 The Neutropenic Child

Neutrophils are a key component in the intervention. However, this definition fails to
defense against infection. They contain take into account important variations in
toxic cytoplasmic granules that, following normal neutrophil number related to age
ingestion of infecting bacteria and fungi, are and ethnicity. Greater than 10% of young
released into the phagocytic vacuole, Arabic and African‐American children will
destroying them. Once released from the have an ANC between 1 and 1.5 × 109/l, and
bone marrow, they circulate in the blood for this level should be considered normal for
a brief time (4–6 hours) before leaving the these groups. This “mild neutropenia” also
circulation and entering the tissue where, in extends into adulthood. Neutropenia is com­
response to the presence of infection, they mon in premature or small for gestational
act to control the infection while sending age (SGA) newborns; 5–10% of these infants
chemotactic signals to recruit additional will have an ANC < 1 × 109/l. Also, the nor­
neutrophils to the area and stimulate the mal range for ANC extends down to 1 × 109/l
accelerated production of neutrophils in in infants between 2 weeks and 6 months of
the bone marrow. While the presence of age and should not be considered abnormal.
adequate neutrophil numbers in the tissue is
the best predictor of the patient’s ability to
fight infection, there is no easy clinical Risk assessment
method to determine the number of tissue
neutrophils, so the number present in the The first question when evaluating a child
blood is used as a surrogate marker. with neutropenia is: “What is the risk that
Neutropenia has traditionally been this patient has or will develop a life‐threat­
defined as a decrease in the absolute neutro­ ening infection?” This is particularly perti­
phil count (ANC) to <1.5 × 109/l. The ANC is nent if the patient presents with fever. The
calculated by multiplying the white blood susceptibility to bacterial infection in neu­
cell (WBC) count by the total percentage of tropenic patients is quite variable and
segmented (mature) neutrophils plus bands. depends on a number of factors. The first is
Mild neutropenia is defined as an ANC of the severity of neutropenia, as described in
1–1.5 × 109/l, moderate neutropenia is an the preceding text. Patients with mild neu­
ANC of 0.5–1 × 109/l, and severe neutropenia tropenia have minimal to no increased risk
is an ANC below 0.5 × 109/l. This division is of infection, those with moderate neutrope­
useful for determining the individual’s risk nia have a mildly increased risk of frequent
for infection and the urgency of medical or severe infections, and those with severe

Handbook of Pediatric Hematology and Oncology – Children’s Hospital & Research Center Oakland,
Third Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
© 2021 John Wiley & Sons Ltd. Published 2021 by John Wiley & Sons Ltd.
120 Chapter 11

neutropenia are highly susceptible to bacte­


Etiology of neutropenia
rial infection. However, patients who are
neutropenic as a result of cytotoxic therapy
A large variety of conditions can lead to
(e.g., chemotherapy, radiation) may be at an
neutropenia in childhood. They can be
increased risk of infection because of the rate
classified as being either acquired causes or
of decline of the neutrophil count or other
inherited causes. Of these, acquired causes
immunosuppressive effects of chemother­
are much more frequent. These are
apy, even before the ANC falls below
presented in Table 11.1.
0.5 × 109/l. Another factor is the cause of the
Viral infection is the most common cause
neutropenia, if known. Patients with neutro­
of mild–moderate neutropenia. Transient
penia caused by intrinsic bone marrow fail­
marrow suppression is seen in children with
ure or hypoplasia have a greater risk of
EBV, respiratory syncytial virus (RSV), influ­
infection than those whose neutropenia is
enza A and B, hepatitis, HHV6, varicella,
due to factors extrinsic to the marrow such
rubella, and rubeola. This typically lasts for
as excessive neutrophil destruction or
3–8 days. Neutropenia can also be seen in
sequestration (e.g., immune neutropenia,
the setting of overwhelming bacterial infec­
splenomegaly). While patients with extrinsic
tion as well as with typhoid fever, Rocky
neutropenia may have a low circulating neu­
Mountain spotted fever, and tuberculosis.
trophil count, their tissue counts are near
Phagocytosis of microbes leads to release of
normal and they are more protected from
toxic metabolites, which then activate the
infection. A third factor is the presence or
complement system, inducing neutrophil
absence of other phagocytic cells. Many
aggregation and adherence of leukocytes to
patients with chronic neutropenia have an
the pulmonary capillary bed. Tumor necro­
elevated circulating monocyte count, which
sis factor and interleukin‐1 (IL‐1), released
provides some additional protection against
by macrophages, likely accelerate this pro­
pyogenic organisms. Conversely, patients
cess. Activated granulocytes sequestered in
being treated with steroids or other immuno­
the lungs may cause acute cardiopulmonary
suppressive therapy will have treatment‐
complications. Neonates have a limited gran­
related abnormalities in cell‐mediated,
ulocyte pool in their bone marrow, which
humoral, and macrophage–monocyte immu­
can be exhausted rapidly during overwhelm­
nity, and may be at increased risk of invasive
ing bacterial sepsis.
infection even with a normal neutrophil
Replacement of the bone marrow (as
count. Patients with a history of splenectomy
occurs with hematologic malignancies, gly­
have an increased risk of overwhelming
cogen storage diseases, granulomas associ­
infection caused by encapsulated bacteria
ated with infection, and fibrosis related to
regardless of their neutrophil count.
chemical or radiation injury or osteoporosis)
The age at presentation with neutropenia
results in neutropenia. Frequently, the eryth­
is an important factor in risk assessment.
roid and megakaryocytic lines are also
The younger the child is at the time of
affected.
diagnosis, the greater the concern that the
Medications are a common cause of neu­
patient could have a severe form of chronic
tropenia. Neutropenia is a common and
neutropenia, while children who are found
expected side effect of anticancer therapy.
to be neutropenic later in life, and who do
Many chemotherapeutic agents have a direct
not have a history of frequent bacterial
toxic effect on the early marrow stem cells.
infections or chronic illness, are more likely
The severity and duration of the neutropenia
to have a more benign form of neutropenia.
The Neutropenic Child 121

Table 11.1 Acquired causes of neutropenia.

Condition Pathogenesis Occurrence Associated findings

Infection Viral marrow suppression, Common EBV, parvovirus,


viral‐induced immune HHV6, other
neutropenia viruses
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
Immune destruction Less common condition
Autoimmune Primary (molecular mimicry) Common Monocytosis
Secondary (SLE, Evans common
syndrome, common variable
immune deficiency)
Newborn Alloimmune: Maternal Rare Antigen difference in
immune sensitization newborn and
mother
Isoimmune: Due to maternal Maternal
autoimmune neutropenia neutropenia
Idiopathic Likely most common cause is Common Consider also
autoimmune with negative familial benign
antibody testing neutropenia
Often asymptomatic
Sequestration Hypersplenism Common if Mild neutropenia
spleen is Enlarged spleen—
enlarged many causes
Nutritional Vitamin B12 or Rare in children Hypersegmented
folate deficiency neutrophils
Copper deficiency Zinc excess

Abbreviations: EBV, Epstein–Barr virus; HHV6, Human herpesvirus‐6; SLE, Systemic lupus
erythematosus.

depend on the particular medication and variable. The underlying mechanism is not
dosage as well as the patient’s underlying known, although studies with certain drugs
disease, state of nutrition, and general have led to various hypotheses including
health. Many other medications can induce immune‐mediated destruction, toxic effect
neutropenia, including antibiotics (chlo­ of the drug or metabolites on the marrow
ramphenicol, cephalosporins, penicillins, stem cells, and toxic effects on the marrow
sulfonamides), anticonvulsants (phenytoin, microenvironment. After withdrawal of the
valproic acid), anti‐inflammatory agents, drug, the marrow can repopulate with early
cardiovascular agents, tranquilizers, and myeloid forms within 3–4 days and appear
hypoglycemic agents. The severity and morphologically normal by 1–2 weeks. The
duration of drug‐induced neutropenia are duration of neutropenia is likely related to
122 Chapter 11

the underlying mechanism; some chronic antigens in the human neutrophil antigen
idiosyncratic drug reactions can last from (HNA) family, cross the placenta and cause
months to years. neutropenia in the fetus and newborn. The
Autoimmune neutropenia (AIN) is consequences of this may be quite severe,
another common cause of neutropenia. including omphalitis, cellulitis, bacteremia,
Primary AIN is a disorder of unknown and meningitis. Although usually resolving
cause and may be associated with develop­ within 6 weeks, the neutropenia can persist
ment of neutrophil‐specific autoantibodies. for as long as 6 months. Management is gen­
It has long been thought that AIN is trig­ erally supportive given the brief duration of
gered by environmental antigens or viral neutropenia. Neonates with severe neutro­
infections, but this has been difficult to penia and serious infection can be treated
prove. Primary AIN most commonly occurs with G‐CSF in hopes of accelerating neutro­
in the first two years of life (occasionally in phil recovery.
the newborn period), and has a high sponta­ Neonatal isoimmune neutropenia is
neous remission rate, perhaps as high as analogous to congenital thrombocytopenia
90%. Cases in which no specific neutrophil in infants of mothers with a history of
autoantibodies are identified are generally immune thrombocytopenia (ITP). The key
given the diagnosis of idiopathic neutrope­ to diagnosis is to consider it; the newborn’s
nia. Collectively these two conditions com­ ANC should be measured whenever a
prise the majority of chronic neutropenia mother is noted to be neutropenic. The risk
cases in young children. Until recently, of severe infection is similar to infants with
detection of antineutrophil antibodies was alloimmune neutropenia, and the manage­
challenging. Development of the granulo­ ment is the same.
cyte immunofluorescence test (GIFT) and Splenomegaly from any cause, including
the granulocyte agglutination test has made chronic hemolytic anemia, portal hyperten­
detection of these antibodies more sensitive, sion, liver disease, hemoglobinopathies, and
and current practice recommends using storage disorders can cause mild neutrope­
both tests for diagnosis. GIFT demonstrated nia, as well as anemia and thrombocytope­
a sensitivity of 60% with a single assay, and nia. Rarely, splenectomy may be necessary
82% with repeated testing. Management of due to the consequences of hypersplenism.
both AIN and idiopathic neutropenia is the Deficiencies of folate and B12 are rare in
same and relies on the history of fever and children; the presence of hypersegmented
infections to guide treatment. Granulocyte neutrophils caused by impaired neutrophil
colony stimulating factor (G‐CSF) is recom­ nuclear maturation is a clue to this condition.
mended only for patients with an Similarly, copper deficiency is a rare cause of
ANC < 0.5 × 109/l and recurrent fever/infec­ neutropenia often secondary to an underly­
tions, using the minimal effective dose ing malabsorptive process or due to zinc
(0.5–3 mcg/kg/day on a daily or every other excess. These patients usually present with
day basis). Prophylactic antibiotics, steroids, concomitant anemia and thrombocytopenia.
or intravenous immunoglobulin (IVIG) are
not recommended for treatment.
Neonatal alloimmune neutropenia is Inherited causes of
analogous to Rh‐related hemolytic disease neutropenia
of the newborn; mothers generate IgG anti­
bodies against paternal neutrophil antigens A number of inherited conditions have neu­
expressed on fetal neutrophils. These anti­ tropenia as one of their characteristic features.
bodies, most commonly directed against Hereditary neutropenias can present similarly
The Neutropenic Child 123

to acquired forms, but almost always present complete blood counts (CBCs) 2–3 times
in the first year of life with recurrent fever and per week over 4–6 weeks to demonstrate the
frequent episodes of fever, mouth ulcers, gin­ periodicity of the cycle. G‐CSF has been
givitis, sinusitis, pharyngitis, cellulitis, and used in patients who develop recurrent
respiratory and perianal infections. The more infections during their nadir.
common of these conditions are presented in Shwachman–Diamond Syndrome is an
Table 11.2. Traditionally these have been autosomal recessive disorder attributable to
named descriptively or using eponyms, but mutations in the SBDS gene. The protein
the current convention is to identify them by product of this gene is a critical component
the specific gene mutation leading to the neu­ of ribosomes, and the associated defect
tropenia, if known. in ribosomal functions affects multiple
Children with severe congenital neutro­ organ systems. Defects include exocrine
penia (SCN) often present in early infancy pancreatic insufficiency, short stature, and
with umbilical infection, skin infections, metaphyseal dysplasia. Mild to moderate
oral ulcers, pneumonia, or perineal infec­ neutropenia is usually the first hematologic
tions. The most common forms are an manifestation, but bilineage and trilineage
autosomal recessive (AR) form (Kostmann hematological abnormalities are frequent.
syndrome) involving mutations in the Affected patients also have an increased risk
HAX1 gene that is involved in signal trans­ of MDS and AML. Intestinal malabsorption
duction, and an autosomal dominant (AD) with failure to thrive commonly occurs,
form involving mutations in the neutrophil especially in infancy and early childhood.
elastase gene (ELANE) or, more rarely, in Benign familial neutropenia is character­
the GFI1 gene that targets ELANE. Bone ized by moderate neutropenia with minimal
marrow aspiration reveals a maturational risk of invasive bacterial infections. It has
arrest at the promyelocyte stage. In addition been postulated that the underlying cause is
to the risk of death due to overwhelming a defect in neutrophil mobilization from the
infection, patients with either of these con­ bone marrow, but the etiology is as yet
ditions have an increased risk of developing unknown. It occurs more commonly in
myelodysplastic syndrome (MDS) or acute individuals of African, Arabic, and Yemenite
myelogenous leukemia (AML). Multiple Jewish descent. Periodontal disease is the
additional SCN mutations have been most frequent complication.
recently discovered. Fanconi anemia is characterized by a
Cyclic neutropenia is characterized by defect in DNA repair leading to extensive
approximately 21‐day cycles of changing chromosomal breakage. Affected patients
neutrophil counts, with frank neutropenia have mutations within the FANC family of
developing at regular intervals and lasting genes, including FANC A, C, and G. It pre­
3–6 days. Fever and oral ulcerations usually sents most commonly during the early
are seen during the nadir, as well as gingivi­ school‐age years; patients with the charac­
tis, pharyngitis, and skin infections. teristic physical findings may be diagnosed
Diagnosis is often delayed because the neu­ sooner. Thrombocytopenia is often the pre­
trophil count has often improved by the senting hematologic abnormality, with ane­
time the patient seeks medical attention. mia and neutropenia developing later. Up to
These patients also demonstrate mutations 10% of patients ultimately develop MDS or
in the ELANE gene, but at different loca­ AML. Bone marrow transplantation is cura­
tions than in those with SCN. They do not tive, but challenging to complete success­
have the same risk of developing MDS or fully given the underlying chromosomal
AML. Diagnosis is made by obtaining serial fragility.
124 Chapter 11

Table 11.2 Inherited causes of neutropenia.

Condition Inheritance Pathogenesis Frequency Associated findings

Neutropenia
syndromes
Severe congenital AD ELANE mutations in neutrophil Rare (2‐3/106) Severe bacterial infections; overall risk of MDS/AML is
neutropenia elastase cause disturbed regulation of 20‐30% in patients treated with G‐CSF for >15 years
myeloid homeostasis with marrow
arrest at the promyelocyte state
Cyclic neutropenia AD Unclear mechanism due to separate Rare (1/106) 21‐day cycle with mouth ulcers, respiratory symptoms,
ELANE mutation leading to risk of cellulitis, abscesses, and severe infections
neutrophil apoptosis
Kostmann syndrome AR Mutation in HAX1 leads to impaired Quite rare, exact Severe bacterial infections; leukemia risk 15‐20%; patients
survival of myeloid precursors through frequency may have developmental delay, cognitive impairment
initiation of programmed cell death unknown and epilepsy
Familial benign AD Decreased marrow release Common Periodontal disease, otherwise not associated with an
increased infection risk; more common in Africans and
Yemenite Jews
The Neutropenic Child 125

Condition Inheritance Pathogenesis Frequency Associated findings

Bone marrow failure


syndromes
Fanconi anemia AR Impaired response to DNA damage Rare (1/106) Dysplastic thumbs, short stature, café au lait spots, 10%
(DNA repair defect) due to risk of AML/MDS
mutations in FANC genes
Dyskeratosis Usually XR Telomerase defect and ribosomal Rare (1/106) Abnormal skin pigmentation, leukoplakia, dystrophic nails
congenita (also AR dysfunction; mutation in DKC1
and AD) (TERC or TERT in AD)
Schwachman‐ AR Defective ribosomal function due to Unknown; several Pancreatic exocrine insufficiency, short stature, metaphyseal
Diamond syndrome SBDS mutation affecting multiple hundred cases dysplasia, marrow failure and leukemia risk (15%)
organs described
Diamond‐Blackfan Sporadic RPS19 mutations that affect a ribosomal Rare (1/10 )
6
Erythroid failure syndrome, rarely with neutropenia; thumb
anemia (75%); AR protein in 25% of families and craniofacial anomalies, increased RBC adenosine
and AD deaminase, many patients respond to corticosteroids;
leukemia risk 2‐3%
Chédiak Higashi AR Nonsense or null mutations in CHS1 Very rare, 200 Oculocutaneous albinism, neurologic features, recurrent
syndrome gene resulting in absence of CHS1/ cases infections, abscesses, mastoiditis, severe immunologic
LYST protein worldwide deficiency with abnormal chemotaxis and NK function
leads to HLH
Glycogen storage AR Impaired glucose homeostasis due to Rare (2/106) Hypoglycemia, hepatosplenomegaly, dyslipidemia,
disease 1b SLC37A4 mutations hyperuricemia, lactic acidemia, seizures and failure to thrive
Selective IgA Unknown May be autoimmune Common (1/600) Infections of the upper and lower respiratory tracts in
deficiency or multi‐ one‐third of patients
factorial
X‐linked neutropenia XR Unique “gain of function” missense 1‐10/106 Associated with eczema, thrombocytopenia and immune
(Wiskott‐Aldrich mutation in the GTPase binding deficiency
syndrome related domain of WASp
disorder)

Abbreviations: AD, autosomal dominant; MDS, myelodysplastic syndrome; AML, acute myelogenous leukemia; G‐CSF, granulocyte colony stimulating factor; XR, X‐linked
recessive; AR, autosomal recessive, RBC, red blood cell; NK, natural killer; HLH, hemophagocytic lymphohistiocytosis; SLC37A4, solute carrier family 37 member 4
126 Chapter 11

Chédiak Higashi syndrome (CHS) is an nary malformations. Neutropenia is rarely


autosomal recessive disease that presents in seen in affected children. Patients also
early childhood with moderate cytopenias have an increased risk of MDS, AML, and
(neutrophil count often 0.2–2.0 × 109/l) and osteogenic sarcoma. Genetic studies have
recurrent pyogenic infections. Most patients identified heterozygous mutations in at
also have oculocutaneous albinism and neu­ least one of eight ribosomal protein genes
rologic abnormalities. The genetic defects in up to 50% of cases. Mutations in riboso­
occur within the CHS1 gene often resulting mal protein L5 (RPL5) are associated with
in absent CHS1/LYST protein (a lysosomal multiple physical abnormalities, including
trafficking protein) leading to abnormal cleft lip/palate, thumb, and heart anoma­
granule formation. The granules are unable lies. The diagnosis is often difficult due to
to release lysosomal enzymes necessary for incomplete phenotypes and a wide varia­
phagocytic activity. Additionally, there is a bility of clinical expression. Up to 80% of
chemotaxis defect as well as the moderate patients respond to steroids with improve­
neutropenia, all contributing to an extreme ment in their anemia.
sensitivity to bacterial infection and sepsis, The etiology of neutropenia in glycogen
particularly with Staphylococcus aureus, storage disease 1b is not definitively known,
Streptococcus pyogenes, Pseudomonas aerug- but appears to be related to increased neu­
inosa, Pneumococcus, and Candida albicans. trophil apoptosis caused by an excess of
Most patients succumb to infection in early reactive oxygen species due to a defect in
childhood. Many patients develop lym­ neutrophil energy metabolism. Patients may
phoproliferative infiltration of the bone benefit from treatment with G‐CSF to pre­
marrow and reticuloendothelium. vent infections.
Dyskeratosis congenita (DKC) is charac­ Almost one‐third of patients with selec­
terized clinically by the triad of abnormal tive IgA deficiency have evidence of autoim­
nails, reticular skin pigmentation, and oral mune disease; neutropenia in this condition
leukoplakia. Bone marrow failure occurs is felt most likely related to the presence of
during early adulthood and is associated antineutrophil antibodies.
with a high risk of developing aplastic ane­ Patients with a mild form of Wiskott–
mia, MDS, leukemia, and solid tumors. Aldrich syndrome (WAS) known as X‐
Patients have very short germline telomeres, linked neutropenia have a unique “gain of
and approximately half have mutations in function” mutation in the GTPase‐binding
one of six genes encoding proteins that domain of the WAS protein (WASp) that
maintain telomere function. Affected leads to increased actin polymerization in
patients rarely present with hematologic neutrophils, causing defective mitosis and
abnormalities during childhood. cell movement and ultimately severe
Diamond–Blackfan anemia (DBA) is a neutropenia.
congenital anemia that presents in infancy.
There is typically a failure of erythropoie­
sis with preservation of myeloid and plate­ Initial evaluation of the child
let production. Only 20–25% of cases with neutropenia
are inherited; the rest are sporadic.
Approximately 50% of affected persons Evaluation of the child with neutropenia
have developmental abnormalities, includ­ begins with a thorough history and physical
ing growth retardation and craniofacial, examination. It is critical to know whether
upper limb/hand, cardiac, and genitouri­ the child has had recent or recurrent bacterial
The Neutropenic Child 127

infections, and whether there is a family Management of the child


history of neutropenia or recurrent bacterial with neutropenia
infections. On examination, attention
should be paid to any phenotypic abnormal­ Children identified with neutropenia but
ities, adenopathy, splenomegaly, evidence of who are free of fever or other signs of infec­
a chronic or underlying disease, and metic­ tion should be evaluated as outlined in the
ulous evaluation of the skin and mucous earlier text, but require no other therapy at
membranes (particularly in the oral and that time. Managing the child with neutro­
perirectal areas). A CBC must be done to penia and fever is more complex and
determine if the patient has isolated neutro­ depends on many factors, including the
penia or neutropenia associated with ane­ nature of the neutropenia (acute or chronic),
mia and/or thrombocytopenia. The its severity, and the association with immune
approach to a child with bilineage or defects, underlying illnesses, or malignan­
trilineage abnormalities is different than cies. Patients with acquired neutropenia aris­
that in a child with isolated neutropenia; ing from malignancy or chemotherapeutic
multiple abnormal lineages increase the drugs have a diminished inflammatory capa­
likelihood of a more generalized marrow bility and are unusually susceptible to sepsis.
failure state such as aplastic anemia or a Fever is not impacted by neutropenia and
marrow infiltrative process such as may be the earliest and only warning sign.
leukemia. It is valuable to repeat the CBC at Sepsis related to chemotherapy‐induced
least once, especially if the child appears neutropenia remains a leading cause of mor­
well, to avoid proceeding with a major tality in these patients. Aggressive manage­
evaluation due to a laboratory error or a ment of the febrile, neutropenic cancer
transient process. patient in the hospital has markedly reduced
Well‐appearing children with mild to morbidity and mortality due to infection.
moderate neutropenia can typically be For management of oncology patients with
observed over the next 2–3 weeks with serial fever and neutropenia, see Chapter 27.
CBCs. Children with persistent or worsen­ The management of the febrile child who
ing neutropenia require further evaluation. is discovered to be neutropenic depends on
Patients with recurrent neutropenia should several factors. Patients with fever and mild
have blood counts checked two to three or moderate neutropenia and symptoms
times per week for 6 weeks to evaluate for and signs suggestive of a viral illness that
cyclic neutropenia. Additional studies include otherwise look well may be managed sup­
antineutrophil antibodies, assessment of cel­ portively without antibiotics, although in
lular and serum immune status, and careful most cases they will receive a long‐acting
review of the peripheral smear for morpho­ cephalosporin such as ceftriaxone prior to
logic abnormalities of the white cells. If SCN discharge from the clinic or emergency
or Kostmann syndrome is suspected, assess­ department. Their parents should be
ment for ELANE, HAX1 and additional SCN instructed to bring them back in 24 hours
mutations should be made. A bone marrow for further evaluation, or sooner if they
aspirate and biopsy may be necessary to iden­ develop additional symptoms or begin to
tify granulocyte precursors and to search for appear unwell. If they become afebrile, their
defects in myeloid maturation. In addition, neutropenia should be evaluated as outlined
bone marrow aspirate and biopsy can be earlier. Patients with fever and mild or mod­
used to exclude hematologic malignancies, erate neutropenia who have evidence of
marrow infiltration, or fibrosis. localized bacterial disease (otitis media,
128 Chapter 11

sinusitis, or local skin infections) may be ●● CT scan of the chest (± sinuses and/or

treated with appropriate oral antibiotics in abdomen/pelvis dependent on symptoms


the outpatient setting. Patients with mild and individual patient factors) looking for
neutropenia and evidence of bacterial pneu­ evidence of occult infection in any patient
monia, GU tract infections, lymphadenitis, persistently febrile for >96 hours without a
or systemic symptoms should have cultures specific cause being identified with initia­
of the infection site and of the blood. They tion of empiric antifungal coverage due to
may be treated with appropriate oral antibi­ the increased risk of invasive fungal infec­
otics if their appearance is not concerning, tion in this patient population.
but should be seen back for re‐evaluation Prophylactic antibiotics have been used
within 24 hours. Similar patients with mod­ in 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 gener­
Febrile patients with moderate or severe ally fallen out of favor. Exceptions include
neutropenia who appear in any way unwell the use of prophylactic penicillin or amoxi­
require emergent medical assessment and cillin in patients following splenectomy, and
initiation of therapy, as do children who are the use of trimethoprim–sulfamethoxazole
known to have severe forms of neutropenia prophylaxis to prevent Pneumocystis jiroveci
such as SCN or Kostmann syndrome. These (previously known as Pneumocystis carinii)
children require: pneumonia in patients with T‐cell dysfunc­
●● Hospital admission. tion in addition to neutropenia.
●● Coverage with broad‐spectrum antibiot­ Empiric antifungal coverage should be
ics due to their increased risk of mortality considered dependent on the severity and
due to sepsis, especially Gram‐negative length of neutropenia; those with prolonged,
organisms. A fourth‐generation β‐lactam profound neutropenia should receive
cephalosporin antibiotic (i.e., cefepime) antifungal prophylaxis with fluconazole at a
provides initial broad coverage and covers minimum due to the increased risk of
Pseudomonas species, but several different invasive fungal infection in this patient
antibiotic choices are appropriate and population. Evidence for broader agents
depend on local practice; patients with skin such as extended‐spectrum azoles (i.e.,
infection should be covered for Gram‐posi­ voriconazole, posaconazole) is lacking.
tive organisms with vancomycin. The use of cytokines, particularly G‐CSF
●● Careful physical evaluation, paying has become increasingly popular in the
meticulous attention to potential sites of management of symptomatic neutropenia.
occult infection (i.e., the oral cavity and It is particularly useful in patients with
perineum). symptomatic cyclic neutropenia or in the
●● Laboratory studies: CBC with differential, neutropenia‐accompanying disorders such
blood cultures, urinalysis (UA) and urine as glycogen storage disease 1b, as these
culture, and cultures from sites of suspected patients typically respond well to low dose
infection, such as the skin, throat, and stool. therapy (1–2 mcg/kg/dose) on a daily or
●● Chest radiograph if any pulmonary signs alternate day schedule with a goal of
or symptoms are present. maintaining an ANC between 1 and
●● Daily blood cultures and a CBC with dif­ 1.5 × 109/l. It is also used frequently in
ferential every 24 hours in the persistently patients with severe chronic neutropenia.
febrile patient to help determine further However, the doses required to raise the
management. ANC into the desired range vary significantly
The Neutropenic Child 129

between patients. A typical starting dose is charged to home on day 2 of life. Her umbilical
3–5 mcg/kg/day, and the dose is increased cord fell off by 2 weeks of life. She has had no
slowly until the desired ANC is achieved. emergency department (ED) visits, hospi­
Patients with severe neutropenia who fail to talizations, recurrent infections, or pro­
respond to doses as high as 50–100 mcg/kg/ longed antibiotic courses. Her growth
day are considered poor responders. These parameters are normal, she has normal
patients are candidates for bone marrow urine and stool output. She is non‐dysmor­
transplantation given their high likelihood phic, and her skin exam is normal. Her
of mortality due to infection as well as an up fevers never correlate with oral ulcers. She
to 30% chance of developing myeloid has no recent travel or medication history.
malignancy. Mom has been told before that “low white
blood cells” run in her family, but no other
family history of recurrent infections is elu­
cidated. Your patient has never had a CBC.
Case study for review b. What initial studies would you like to
obtain?
A 2‐year‐old African‐American female A CBC is vital to assess her current white
presents to your office for evaluation of a blood cell count and differential, and to
fever to 38.7 °C. Her parents state that she determine if additional cell lines are affected.
has had fever on and off for about a week Given the number of days of fever, it would
with a similar fever occurring about once a be worth obtaining a chest X‐ray (CXR) to
month for the past few months, each time rule out a viral or bacterial pneumonia, as
associated with viral upper respiratory well as urine studies (ideally catheterized
infection (URI) symptoms. Otherwise, she urinalysis with micro and urine culture) to
has been healthy, and her parents are with­ assess for occult infection. Viral studies
out major concerns, as the fevers resolve on including RSV and influenza can be consid­
their own. Her vital signs are currently ered based on the time of year and clinical
within normal limits. Her physical exam picture.
reveals a well female, nontoxic‐appearing. CBC shows a WBC of 8 × 109/l, hemo­
There is no obvious source of infection globin of 11 g/dl, and a platelet count of
noted on exam. 200 × 109/l. Differential reveals 8% neutro­
a. What additional information would phils, 78% lymphocytes, and 12% mono­
you like? cytes, with an ANC of 640. CXR is normal,
Additional history including birth history, UA with micro is normal and urine culture
recurrent hospitalizations/prolonged antibi­ is subsequently normal, RSV and flu studies
otic courses, or recurrent or unusual infec­ are negative. Given the fever and moderate
tions will help to guide differential diagnosis. neutropenia, a blood culture is drawn which
Additional important information to obtain is later negative.
should include travel/exposure history, c. What are your considerations now?
medication usage, family history, presence This degree of marrow suppression may be
of any dysmorphic features on physical viral, the most common cause of mild to
exam, and availability of any prior compara­ moderate neutropenia. Clinical symptoms
tor labs. usually last for 7–8 days and neutropenia
She was born full‐term, via normal sponta­ should resolve within weeks. The patient’s
neous vaginal delivery (NSVD) after an increased monocyte count may be a harbin­
uncomplicated pregnancy and delivery, dis­ ger of impending neutrophil recovery.
130 Chapter 11

Autoimmune neutropenia is an additional can be in the outpatient setting if a family is


common cause, and difficult to distinguish in close proximity and able to return in
from viral suppression though is more pro­ 24 hours or with any signs of clinical wors­
longed in duration (i.e., several months to 2 ening. Any patient with a specific focus on
years), with infection risk proportionate to infection (i.e., otitis media, UTI, etc.) noted
the level of neutropenia. on exam should be treated with appropriate
Familial benign neutropenia should be a antibiotic coverage as needed, though broad
consideration given her family history of coverage should still be ensured.
“low white blood cells in the family,” which is In general, given a well‐appearing child,
more common in the African‐American close outpatient clinical monitoring and fol­
population and can be seen in 25–40% of low‐up with serial CBCs over the subsequent
those of African descent. African‐Americans 2–4 weeks until normalization are sufficient
and Middle Easterners in general have a assuming continued well‐appearance. It is at
lower WBC count, with WBC > 4.0 × 109/l the discretion of the provider to determine if
and ANC > 1000 considered normal. antibiotic coverage is necessary.
Given the moderate degree of neutrope­ In the setting of persistent neutropenia,
nia, congenital neutropenia syndromes (i.e., further workup may be required such as
SCN and Kostmann syndrome) are highly evaluation for cyclic neutropenia, antineu­
unlikely, as they require ANC < 500 for trophil or additional infectious antibody
potential diagnosis. Cyclic neutropenia is a testing, genetic testing for inherited neutro­
possible diagnosis, though is seen more penic syndromes, and possibly bone mar­
often in 19–21‐day cycles and associated row studies. Chronic benign neutropenia or
with fevers, oral lesions, pharyngitis, or skin autoimmune neutropenia are the most likely
infections. The lack of dysmorphic or other diagnoses with persistent neutropenia in the
characteristic abnormal physical exam fea­ well‐appearing child with eventual self‐res­
tures makes Fanconi anemia and dyskerato­ olution, though this may take many months.
sis congenita unlikely, though physical exam Families must be advised to monitor their
features may be quite subtle. Diamond– child closely for fever and should be evalu­
Blackfan anemia is typically a pure red cell ated urgently if febrile or ill‐appearing given
aplasia, though may have associated neutro­ the potential risk of bacterial infection with
penia. DBA is also more often diagnosed in underlying severe neutropenia.
the first year of life. Shwachman–Diamond
syndrome should be considered, though
usually presents with exocrine pancreatic Multiple choice questions
dysfunction.
d. What would be your next step? 1. You are seeing a 2-year-old patient in
Initially, given a fever in the setting of mod­ your clinic who is asymptomatic but ends up
erate neutropenia (ANC 500–1000), in a getting a CBC due to a low finger stick
well‐appearing child, coverage could begin hemoglobin. The CBC incidentally shows
with a broad‐spectrum antibiotic such as a neutropenia with an absolute neutrophil
third‐generation cephalosporin, such as cef­ count (ANC) of 300 x 106/l. There is mild
triaxone with activity against Gram‐positive microcytic anemia most likely secondary to
and Gram‐negative aerobic organisms. iron deficiency anemia. The child is well
Length of treatment is determined through appearing with no significant infections.
pending blood culture results and usually There were baseline counts done at birth
The Neutropenic Child 131

due to rule out sepsis and at that time the hypoventilation syndrome. N‐Myc is a
ANC was 4200 x 106/l. All of the following prognostic genetic marker in neuroblas­
are potential diagnoses except: toma. The answer is e.
a. Chronic benign neutropenia
b. Viral suppression 3. You are seeing a well appearing newborn
c. Kostmann syndrome who has a CBC done as part of a rule out sepsis
d. Cyclic neutropenia work up due to prolonged rupture of mem­
e. Autoimmune neutropenia branes with a maternal fever in a GBS+ mother.
Explanation: At this age, given the normal Maternal history is otherwise unremarkable.
exam and no history of infections, the most The CBC shows a WBC of 5.8 × 109/l but an
likely diagnoses are viral suppression and ANC of only 440 × 106/l. Assuming underly­
chronic benign neutropenia. Kostmann ing infection is not the cause, of the following,
syndrome is the autosomal recessive variant the most likely diagnosis is:
of severe congenital neutropenia and given a. Autoimmune neutropenia
the normal ANC at birth is ruled out as the b. Alloimmune neutropenia
neutrophil count should never be normal in c. Severe congenital neutropenia
these conditions. Cyclic neutropenia is pos­ d. Fanconi anemia
sible in this case as for many patients there e. Chronic benign neutropenia
are no significant infections. Autoimmune Explanation: A normal ANC is >1000‐1500
neutropenia occurs in a similar fashion as x 106/l. Neonates <32 weeks of age have
ITP or autoimmune hemolytic anemia and only 20% of adult neutrophil mass so neutro­
is usually a transient antibody process at this penia is common secondary to age but
age. The answer is c. should normalize by 4 weeks post‐birth.
Certain populations, namely Africans,
2. You are seeing a 1-week-old neonate who African‐Americans and Yemenite Jews, will
presents with severe omphalitis. A CBC is have a normal, lower ANC though this still
checked which shows a WBC of 12 × 109/l tends to run >1000 × 106/l. Although it may
with 67% lymphs, 32% monos and 1% be difficult to differentiate allo/autoimmune
neutrophils (ANC 120 × 106/l). You suspect neutropenia from severe congenital neutro­
a congenital neutropenia. The mutated gene penia at birth, alloimmune neutropenia is
would be: more common. And given the ANC number
a. ELANE and lack of infection, alloimmune neutro­
b. HAX1 penia is also more likely. Similar to auto­
c. PHOX2b immune thrombocytopenia, the lack of
d. N‐Myc maternal history makes auto­ immune
e. Either a. or b. neutro­penia less likely. Alloimmune neutro­
Explanation: Congenital neutropenia can penia is due to maternal alloimmunization
have an autosomal dominant form, severe of human neutrophil antigen in the father
congenital neutropenia, which is due to a (HNA‐1A, ‐1B, ‐1C). This transient anti­
defect in the ELANE gene, or an autosomal body usually clears by 2 months of age. Most
recessive form, Kostmann syndrome, which infants are asymptomatic though may pre­
is due to a defect in the HAX1 gene. A differ­ sent with infection including cellulitis,
ent mutation in the ELANE gene leads to omphalitis, pneumonia or sepsis. If with
cyclic neutropenia. PHOX2b is a gene seen infection these infants should receive IVIG
in familial neuroblastoma as well as central and GCSF. The answer is b.
132 Chapter 11

4. You are seeing a 4 year old male that has Explanation: Severe aplastic anemia can be
failure to thrive. He has had a work up for due to multiple causes including bone mar­
cystic fibrosis which was negative after stool row failure syndromes and acquired causes
studies revealed increased fecal fat. Baseline such as viral hepatitis and drugs. The majority
CBC shows neutropenia. He has short stat­ of cases are idiopathic. In addition to sugges­
ure. The likely diagnosis is: tive peripheral blood counts, bone marrow
a. Fanconi anemia biopsy is required to determine bone marrow
b. Dyskeratosis congenita cellularity. Studies to rule out paroxysmal
c. Diamond‐Blackfan nocturnal hemoglobinuria, congenital bone
d. Shwachman‐Diamond Syndrome marrow failure syndromes and viral hepatitis
e. Cartilage‐hair hypoplasia should be conducted. The answer is b.
Explanation: The constellation of findings,
exocrine pancreatic insufficiency, neutro­ 6. You are seeing a 7-year-old with multiple
penia and short stature are consistent with medical problems on a host of medications
Shwachman‐Diamond syndrome, the including valproic acid for seizures, famoti­
­second most common cause of exocrine dine for gastroesophageal reflux, and INH
pancreatic insufficiency after cystic fibro­ for latent TB infection. He also takes ibupro­
sis. Neutropenia is common though may fen and acetaminophen for fever and pain
intermittent, with or without concomitant control. You note that his ANC tends to run
anemia and/or thrombocytopenia. Exocrine low, between 600‐800 x 106/l with a low total
pancreatic function may improve with age. WBC count and you suspect drug effect. He
Skeletal abnormalities are common. The has not had any notable infections on this
SBDS gene on chromosome 7 is mutated in cocktail of medications. Which of the below
this condition. Fanconi anemia is a bone should not affect his neutrophil count?
marrow failure syndrome that typically a. Acetaminophen
presents with short stature, skeletal abnor­ b. Ibuprofen
malities, skin abnormalities (often café au c. Isoniazid
lait spots) due to a defect in DNA repair. d. Ranitidine
Dyskeratosis congenita is also a bone e. Valproic acid
marrow failure syndrome characterized by Explanation: The practitioner should be
shortened telomeres and presents with aware of a host of medication classes which
nail dystrophy, abnormal skin pigmen­ can lead to neutropenia including anti‐
tation and oral leukoplakia. Finally, inflammatories, antipsychotics and antide­
Diamond‐Blackfan anemia is a pure red pressants, anticonvulsants, antithyroid
cell aplasia and may also be characterized medications, antihistamines, antimicrobials
by short stature, skeletal anomalies and car­ and cardiovascular drugs (in addition to
diac anomalies. Cartilage‐hair hypoplasia is chemotherapeutic agents). The answer is a.
a rare condition with skeletal dysplasia,
dwarfism and immunodeficiency. The
answer is d.
Suggested reading
5. All of the following are criteria for severe Dale, D.C. (2017). How I manage children with
aplastic anemia EXCEPT: neutropenia. Br. J. Haematol. 178: 351–363.
a. ANC < 500 x 106/l Al Ghaithi, I., Wright, N.A., Breakey, V.R. et al.
b. Platelets <50 x 109/l (2016). Combined autoimmune cytopenias
c. Bone marrow cellularity < 25% presenting in childhood. Pediatr. Blood Cancer
d. Absolute reticulocyte count <20 x 109/l 63: 292–298.
12 Thrombocytopenia

Thrombocytopenia, or low platelets, can The most common cause of destructive


occur as an isolated finding or in conjunc­ thrombocytopenia is autoimmunity. The
tion with a multitude of underlying clinical markedly shortened platelet survival seen in
conditions. Normal platelet counts range this condition is a result of platelet auto‐anti­
from 150 to 400 × 109/l and thrombocytope­ body production (usually IgG), often stimu­
nia is generally defined as a platelet count lated by infection or drug exposure.
of less than 100 × 109/l. Thrombocytopenia Suppression of platelet production may also
may further be defined as mild (50– be present, and therefore ITP may actually
100 × 109/l), moderate (20–50 × 109/l), and be a disorder in which decreased platelet
severe (<20 × 109/l). Thrombocytopenia is production and destruction occur simulta­
typically subdivided into immune and non­ neously. IgM antibodies and complement
immune causes. Immune causes generally activation are less frequently found but can
cause increased platelet destruction. also be seen in childhood ITP.
Nonimmune causes may lead to increased ITP is an acute, self‐limited disease of iso­
destruction or decreased bone marrow pro­ lated thrombocytopenia that typically occurs
duction. In patients with splenomegaly, in children aged 2–5 years (though may
platelet sequestration may also lead to occur at any age from infancy to adoles­
thrombocytopenia. cence), resolving in more than 80% of chil­
dren within 6 weeks to 6 months. The
incidence in boys and girls is approximately
Acute immune equivalent. When ITP occurs in the child
thrombocytopenic purpura over 10 years of age, the course may more
likely become chronic or be associated with
Immune thrombocytopenic purpura (ITP; an underlying autoimmune disorder. The
also referred to as idiopathic thrombocyto­ otherwise healthy child presents with sud­
penic purpura) is an acquired, isolated disor­ den onset of severe thrombocytopenia (usu­
der in which the patient typically presents ally <20 × 109/l), manifested by petechiae and
with a low platelet count and symptoms of purpura and less frequently mucosal bleeding
mucocutaneous bleeding. The diagnosis of such as severe epistaxis, hemoptysis, menor­
ITP is often one of exclusion (see Table 12.1 rhagia, hematuria, and hematochezia. There
and Figures 12.1–12.3). is a seasonal fluctuation in ITP (highest in

Handbook of Pediatric Hematology and Oncology – Children’s Hospital & Research Center Oakland,
Third Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
© 2021 John Wiley & Sons Ltd. Published 2021 by John Wiley & Sons Ltd.
134 Chapter 12

Table 12.1 Differential diagnosis of thrombocytopenia.

Destructive thrombocytopenias
Immunologic ITP
Drug‐induced (valproic acid, amphotericin B,
digoxin)
Infection‐induced (HIV, VZV, CMV, EBV,
parvovirus B19, TB, hepatitis, pertussis)
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 hereditary TAR syndrome
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
Thrombocytopenia 135

Table 12.1 (Continued)


Associated with chromosomal defect Trisomy 13 or 18
Metabolic disorders Marrow infiltration
Malignancies (leukemia, neuroblastoma, metastatic
solid tumors)
Storage disease
Myelofibrosis
Acquired processes Aplastic anemia, paroxysmal nocturnal
hemoglobinuria
Liver disease/failure
Drug‐induced
Radiation‐induced
Anemia: severe iron deficiency, megaloblastic
(folate or vitamin B12 deficiency)
Sequestration Hypersplenism (portal hypertension, neoplastic,
infectious, metabolic storage disease, cyanotic
heart disease)
Hypothermia

Abbreviations: ITP, immune thrombocytopenic purpura; CMV, cytomegalovirus; SLE, systemic


lupus erythematosus; 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; TB, tuberculosis; ATRUS, amegakaryocytic thrombocytopenia
radio‐ulnar synostosis; RDS, respiratory distress syndrome; VZV, varicella zoster virus.

spring and lowest in fall) and the patient


Evaluation
often gives a history of an antecedent viral
illness within the past 1–3 weeks in up to
The initial evaluation of the child with sus­
60–70% of children or following vaccination
pected ITP begins with a complete history
with Measles‐Mumps‐Rubella (MMR) or
and physical examination. Other than a pos­
Varivax (live virus vaccines). This supports
sible antecedent illness (1–3 weeks prior)
the hypothesis that an aberrant immune
and the acute onset of minor bleeding and
response may be triggered by infection.
bruising, the child should be otherwise clin­
Severe bleeding such as protracted epistaxis,
ically well‐appearing. More significant
hemoptysis, or gastrointestinal bleeding
bleeding may be associated with trauma.
leading to severe anemia and need for trans­
Bleeding into joints (hemarthroses) should
fusion is rare. Intracranial hemorrhage
lead one to consider an alternative bleeding
(ICH) is the most feared complication, with
disorder (e.g., congenital or acquired factor
an estimated incidence of 0.1–0.5%; more
deficiency such as hemophilia). There
than 50% of patients with ICH present with
should be no history of unexplained fevers,
this finding at diagnosis or within 1 week of
bone pain, or weight loss which would be
diagnosis. More than 75% of patients with
concerning for an underlying malignancy or
ITP and ICH survive. It has yet to be defined
chronic infection. A medication history is
what risk factors predispose children with
critical as many drugs have been impli­
ITP to sustain this rare but extremely serious
cated in inducing drug‐mediated immune
complication.
136 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.

thrombocytopenia. Implicated drugs history should include an assessment for


include heparin, aspirin, aspirin‐containing autoimmune diseases, immune deficiency,
cold medications, nonsteroidal anti‐inflam­ or congenital syndromes associated with
matory drugs (NSAIDs), and seizure medi­ thrombocytopenia (see Table 12.1). The
cations such as valproic acid. Chronic physical exam should assess for lymphade­
infections with HIV, cytomegalovirus nopathy, hepatosplenomegaly, short stature,
(CMV), and hepatitis C may cause a low dysmorphic features, or the presence of con­
platelet count and appropriate screening genital anomalies (e.g., radius, thumb, and
should be done to determine risk. fingers) that may be associated with other
Helicobacter pylori has been implicated in diagnoses.
adults with ITP and there are some reports The laboratory features of ITP include a
of this association in children as well. Family low platelet count in the face of an otherwise
Thrombocytopenia 137

History, physical examination,


CBC, differential, platelet count,
peripheral smear evaluation

Isolated thrombocytopenia

Assess platelet morphology

Normal Abnormal

Assess for splenomegaly Negative Congenital Large platelets


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:
Malignancy Drug-induced
HIV Skeletal
Infection Fanconi anemia
Autoimmune disease
HIV, EBV, CMV TAR, ATRUS
Infant
Storage disease Cyanotic heart disease
Alloimmune (NAIT)
Gaucher disease Eczema
Autoimmune (maternal Ab transfer)
Hypersplenism Wiskott-Aldrich syndrome
Hemangioma (may be visceral)
Aplastic anemia Hemangiomas
Fanconi anemia Kasabach–Merritt syndrome
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 alloimmune
thrombocytopenia; Ab, antibody; TAR, thrombocytopenia absent radius; ATRUS, amegakaryocytic
thrombocytopenia radio‐ulnar synostosis.

normal complete blood count (CBC). studies other than a CBC truly need to be
However, the hemoglobin, white blood cell done if the diagnosis of ITP is strongly sus­
count, and/or absolute neutrophil count pected. The platelet count should be con­
may be altered due to a recent infection, and firmed to be low on a second evaluation if
the hemoglobin may be low if there has the physical findings do not correlate with
been significant or prolonged bleeding. the laboratory value to rule out a falsely low
Coagulation screening tests including PT, platelet count (i.e., platelet clumping or anti­
PTT, and fibrinogen and the complete meta­ body to ethylenediaminetetraacetic acid
bolic panel should be normal. Therefore, no [EDTA]).
138 Chapter 12

History, physical examination,


CBC, differential, platelet count,
peripheral smear evaluation

Thrombocytopenia with anemia

DAT test no Assess for intravascular yes


Microangiopathic
(Coombs) hemolysis, red cell process
fragments Consumption
negative positive

Reticulocyte DDx:
count Evans syndrome
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 antiglobulin test (Coombs); DDx, differential diagnosis; DIC, disseminated
intravascular coagulation; HUS, hemolytic uremic syndrome; TTP, thrombotic thrombocytopenic
purpura; CHD, congenital heart disease.

If two cytopenias are present with any platelet precursors. By convention, a BMA is
findings on the history or physical sugges­ done on individuals with suspected ITP
tive of another diagnosis such as malignancy only if steroids are to be part of the treat­
or marrow failure, consideration should be ment plan due to the small chance the
given to performing a bone marrow aspira­ thrombocytopenia may be an early manifes­
tion (BMA). However, a bone marrow eval­ tation of leukemia. However, even this prac­
uation is rarely necessary to confirm the tice is controversial and the recent American
diagnosis of ITP. Marrow morphology in Society of Hematology guidelines state this
ITP typically shows immature megakaryo­ is not routinely necessary for most children
cytes in normal or increased numbers (often who present with classic features of ITP.
with maturation arrest) and normal eryth­ Additional laboratory evaluations may be
roid and myeloid lineages. Megakaryocytes required for specific clinical indications (e.g.,
can be decreased in a subset of patients in screening for autoimmune or thyroid dis­
which immune destruction may affect early ease; viral testing for HIV, hepatitis C, CMV,
Thrombocytopenia 139

or parvovirus B19; direct antiglobulin testing and is referred to as acute within 3 months
[DAT; Coombs] and reticulocyte count if of diagnosis, persistent from 3 to 12 months,
concern for autoimmune hemolytic anemia and chronic if the symptoms have persisted
(AIHA); and immunoglobulin levels and beyond 12 months. Despite the relatively
T/B cell numbers if concern for underlying benign course experienced by most children
immunodeficiency syndrome). Antiplatelet with ITP, the sudden onset, concern for a
antibody tests are not sensitive or specific possible serious condition such as leukemia
and not routinely helpful in diagnosing ITP. or aplastic anemia, and concern for life‐
Review of the peripheral blood smear con­ threatening bleeding often guide the initial
firms a low platelet count, and the few remain­ treatment decision‐making. Parents often
ing platelets typically are large. If platelet size bring their child to see the primary care or
(mean platelet volume; MPV) is determined emergency physician, many of whom are
by an automated cell counter (Coulter), it may not accustomed to encountering such severe
be elevated, consistent with young platelet age thrombocytopenia. Risk factors for the rare
(due to early marrow release associated with life‐threatening hemorrhage have yet to be
rapid peripheral platelet destruction). More elucidated and dogma has been to treat the
often the platelet count is underestimated, as patient according to platelet count rather
very large immature platelets (megakaryocytic than being guided by the degree of clinical
fragments) are not counted by the Coulter. symptoms. It is also not possible to predict
The presence of these premature granular which patients will have a short or a
platelets may in part explain the relative lack of prolonged course, though in general
bleeding symptoms in children with very low younger children tend to have shorter
platelet counts in ITP as compared with courses and adolescents may be more at risk
decreased production in association with mar­ for a chronic course. The concern for the
row failure or following chemotherapy. The possibility of life‐threatening hemorrhage
erythrocyte and leukocyte counts and mor­ and lack of predictability has led to
phology are typically normal and there should significant controversy as to whether
be no evidence of hemolysis or microangio­ intervention is warranted, when it is
pathic disease (e.g., schistocytes or burr cells). warranted, and with what treatment.
False values for platelet counts can result Contemporary therapies have not been
from aggregation of platelets in the syringe shown to alter the course or outcome of
or collection tube, counting of small non‐ children with acute ITP. As such, the concept
platelet particles (fragmented red or white of nontreatment in ITP is increasingly
cells) by automated cell counters, and pseu­ becoming the standard of care in most
dothrombocytopenia due to in vitro platelet pediatric hematology oncology centers.
agglutination by anticoagulant‐dependent Preventative care is an important element
EDTA antibodies. Review of the blood film in the management of patients with ITP. All
should assess for clumps of agglutinated patients and families should be counseled
platelets at the periphery of the slide. regarding rough play, contact sports, and
general anticipatory guidance such as the
use of protective gear (helmets) and seat
Treatment belts. Intramuscular injections should be
withheld until platelet counts increase.
The natural history of acute ITP is complete Depo‐Provera or other progesterone
resolution in the majority of children. ITP is intervention can be helpful in decreasing or
classified according to length of symptoms stopping menstruation. Specific medications
140 Chapter 12

that interfere with platelet function (such as With observation, the majority of patients
fish oil, NSAIDs, selective serotonin with ITP will have resolution of symptoms
reuptake inhibitors [SSRI], anticonvulsants, and the underlying thrombocytopenia
and aspirin) should be avoided. while avoiding unnecessary costs and side
The use of a bleeding score as the effects of medications and hospitalization.
framework for a consistent treatment Pharmacological treatments to raise the
strategy is gaining in popularity and several platelet count in children with moderate‐to‐
scoring systems have been published to date. severe bleeding include two first‐line
Patients are categorized by the presence of therapies: intravenous immune globulin
clinical signs and symptoms of bleeding; (IVIG) and RhoGAM (anti‐Rh [anti‐D]
more than 80% present with mild clinical immune globulin, also Rhophylac).
bleeding (manifested by bruising, petechiae, Although these infusion therapies may be
mild epistaxis, and no anemia). Moderate more expensive than corticosteroids, they
clinical bleeding is characterized by more are generally the preferred initial approach
significant mucosal bleeding such as more in children due to less toxicity and
severe epistaxis or menorrhagia, and severe theoretical concerns over partially treating
bleeding (usually <5% of patients) is marked unrecognized acute lymphoblastic leukemia.
by unusual or severe bleeding requiring Response to either of these infusion
hospitalization for control or blood therapies also may help confirm the diagno­
transfusion support, with symptoms sis of ITP.
seriously interfering with quality of life. All The mechanism of IVIG is thought to be
patients may present with very low platelet saturation of Fc receptors on the reticuloen­
counts (i.e., 1–20 × 109/l) though those with dothelial cells in the spleen and liver, thereby
severe symptoms tend to have initial platelet decreasing the clearance of opsonized plate­
counts <10 × 109/l. lets. Recent studies suggest that inhibition of
Utilizing the bleeding score, many Fc receptors may be upregulated. IVIG
patients with clinically mild or moderate treatment typically results in a quick yet
bleeding can be effectively managed with transient increase in the platelet count, often
observation. The anxiety of patients, fami­ sustained 2–4 weeks. More than 80% of
lies, and other caregivers can often be patients respond with an increase in the
alleviated with education on the patho­ platelet count which is sufficient to decrease
physiology and natural history of ITP in or eliminate bleeding symptoms. A recent
addition to general preventive measures review suggests that responsive first‐line
and periodic clinical and laboratory assess­ treatment with IVIG may be predictive of a
ments. Platelet counts do not need to be lower rate of chronic disease. Families
checked more often than every 1–2 weeks should be counseled as to the transitory
(or even less often in the clinically stable effect of IVIG treatment due to the miscon­
child). Disruption of school or work is not ception that a subsequent decrease in plate­
necessary, though participation in competi­ let count is indicative of increased disease
tive contact sports or other situations that severity or recurrence. The dose of IVIG is
could lead to head or abdominal injury 0.8–1 g/kg over 4–6 hours. If necessary,
should be avoided until the platelet count IVIG may be repeated two to three times for
has returned to a safe level (typically a total dose of 2.4–3 g/kg; doses should be
>50 × 109/l). Children should not sleep in given at least 24 hours apart to allow suffi­
the top bunk bed and activities such as cient time to determine response. A rise in
climbing play structures should be avoided. the platelet count is usually seen within
Thrombocytopenia 141

24–72 hours and peaks at approximately hemoglobin for age). RhoGAM does cause
9 days. A good initial response is an increase transient hemolysis and a decrease in hemo­
in platelet count by 20–50 × 109/l. Side globin of 1–2 g/dl over the ensuing 3–4 days
effects of IVIG are usually immediate, should be expected. The hemoglobin should
related to the rate of infusion, and include subsequently recover within 10 days.
nausea, lightheadedness, and headache. At Though a rare complication, the U.S. Food
times, onset of a severe headache can and Drug Administration (FDA) currently
prompt an emergent head CT to assess for mandates that patients be monitored for 8
potential ICH. These symptoms can be alle­ hours with serial dipstick urinalysis checks
viated by slowing the rate of infusion and for hematuria and hemoglobinuria after
premedicating with diphenhydramine if administration of anti‐D due to reported
further doses are needed. Fever may also cases of severe, life‐threatening hemolysis.
occur, and premedication with acetami­ Despite anti‐D therapy being considered a
nophen is advisable. Rare side effects include first‐line intervention for ITP, its use as such
anaphylaxis in an IgA‐deficient patient, has diminished significantly.
aseptic meningitis, acute renal failure, pul­ Corticosteroids may be used in the initial
monary insufficiency, and thrombosis. medical management of ITP, though typi­
Although IVIG is a pooled blood product, it cally are considered a second‐line therapy in
is thought to be safe with regard to viral pediatrics. There are several mechanisms by
transmission and does not require consent which steroids affect the platelet count: inhi­
as with blood product administration. bition of phagocytosis of the antibody‐
Subsequent dosing with IVIG may be indi­ coated platelets leading to prolonged platelet
cated depending on clinical symptomatol­ survival; inhibition of antibody production
ogy and continued thrombocytopenia. by B‐lymphocytes; improvement of capil­
RhoGAM is an alternative therapy to lary integrity reducing clinical bleeding; and
IVIG. The dose is 75 mg/kg IV over an increase in platelet production. Response
20–30 minutes. Response rate and time to is good to oral or intravenous steroids but is
onset are similar to IVIG. Anti‐D works by slower than with IVIG or RhoGAM and is
binding the RhD antigen expressed on the usually within 3–4 days. Typically, steroids
surface of red blood cells, leading to their are prescribed at higher doses initially and
recognition by Fc receptors on cells of the then tapered as tolerated to maintain effect
reticuloendothelial system. The coated red and limit adverse reactions. The most com­
cells are thought to compete with the anti­ monly prescribed steroid is prednisone at
platelet–antibody‐coated platelets for the 1–2 mg/kg/day for 2–4 weeks, tapering the
activated Fc receptor sites, thereby slowing dose post‐platelet response (i.e., >100 × 109/l)
platelet clearance. Side effects include fever over several weeks. The initial response rate
and chills, intravascular hemolysis, head­ is 70–90%. An immune rebound may occur
ache, emesis, and rarely anaphylaxis. with a taper that is too rapid and patients
Premedication with acetaminophen and will often require prolonged steroid therapy
diphenhydramine may prevent these to maintain the desired platelet count. Side
adverse effects with subsequent infusions. effects may occur with repeated treatment
A response is typically seen within or chronic use including gastritis, fluid
24–48 hours. Patients must be Rh‐positive, retention, weight gain, mood lability, acne,
have a functional spleen (at least not known striae, high blood pressure, osteopenia,
to be splenectomized or asplenic), and not cataracts, increased infection risk, and
be anemic (i.e., >2–4 g/dl below expected elevated serum glucose. An alternative to
142 Chapter 12

oral prednisone is high‐dose pulse infusion thrombocytopenic patient, in conjunction


of intravenous methylprednisolone. Some with other therapies. Off‐label recombinant
patients become thrombocytopenic again factor VIIa (rFVIIa) may also be considered.
after therapy and require retreatment. A
relapse may be managed safely by
observation and restriction of activity and Chronic immune
medications, or with intermittent IVIG or thrombocytopenic purpura
pulse steroids. High‐dose dexamethasone
has also been used with efficacy in refractory Approximately 10–20% of children with
patients (see in the following text), but acute ITP that persists will have chronic ITP.
durable responses in children have not been The child with chronic ITP is still likely to
as likely as in adults. have complete resolution within 2 years or
Amicar (ε‐aminocaproic acid) or may have (or develop) an associated
tranexamic acid are antifibrinolytics which autoimmune disease or underlying
may be utilized in patients with prolonged immunodeficiency state. Most patients with
thrombocytopenia and persistent mucosal chronic ITP do not need treatment, as the
bleeding without anemia (mild bleeding). platelet count is often above 20 × 109/l and
Reports of efficacy with minimal side effects bleeding is minimal. Platelet count alone
make low‐dose therapy a good option (i.e., does not correlate with the risk of
Amicar 10 mg/kg/dose every 6–8 hours). hemorrhage, as platelets are large due to a
Another option is low‐dose prednisone healthy marrow response to peripheral
which may benefit the patient with a long thrombocytopenia and, as a consequence,
course of ITP who secondarily develops have greater than normal procoagulant
alteration in vascular integrity. Low‐dose activity. In the rare patient with chronic,
steroids would not be expected to raise the refractory ITP who has clinical hemorrhage
platelet count, however, they would have or cannot tolerate the living restrictions
minimal side effects. imposed by the thrombocytopenia,
intervention with medical therapies or
splenectomy should be considered.
Emergency therapy ITP may be the initial manifestation of
aplastic anemia including Fanconi anemia
Patients with known or suspected life‐ (before progression to pancytopenia), Evans
threatening hemorrhage such as central syndrome (association of ITP with AIHA),
nervous system hemorrhage from ITP systemic lupus erythematosus (SLE), or
(usually with concomitant platelet count autoimmune lymphoproliferative syndrome
<10 × 109/l) should be treated emergently (ALPS). HIV infection should be considered
with combination therapy including IVIG in all patients with isolated thrombocytope­
1 g/kg daily for 2–3 days and intravenous nia, as this is frequently the first manifesta­
high‐dose methylprednisolone 30 mg/kg/ tion of disease in children. ITP may also be
day (up to 1 g/day) for 1–3 days. Active life‐ present in children years before the diagno­
threatening hemorrhage with refractory sis of immunodeficiency (e.g., common vari­
thrombocytopenia is one of the only current able immunodeficiency) is evident.
indications for emergency splenectomy. Therapies utilized in the treatment of
Continuous infusion of platelets should be acute ITP may be utilized for chronic ITP,
considered in an emergent situation and though continued administration, cost,
during surgery or childbirth in a severely and side effects frequently limit their use.
Thrombocytopenia 143

Other considerations include additional destruction, these drugs may have some
medical approaches as discussed in the fol­ role in treatment to stimulate increased
lowing text. production, even in patients with normal
Rituximab is a chimeric human–mouse thrombopoietin levels. The FDA recently
monoclonal antibody directed against the approved eltrombopag (Promacta®) for
transmembrane CD20 antigen present on B pediatric patients with chronic ITP. The ini­
cells, leading to apoptosis and antibody‐ tial dosing is 50 mg orally once daily for
dependent cellular cytotoxicity. Children adult and pediatric patients aged 6 years
with chronic ITP may have a 30–50% and above, with dose reduction to 25 mg
response rate with rituximab. The standard once daily for patients of East Asian ethnic­
dosage is 375 mg/m2 IV weekly infusion ity or those with a history of mild to moder­
(over 4–6 hours) for 4 weeks. Most patients ate hepatic impairment. The maximum
start to show a response by the second week, daily dose is 75 mg and is available as a tab­
though delayed responses several weeks later let and an oral suspension. Children ages
may occur. Responses may be short‐lived or 1–5 years, regardless of ethnicity, start at a
last for years. Acute toxicity is minimal, dose of 25 mg once daily. The dose is then
though patients may experience fever and adjusted at 2‐week intervals by 12.5 or
chills (often abates with subsequent infu­ 25 mg to maintain a platelet count of ≥50
sions), serum sickness, headache, nausea, and <200 × 109/l. There are certain dietary
emesis, and mucocutaneous reactions (con­ considerations with respect to timing of
tinuum from hives and dermatitis to rarely drug administration due to interference of
reported Stevens–Johnson syndrome and absorption. The medication needs to be
toxic epidermal necrolysis). B‐cell depletion taken on an empty stomach (1 hour before
lasts for approximately 6 months with a sig­ or 2 hours after a meal) and 2 hours before
nificant decrement in humoral response, or 4 hours after consumption of polyvalent
although decreased total IgG immunoglobu­ cations (i.e., iron, calcium, aluminum, mag­
lin levels and increased infections have not nesium, selenium, zinc) which are primar­
been frequently reported. For the treatment ily found in dairy products, supplements,
of ITP without an underlying immunodefi­ and antacids. The two studies leading to
ciency, it remains unclear if there exists an FDA approval demonstrated that in chil­
increased infection risk in pediatric patients dren with ITP, eltrombopag improved
after receiving rituximab. Although consid­ platelet counts, allowed for reduction or
ered a T‐cell‐dependent infection, B‐cell discontinuation of concomitant ITP medi­
depletion may lead to an increased risk of cations and decreased the need for rescue
PCP (Pneumocystis jiroveci pneumonia) and therapy. Concerning toxicities include mar­
cotrimoxazole prophylaxis should be consid­ row reticulin fibrosis, cataracts, liver
ered. Hepatitis B reactivation and progres­ enzyme abnormalities, and thromboembo­
sive multifocal leukoencephalopathy lism, as well as rebound thrombocytopenia
secondary to JC virus are rare considerations with abrupt discontinuation. It is likely that
in this population. these drugs or subsequent generations will
The thrombopoietin (TPO) receptor provide potential new therapeutic options
agonists (RAs) are a novel class of drugs and in the future.
may diminish or eliminate the need for Another TPO RA under investigation is
immunosuppressive therapy in ITP. As ITP romiplostim (Nplate®). This drug is also
is likely the result of impaired production in likely soon to receive a pediatric indication
addition to peripheral immune‐mediated for chronic ITP. It is administered as a weekly
144 Chapter 12

intramuscular injection and does not impose Vinca alkaloids (vincristine and vinblas­
the dietary timing issues of eltrombopag. tine), danazol (a virilizing androgen), and
Clinical evidence demonstrates that eltrom­ immunosuppressive agents such as azathio­
bopag and romiplostim do not have cross‐ prine and cyclophosphamide have also been
resistance, likely due in part to different used with some success. Ascorbic acid,
binding sites on the TPO receptor. Therefore, cyclosporine, and interferon α‐2b are other
patients not responsive or unable to tolerate agents that have also been investigated for
one TPO RA may still benefit from treat­ use in chronic ITP. Long‐term management
ment with the other. Early data suggest that of chronic ITP includes periodic assess­
treatment with TPO RAs may help increase ments for development of other manifesta­
the number of ITP patients who achieve a tions of immunodeficiency or autoimmune
remission before becoming chronic, and disease, counseling regarding activities and
therefore there may be a role for these medi­ medications, and periodic assessments of
cations in patients with acute or persistent the platelet count to assess for continuation
ITP. Early treatment with this class of drugs or resolution of the underlying process.
may become a preferred intervention to
avoid exposure to conventional therapies
and the associated risks and may alter the Neonatal alloimmune
disease course. More studies need to be con­ thrombocytopenia
ducted to elucidate these potential benefits.
Splenectomy may be considered for Neonatal alloimmune thrombocytopenia
patients with chronic ITP with bleeding or (NAIT) is the result of early transplacental
limitation in activities negatively impacting passage of maternal alloantibodies directed
quality of life. Current guidelines agree that against fetal platelets, similar in pathophysi­
it should only be considered in patients with ology to hemolytic disease of the newborn.
chronic ITP or the rare refractory patient Unlike hemolytic disease of the newborn,
with life‐threatening bleeding. Splenectomy NAIT often affects the first‐born offspring.
is successful in 60–85% of patients; however, Although rare, NAIT is the most common
relapse of ITP may occur due to immune‐ cause of severe thrombocytopenia in the first
mediated platelet destruction in other few days of life. Alloantibodies are present
organs, especially the liver. Patients should due to human platelet antigen incompatibil­
ideally receive appropriate immunization ity, as fetal platelet antigens are inherited
several weeks prior to surgery due to the risk from the father. Immunization in the mother
of infection with encapsulated organisms against fetal platelet antigens can occur dur­
(i.e., pneumococcus, Haemophilus influenza, ing the current or a previous pregnancy or
or meningococcus). Lifelong penicillin secondary to a previous platelet transfusion.
prophylaxis is recommended postsplenec­ NAIT may lead to severe thrombocytopenia
tomy. Given the lifelong risk of overwhelm­ in the fetal‐newborn period, with a high risk
ing sepsis, and the recommended prophylaxis of fatal hemorrhage. Thrombocytopenia
and preemptive therapy for presumed bacte­ leading to fetal loss and hemorrhage has
rial infection, the benefit of this approach been reported as early as 16–24 weeks of ges­
must be considered individually. In the field tation. Although several platelet antigens
of emerging innovative therapies, including have been implicated, 75% of cases are
the recently introduced TPO RAs, the ques­ related to human platelet antigen 1a (HPA‐1a)
tion of who needs splenectomy is becoming incompatibility (i.e., HPA‐1a‐negative
a more challenging question to answer. mother with a HPA‐1a‐positive fetus).
Thrombocytopenia 145

HPA‐1a resides on the GP IIb/IIIa complex thrombocytopenic secondary to birth


that is responsible for fibrinogen receptor asphyxia, infection, congenital bone mar­
activity and thus important in aggregation row hypoplasia, or prematurity. The pres­
and platelet‐plug formation. Therefore, ence of hepatosplenomegaly, intrauterine
development of anti‐HPA‐1a antibodies may growth retardation, or intracranial calci­
interfere with normal platelet aggregation in fications with thrombocytopenia should
addition to decreasing platelet number, suggest a congenital viral infection.
resulting in both a qualitative and quantita­ However, if a secondary diagnosis is not
tive platelet defect. This may in part explain clearly delineated, it is important to exclude
the high incidence of serious bleeding in alloimmune thrombocytopenia by appro­
these infants as compared with those born to priate immunologic testing as future‐
mothers with ITP with alloantibodies affected pregnancies tend to be more severe
directed against alternate antigens. More and require close antenatal monitoring.
than 20 antigens are involved in NAIT with NAIT should be considered in all newborns
HPA‐1a being most common in the with thrombocytopenia.
Caucasian population, in addition to Early platelet alloantigen evaluation of
HPA‐5b. HPA‐4 incompatibility is more the newborn and parents is important, both
common in Asian populations. in offering the affected infant appropriate
The infant with NAIT typically presents treatment and in order to minimize the risk
with moderate‐to‐severe thrombocytope­ of devastating complications with future
nia (i.e., <20–50 × 109/l) and may rapidly pregnancies. Evaluation includes platelet
develop petechiae, purpura, hematuria, and typing on the mother and father, specifically
bleeding from the umbilical cord, veni­ assessing for known antigens responsible for
puncture sites, or gastrointestinal tract. alloimmunization such as HPA‐1, ‐3, and ‐5
ICH is common, occurring in 10–20% of antibodies as well as HPA‐4 antibodies in
neonates and may occur prenatally or ante­ those of Asian descent. HPA‐9 and ‐15 are
natally. Therefore, head ultrasound should the next most common antigen
always be part of the initial diagnostic incompatibilities. Serum from the mother
workup. ICH may be detected on ultra­ and infant is screened for antiplatelet
sonography during an apparently uncom­ antibodies against either the infant’s or the
plicated pregnancy and complications of father’s platelets. We send blood for the
early central nervous system hemorrhage NAIT workup to the Blood Center of
include hydrocephalus, porencephaly, sei­ Wisconsin™.
zures, and fetal loss. Early jaundice occurs Several treatment options are available
in 20% of cases owing to resolution of for the infant with neonatal alloimmuni­
intracranial or intraorgan hemorrhage. The zation. Transfusion with antigen‐negative
infant is otherwise generally healthy with­ platelets has been the mainstay of treat­
out other perinatal complications and the ment, when available. Since HPA‐1a‐neg­
mother typically has an unremarkable ative platelets are present in only 2% of the
hematologic history (other than possibly a Caucasian population, the most available
previously affected fetus or newborn). For a source of platelets is from the mother
mother with a previous low platelet count, (though this is not always technically
the differential diagnosis should include feasible). Random platelet transfusions
maternal idiopathic, autoimmune, or drug‐ (10–20 ml/kg) may provide a transient
dependent thrombocytopenia, infection, increase, lasting 1–2 days, and should be
and preeclampsia. Infants may also be used in cases of serious hemorrhage, while
146 Chapter 12

antigen‐negative platelets are being appropriately selected donor should be


obtained and prepared. If random donor obtained and prepared prior to delivery in
platelets are ineffective, then cross‐ the event of extreme thrombocytopenia or
matched donor platelets or washed mater­ hemorrhage. The infant’s platelet count
nal platelets may be used if available. An should be checked at birth and every
excellent alternative treatment is IVIG. 6–12 hours for 1–2 days and be kept
The recommended dose is 1 g/kg/24 hours >50 × 109/l. Frequency of monitoring can be
for 1–2 doses (until the platelet count is decreased depending on the stability of the
≥50 × 109/l). Platelet transfusion may still platelet count and the clinical status of the
be necessary with IVIG if immediate cor­ infant.
rection of the thrombocytopenia is needed.
Corticosteroids can be effective in reduc­
ing platelet destruction and increasing Neonatal autoimmune
vascular integrity. Methylprednisolone, thrombocytopenia
1 mg/kg IV every 6–8 hours (with IVIG), is
frequently effective. Regular head ultra­ Neonatal autoimmune thrombocytopenia
sounds should be done while the infant occurs due to the passive transfer of auto‐
remains severely thrombocytopenic antibodies from mothers with ITP or may
(<50 × 109/l). If intracranial bleeding is be caused by other maternal disorders
present, platelets should be kept including autoimmune diseases such as
≥100 × 109/l. Head MRI is necessary to fur­ systemic lupus erythematosus, hypothy­
ther define the hemorrhage. Ultrasound roidism, and lymphoproliferative states, or
should be repeated at 1 month in children secondary to m ­ edications. Unlike NAIT,
with ICH to identify early hydrocephalus. these antibodies recognize both maternal
Resolution of thrombocytopenia typically and fetal platelet antigens. Maternal ITP
occurs within 2–4 weeks. should be distinguished from gestational
Mothers with an affected infant should thrombocytopenia that tends to occur late
be counseled on the necessity for careful in pregnancy and leads to mild thrombocy­
monitoring with future pregnancies due to topenia in the mother (i.e., 70–100 × 109/l)
the risk of increasingly severe NAIT. Fetal without the development of antibodies, in
platelet counts should be obtained starting which case the infant is not at risk for
at 20 weeks of gestation (12 weeks if prior thrombocytopenia. In gestational throm­
history of ICH), with ultrasound monitoring bocytopenia, maternal platelet counts
for hemorrhage. Maternal antiplatelet titers return to normal shortly after birth. The
cannot be used to accurately predict affected degree of thrombocytopenia seen in neo­
fetuses. IVIG 1 g/kg/week given to the nates born to mothers with ITP is less
mother from mid‐gestation until near term severe than with NAIT, with only 10–15%
has been shown to effectively increase fetal having platelet counts <50 × 109/l. Bleeding
platelet counts in the majority of cases. is minimal and ICH is rare (i.e., 0–3%). The
Delivery should be planned near term with platelet count may be normal at birth but
an elective cesarean section or planned falls within 1–3 days after delivery. Platelet
induced vaginal delivery after documented counts should be monitored closely and a
increase in fetal platelet count following head ultrasound obtained to exclude ICH.
administration of maternal IVIG. Antigen‐ Treatment should be initiated if the platelet
negative platelets, which can be obtained count falls below 30–50 × 109/l with the
from the mother by apheresis or from an same treatment regimen as for NAIT:
Thrombocytopenia 147

IVIG, IV methylprednisolone, and random risk for skin necrosis at subcutaneous hepa­
donor platelet transfusion for treatment of rin injection sites. If suspected, based on
hemorrhage. The duration of thrombocy­ clinical scoring that defines likelihood, func­
topenia is usually 3–6 weeks but may last as tional studies of platelet activation under the
long as 12 weeks. presence of heparin can be done by special­
ized laboratories. This testing includes the
serotonin release assay and heparin‐induced
Drug‐induced platelet aggregation assay. Heparin should be
thrombocytopenia discontinued and replaced by an anticoagu­
lant that does not lead to antibody formation
In addition to immune‐mediated mecha­ such as argatroban. Warfarin should not be
nisms induced by drugs, many bone marrow utilized immediately due to associated pro­
suppressive agents such as chemotherapy tein C deficiency with risk of microthrombo­
cause thrombocytopenia, though typically in sis leading to skin necrosis and gangrene.
the face of pancytopenia. Management is
usually with platelet transfusion, to prevent
or treat bleeding. The bone marrow effects of Nonimmune thrombocytopenia
these agents often define their dose‐limiting
toxicity, and thrombocytopenia is a common Many nonimmune‐mediated processes
effect. Patients respond well to platelet trans­ lead to increased platelet consumption.
fusion but can become refractory because of Generalized platelet activation with trap­
the underlying illness, organomegaly and ping of microaggregates in the small vascu­
sequestration, development of alloimmun­ lature contributes to microangiopathic
ization, sepsis, and other medications. hemolytic anemia (MAHA) occurring in
Other potential offending drugs include congenital heart disease, hemolytic uremic
anticonvulsants such as valproic acid, chlo­ syndrome (HUS), and thrombotic throm­
rothiazides, estrogenic hormones, ethanol, bocytopenic purpura (TTP). HUS is pri­
ristocetin, and protamine sulfate. marily associated with a prothrombotic state
Heparin‐induced thrombocytopenia induced by exposure to shiga‐toxin‐produc­
(HIT) is caused by antibody formation to ing Escherichia coli (O157:H7) or Shigella
complexes of heparin and platelet factor 4 dysenteriae 1, particularly affecting the renal
leading to platelet activation, often resulting vasculature and leading to the triad of
in severely low counts as well as risk for MAHA, platelet consumption, and renal
thrombosis. It is much less common in chil­ failure. HUS is the most common cause of
dren than adults. HIT should be suspected in acute renal failure in children. Patients may
the child receiving heparin (usually second­ present with abdominal pain and bloody
ary to unfractionated but can also occur with diarrhea and are treated with supportive
low molecular weight) who develops unex­ care (i.e., red cell transfusions, dialysis as
plained thrombocytopenia of any degree necessary). Platelet transfusion may worsen
within 5–10 days of exposure, often defined the clinical status and should be used with
as a decrease of ≥50% from baseline (even if caution.
still in the normal range). Testing for hepa­ Idiopathic TTP is a rare disease in chil­
rin‐induced antibodies is not very specific, dren, characterized by the pentad of throm­
as many patients without HIT will have bocytopenia, hemolytic anemia, renal
­circulating antibody. Patients may develop impairment, neurologic symptoms, and
thrombosis (venous or arterial) and are at fever, although few patients present with
148 Chapter 12

the full gamut of symptoms. Idiopathic TTP prolonged PT/PTT, elevated D‐dimer and
is often clinically indistinguishable from hypofibrinogenemia, as well as elevated
diarrhea‐negative HUS. There is also a rare blood urea nitrogen (BUN) and creatinine.
congenital form in which affected neonates Without treatment, mortality is >90%.
present with jaundice and thrombocytope­ Plasmapheresis is the mainstay of therapy in
nia, although patients may not have an epi­ the acquired form. Fresh frozen plasma is
sode of overt TTP for years until triggered usually sufficient to treat the congenital
by infection, pregnancy, or stress. Patients form. Patients may also benefit from rituxi­
with congenital TTP have extremely low mab and other immunosuppressive drugs
levels of ADAMTS13, a protein that cleaves including steroids, cyclosporine, cyclophos­
unusually large multimers of von phamide, vincristine, and azathioprine.
Willebrand factor into biologically less‐ Increased utilization of platelets may
active forms. Absence of ADAMTS13 occur in active bleeding, infection, or sepsis.
inhibits cleavage of these large multimers In disseminated intravascular coagulation
allowing spontaneous platelet adhesion and (DIC), there is an imbalance between intra­
aggregation. vascular thrombosis and fibrinolysis, with
Patients with acquired TTP, which is increased platelet consumption, depletion of
often idiopathic, commonly demonstrate plasma clotting factors, and formation of
antibodies to the protein, unlike the con­ fibrin. DIC can be initiated by many events,
genital form in which there is a constitutive including sepsis due to bacteria, viruses, or
deficiency. Affected individuals are more fungi; malignancy, particularly acute pro­
commonly female, of African descent, and myelocytic leukemia and neuroblastoma;
diagnosis can be associated with pregnancy, hemolytic transfusion reactions; and
autoimmune disease, infection, or trans­ trauma. Therapy is aimed at treating the
plantation. The hallmark of disease is the underlying etiologic process. Supportive
presence of segmental hyaline micro­ care consists of platelet transfusion to main­
thrombi in the microvasculature that can tain platelet counts >50 × 109/l and plasma
also be seen in the lymph nodes and spleen. protein replenishment to correct coagulopa­
Classic signs and symptoms include fever, thies and maintain fibrinogen >100 mg/dl.
malaise, nausea and vomiting, abdominal Thrombocytopenia can occur in the sick
and chest pain, arthralgia and myalgia, pal­ newborn for many reasons, most commonly
lor, jaundice, purpura, progressive renal in association with infection, prematurity,
failure, and fluctuating neurologic signs asphyxia, respiratory distress syndrome,
and symptoms. Laboratory features include pulmonary hypertension, or meconium
thrombocytopenia and MAHA. The aspiration. These infants appear to have
peripheral blood smear will show polychro­ normal to increased platelet production, but
masia, basophilic stippling, schistocytes, a decreased platelet life span for reasons that
microspherocytes, and nucleated red cells. are unclear. Thrombocytopenia is a frequent
The direct antiglobulin test (DAT; Coombs) occurrence in congenital cyanotic heart
should be negative, as TTP is not an AIHA. disease associated with compensatory
The LDH and unconjugated bilirubin will polycythemia.
be elevated and haptoglobin reduced due to The association of thrombocytopenia
the MAHA, with associated hemoglobinu­ and giant hemangiomas occurs in the
ria. Thrombocytopenia is often more severe infant with Kasabach–Merritt syndrome
than the degree of hemorrhage would pre­ and represents a form of localized intravas­
dict. Evidence of DIC may be present with cular coagulation. The hemangiomas may
Thrombocytopenia 149

be multiple and may involve only viscera. but carry the risk of causing platelet
Therefore, in an infant with unexplained dysfunction in addition to the existing
­
thrombocytopenia, imaging studies should thrombocytopenia.
be done to look for a vascular anomaly. A variety of conditions that result in sple­
Hemangiomas are proliferative lesions that nomegaly are associated with thrombocyto­
grow rapidly for several months and then penia. The large spleen sequesters and
regress spontaneously. Platelet thrombi destroys circulating platelets. Anemia, leu­
may develop in these lesions and platelet kopenia, and neutropenia may also be pre­
life span may be decreased. These infants sent. Megakaryocytic production in the
may also have a consumptive coagulopathy marrow is normal and may be accelerated in
with low fibrinogen levels and elevated response to a decrease in the circulation.
concentrations of fibrin degradation prod­ Storage diseases, early portal hypertension,
ucts. The lesions are also prone to necrosis hemolytic conditions (red cell membrane
and infection. A particular hemangioma’s defects), infections such as with HIV,
size or location cannot predict whether it Epstein–Barr virus (EBV), and CMV, and
will lead to platelet trapping and thrombo­ malignancies are frequently associated with
cytopenia. These infants should be man­ splenomegaly and may result in hypersplen­
aged by close observation and hematologic ism (increased splenic activity and resultant
monitoring while waiting for regression to red cell destruction).
occur. However, the lesions may become
large enough to compromise the infant by
impinging on the airway or vital organs, Decreased platelet production
leading to compartment syndrome, and
resulting in serious illness or death. Thrombocytopenia due to decreased produc­
External compression of hemangiomas by tion of platelets may be a result of an acquired
firm bandaging, when possible due to loca­ or inherited disease process. Decreased plate­
tion, may reduce blood flow and platelet let production may be a direct effect of mar­
trapping. Propranolol has become first‐line row crowding due to malignancy (leukemia
therapy for regression of hemangiomas, or metastatic solid tumors such as lymphoma,
likely secondary to inhibition of angiogen­ neuroblastoma, medulloblastoma, and rhab­
esis. Corticosteroid treatment at a dose of domyosarcoma) or storage disease (Gaucher,
1–2 mg/kg/day may also bring about Neimann–Pick, etc.). Drugs may also be
regression of the lesion and normalization implicated in decreased platelet production.
of the platelet count. Interferon α‐2a has The liver is the site of TPO production and
been shown to be beneficial in correcting liver disease is associated with chronic severe
the platelet count and shrinking the lesion. thrombocytopenia. Severe iron deficiency can
Supportive transfusion therapy is indicated result in decreased production of platelets,
with active bleeding due to thrombocyto­ though early iron deficiency states are associ­
penia. Platelet transfusion as well as infu­ ated with an elevated platelet count likely due
sion of coagulation factors (fresh frozen to marrow stress. Diseases affecting the mar­
plasma, cryoprecipitate, and antifibrino­ row matrix (aplastic anemia and myelofibro­
lytic drugs) may be helpful but usually are sis) cannot support stem cell growth and
of only transient benefit. Antiplatelet med­ maturation with resultant development of
ications (aspirin and dipyridamole) have thrombocytopenia.
been used in the past to interfere with Thrombocytopenia related to an inher­
platelet trapping within the hemangioma, ited condition is frequently distinguished by
150 Chapter 12

characteristic clinical features, early presen­ receptor c‐mpl. Bleeding symptoms lead to
tation and chronic course, family history, diagnosis in infancy, although CAMT is often
platelet morphology, and lack of response to initially confused with other more common
classic treatments for ITP. A number of neonatal causes of thrombocytopenia such as
these conditions are associated with alloimmune‐ and autoimmune‐mediated pro­
­macrothrombocytes and mild‐to‐moderate cesses. However, unlike these other conditions,
thrombocytopenia including Bernard– the thrombocytopenia does not resolve with
Soulier syndrome, MYH9‐related disorders time. There are no classic physical features.
(May–Hegglin anomaly with bluish cyto­ Diagnosis is based on markedly reduced mega­
plasmic inclusions including Sebastian‐, karyocytic precursors in the bone marrow with
Fechtner‐, Epstein‐, and Alport‐like syn­ normal erythroid and myeloid lineages with
dromes), platelet‐type von Willebrand dis­ elevated TPO levels. Current treatment is sup­
ease, gray platelet syndrome (storage pool portive care, platelet transfusion as needed, and
disease), benign Mediterranean macro­ consideration for curative hematopoietic stem
thrombocytopenia, Paris‐Trousseau‐type cell transplantation.
thrombocytopenia, and more poorly Children with ATRUS present similarly
defined syndromes such as Montreal plate­ to CAMT with severe thrombocytopenia at
let syndrome. Microthrombocytes are seen birth, but in association with skeletal anom­
in Wiskott–Aldrich syndrome, an X‐linked alies and sensorineural hearing loss.
disorder resulting from a mutation on the Patients may have fusion of the radius and
WAS gene. Wiskott–Aldrich syndrome is ulna at the elbow, often with minor clinod­
characterized by thrombocytopenia, recur­ actyly or anomalies involving the humeri or
rent bacterial and viral infections secondary lower limbs. The disease may progress to
to T‐cell dysfunction, chronic eczema, and aplastic anemia or leukemia. A HOXA11
an association with autoimmune disorders. mutation has been identified in two kin­
Patients with defects in the WAS gene may dreds and this mutation inhibits megakar­
also have a milder syndrome called X‐linked yocyte differentiation. Most cases of TAR
thrombocytopenia with small platelets and are diagnosed at birth or in utero due to
immune dysregulation which may develop bilateral absence of the radii manifested
over time. Of note, it is difficult to appreciate as a shortening of the forearms and flexion
small platelets in the newborn and the mean at the elbows, with preservation of the
platelet volume reported on the CBC is thumbs. Patients may have platelet dys­
unreliable in the face of thrombocytopenia. function and are at risk for significant
Patients with inherited thrombocytopenia bleeding episodes. Typically, the thrombo­
may have normally sized platelets in certain cytopenia improves through childhood.
conditions including congenital amegakaryo­ Associated clinical features include addi­
cytic thrombocytopenia (CAMT), thrombocy­ tional skeletal limb defects, renal and
topenia with absent radii (TAR), familial ­cardiac anomalies, facial capillary heman­
platelet disorder and predisposition to acute giomas, and cow’s milk intolerance. Fanconi
myelogenous leukemia (AML), amegakaryo­ anemia is an inherited autosomal recessive
cytic thrombocytopenia with radio‐ulnar syn­ (>99%) and rarely X‐linked recessive
ostosis (ATRUS), and autosomal dominant (FANCB, <1%) disorder characterized by
thrombocytopenia. CAMT, a rare cause of chromosomal instability with skeletal
neonatal thrombocytopenia, is a bone marrow anomalies and hypoproductive thrombocy­
failure syndrome inherited in an autosomal topenia, although other cell lines are even­
recessive manner due to deficiency in the TPO tually affected. These patients have an
Thrombocytopenia 151

increased susceptibility to develop leuke­ diagnoses such as von Willebrand disease


mia and may benefit from early hematopoi­ and acute leukemia and ask questions
etic stem cell transplantation if a suitable related to these possibilities. In this case,
donor is available. the mother states that he has been healthy,
with no prior medications, illnesses, hospi­
talizations, or other comorbidities. There is
Case study for review no family history of a diagnosed bleeding
disorder (though you make a mental note
A 9‐year‐old previously well Hispanic boy to pursue these questions later, as family
presents to the emergency department with members may not have been evaluated for
a 2‐hour history of profuse epistaxis. On symptoms suggestive of von Willebrand
presentation he is noted to be well‐developed disease). The mother states that he on
and well‐nourished, frightened, fatigued, awakening this morning had unexplained
and pale, with numerous ecchymoses and bruises over his torso and extremities and
blood pouring from both nares. He is blood blisters in his mouth. He complained
emergently triaged and has bilateral nasal of feeling sick to his stomach and began to
packing placed. He continues to have have simultaneous emesis and nose bleed­
massive bleeding and has repeated emesis of ing, which led to her calling an ambulance
digested and bright red blood. His vitals are as she could not stop it.
T 37.2° C, HR 156, RR 30, and BP 62/38. c. What findings do you look for on
a. What are your immediate thoughts examination to either confirm or dispute
regarding possible diagnosis? How do your suspected diagnosis?
you confirm this and what steps do you On examination, you note he is alert and
take in treatment and stabilization of oriented, though frightened to see so much
your patient? blood and expresses to you he thinks he is
b. What pertinent history should you going to die. You provide reassurance and
obtain? quickly note his oral and skin findings of
The first concern is to stop the bleeding and diffuse petechiae and purpura. Purpuric
treat the patient for acute, symptomatic lesions on the torso are very unusual and
hemorrhage. You suspect a bleeding disor­ frequently not associated with trauma,
der and see that the patient is an otherwise reflecting more spontaneous bleeding. He
healthy child with no known inherited has no adenopathy, organomegaly, other
bleeding diathesis. Although ITP presents skin rashes, and no pain on palpation of the
with acute massive bleeding in fewer than extremities and abdomen with full range of
5% of cases, you are appropriately concerned motion of all joints, and in general appears
that he has mucosal‐type bleeding associ­ well‐developed and appropriate for age. All
ated with either an acquired or inherited these findings are supportive of a diagnosis
platelet problem, which may be qualitative of ITP.
or quantitative. You order a emergent CBC Your patient’s CBC comes back with a
to determine both platelet and hemoglobin platelet count of 2 × 109/l, hemoglobin
levels and initiate fluid resuscitation await­ 6.2 g/dl, and white blood cell count of
ing the results. 7.4 × 109/l with a normal differential. You
Additional historical points to elicit explain the anemia as secondary to massive
include recent illness, medication expo­ epistaxis.
sure, underlying disease, and prior symp­ Your patient now has evidence of gross
toms. You are contemplating other hematuria and melena, with a further drop
152 Chapter 12

in blood pressure despite having received a peripheral destruction or sequestration


unit of packed red blood cells and volume has been removed. Splenectomy is not suc­
support. The complete metabolic panel and cessful in ≥30% of cases and it cannot be
coagulation studies are normal. predicted who will respond. Our patient
d. What is your next thought for continues to receive massive blood product
treatment? support for 1 week, then starts to stabilize
As you are now certain that this is acute with an increase in hemoglobin to 8 g/dl and
ITP and your patient is experiencing life‐ platelet count of 54 × 109/l. He continues on
threatening hemorrhage, you initiate therapy a course of steroids, eventually transitioning
with IVIG 1 g/kg IV over 4 hours, methyl­ to oral prednisone, and tapering off by 6
prednisolone 1 mg/kg IV every 8 hours, weeks. By this time his platelet count nor­
continuous infusion of packed red blood malizes, and several years later he continues
cells, and fluid support with lactated Ringer’s to have a normal platelet count.
or normal saline. Consideration should also Of interest, the patient represents with
be given to continuous infusion of platelets. moderate epistaxis, no anemia, and a nor­
Consultation with an otolaryngologist mal platelet count 1 year later. Further diag­
should be obtained for nasal packing and nostic workup reveals a diagnosis of type 1
control of the bleeding in the posterior von Willebrand disease.
pharynx. f. How would the knowledge of this
e. How do you counsel the family? comorbidity have influenced your man­
The prognosis of ITP, even in the face of agement of the patient with the initial
such a dramatic presentation, is excellent. presentation of ITP and massive
However, this situation is tenuous due to epistaxis?
lack of therapies that could cause an imme­ Knowledge of this disease may have
diate rise in the platelet count. At this time, led to the use of additional therapies
his prognosis is guarded. such as DDAVP to stimulate release of
Your patient is hospitalized in the von Willebrand factor, and Humate‐P, a
Pediatric Intensive Care Unit (PICU) and factor VIII concentrate that also con­
continues to receive red cell transfusions, tains von Willebrand factor and is used
continuous platelet transfusion (1 pheresed for von Willebrand factor replacement.
unit every 2–3 hours), fresh frozen plasma It is unknown whether such additional
(FFP) infusion, further doses of methyl­ therapies may have impacted the clinical
prednisolone, and a second dose of IVIG. course.
On day 3, the hemoglobin drops to 4.6 g/dl
and the decision is made to perform an
emergent splenectomy. The family is coun­ Multiple choice questions
seled that he may not survive this procedure.
Fortunately, however, he does very well 1. You are in the newborn nursery and have
and is supported with transfusions without a 1-day-old male infant, 2.6 kg, born at 37
a worsened clinical course. Unfortunately, weeks GA to a G1P1 healthy 34-year-old
there is no immediate response to splenec­ mother with PROM and GBS+ leading to a
tomy. Typically, in patients that do respond, sepsis rule out. The baby is well appearing
there is an immediate increase in postoper­ with a small cephalohematoma but other­
ative platelet count, often to above normal wise normal exam. The baby’s CBC checked
levels due to an exaggerated marrow at birth is normal except for a platelet count
response that persists after the source of of 27 × 109/l. Bcx is pending on antibiotics.
Thrombocytopenia 153

Assuming the sepsis work up is unremarkable, and father should be sent to the Blood
the most likely diagnosis is Center of Wisconsin™ for work up as it is
a. Autoimmune thrombocytopenic important, if positive, for future pregnan­
purpura cies. IVIG can also be given to help dilute
b. Alloimmune thrombocytopenia the antibody response as well as compete
c. Bernard‐Soulier for Fc receptors in the spleen and liver to
d. Wiskott‐Aldrich decrease platelet clearance. Resolution of
e. CMV infection thrombocytopenia occurs in 2‐3 weeks and
Explanation: Neonatal thrombocytopenia rules out other congenital causes which
is common, affecting 1%‐3% of newborns should not have normalization of the plate­
and 20%‐30% of premature neonates. Neo­ let count. The answer is d.
natal alloimmune thrombocytopenia (NAIT)
occurs in about 1 in 1000 births and can 3. You are following a 4-year old male
occur in the firstborn unlike Rh‐incom­ patient who is admitted for autoimmune
patibility. Patients typically present at birth hemolytic anemia (AIHA) with a hgb of
with a platelet count <50 × 109/l as com­ 4.5 g/dl with elevated reticulocyte count,
pared to autoimmune thrombocytopenia indirect bilirubin, LDH, AST and a positive
which presents after 1‐3 days and usually DAT (direct Coombs). He is on steroids for
with a higher platelet count. The answer is b. the AIHA. You note that the platelet count
is 34 × 109/l. The most likely diagnosis is
2. Given the above case and the presumed a. Splenic sequestration
diagnosis of NAIT, all of the following are b. Decreased production due to bone
appropriate next management steps EXCEPT: marrow suppression
a. Check a head ultrasound for intracra­ c. Concurrent immune thrombocytopenic
nial hemorrhage purpura (ITP) (i.e, Evans syndrome)
b. Give random donor platelets d. Suppression from steroids
c. Give maternal platelets if available e. Increased destruction due to bystander
d. Give paternal platelets if available hemolysis
e. Give IVIG Explanation: Evans syndrome is the combi­
Explanation: Platelet count should be kept nation of AIHA and ITP. Evans syndrome
>50 × 109/l at birth to prevent intracranial can occur due to transient antibodies which
hemorrhage (ICH). Additionally, all neo­ occur concurrently after infection in a
nates below this level should have a head young child although often signify, espe­
ultrasound to rule out intracranial hemor­ cially in the older child, the presence of an
rhage which can be a presenting finding underlying autoimmune condition. If there
with NAIT. Early exposure to paternal is resolution without recrudescence of cyto­
human platelet antigen (HPA) leads to penias after being weaned off steroids, the
maternal alloimmunization, usually due to patient can be monitored. If there is recur­
HPA1a in the father. The second most com­ rent AIHA and/or ITP, this should prompt a
mon difference is HPA5b and HPA4 should more thorough work up searching for an
be tested in the Asian father. There is a underlying autoimmune process. The
10%‐20% risk of ICH and presentation can answer is c.
be worse with subsequent pregnancies.
The patient should receive random donor 4. You are seeing a 3-year-old in the emer­
platelets and can receive maternal platelets, gency department who presents with “red
if available. Blood from the neonate, mother dots” on the extremities and easy bruising.
154 Chapter 12

You note that the red dots are non‐blanch­ a. Administer IVIG
ing and diagnose these as petechiae. The b. Administer platelets
child is otherwise well with no other phys­ c. Administer steroids
ical findings and without wet bleeding d. Administer RhoGAM
(i.e., mucosal bleeding) although did have a e. Provide reassurance
viral illness a few days prior. The presumed Explanation: In the case of the patient with
diagnosis is immune thrombocytopenic wet bleeding, observation alone is not suffi­
purpura (ITP). Follow up CBC confirms cient and the patient should be treated to
this with normal WBC/diff and hgb with a increase the platelet count. IVIG is the pre­
platelet count of 9 ×109/l. The most appro­ ferred primary treatment. IVIG dilutes the
priate management is: antibody response as well as competes for
a. Administer IVIG the Fc receptors in the spleen and liver. IVIG
b. Administer platelets is generally well tolerated although may lead
c. Administer steroids to headache secondary to aseptic meningi­
d. Administer RhoGAM tis. Families should be advised of this risk as
e. Provide reassurance the patient with thrombocytopenia and
Explanation: Acute ITP is a common pres­ severe headache would subsequently require
entation in young children often secondary a head CT to rule out ICH. Platelets should
to a transient antibody which can occur due not be administered unless with a life‐
to a viral illness. The majority of these threatening bleed as these will be rapidly
patients will clearance of the antibody and consumed. Steroids can be administered by
resolution of the thrombocytopenia over do not have an immediate onset of action.
weeks. In rare cases and in the case of RhoGAM is a potential choice in the Rh+
older children/adolescents, ITP may become patient and anti‐D coats the red blood cells
a chronic condition and may be due to an and thus competes with antibody‐coated
underlying autoimmune condition. Sponta­ platelets at the Fc receptors in the spleen and
neous bleeding can occur when the platelet liver. Thus the major side effect of RhoGAM
count is <50 x 109/l and especially when the is hemolysis and a drop in the hgb as much
platelet count is <20 x 109/l. Spontaneous as 2 g/dl or more which makes it a second­
bleeding may be the presenting sign of ary choice to IVIG. The answer is a.
ITP but in large multicenter analysis, treat­
ment for asymptomatic thrombocytopenia
has not been shown to mitigate this risk. Suggested reading
Therefore, the current recommendation is
to observe patients without active bleeding Bennett, C.M., Neunert, C., Grace, R.F. et al. (2018).
rather than treating a number. Shared deci­ Predictors of remission in children with newly
sion‐making should occur and parental diagnosed immune thrombocytopenia: data
from the intercontinental cooperative ITP study
anxiety be considered when making this
group registry II participants. Pediatr. Blood
decision. The answer is e.
Cancer 65: e26736.
D’Orazio, J.A., Neely, J., and Farhoudi, N. (2013).
5. How would your decision in the above ITP in children: pathophysiology and current
case be altered if the child had mucosal treatment approaches. J. Pediatr. Hematol.
bleeding (i.e., from the mouth, nose, or if Oncol. 35: 1–13.
with hematuria)? Which of the following Fernández, K.S. and de Alarcón, P. (2013).
would be the most appropriate treatment: Neonatal thrombocytopenia. NeoRev 14: e74.
Thrombocytopenia 155

Grainger, J.D. and Thind, S. (2017). A practical Schultz, C.L., Mitra, N., Schapira, M.M., and
guide to the use of eltrombopag in children Lambert, M.P. (2014). Influence of the American
with chronic immune thrombocytopenia. Society of Hematology guidelines on the man­
Pediatr. Hematol. Oncol. 34: 73–89. agement of newly diagnosed childhood immune
Neunert, C., Lim, W., Crowther, M. et al. (2011). thrombocytopenia. JAMA Pediatr. 168: e142214.
The American Society of Hematology 2011 Zdravic, D., Yougbare, I., Vadasz, B. et al. (2016).
evidence‐based practice guidelines for immune Fetal and neonatal alloimmune thrombocyto­
thrombocytopenia. Blood 117: 4190–4207. penia. Semin. Fetal Neonatal Med. 21: 19–27.
13 Evaluation of the Child
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, often providing important
diagnosed with cancer in the United States. clues. Most of the symptoms of childhood
The likelihood of a young adult having a his­ cancer are either due to a mass and its effect
tory of childhood cancer is approximately 1 on the surrounding tissues, invasion of the
in 300. Although cancer remains the leading marrow, or secretion of a substance by the
cause of death in children except for acci­ tumor that disturbs normal function. The
dents, survival continues to steadily increase. most common presenting symptoms can be
In adolescents, cancer deaths are less com­ insidious in onset and include fatigue, fevers,
mon than those caused by accidents, homi­ weight loss, swelling, or a mass. Some of
cide, and suicide. More than 80% of children these may be easily ascribed to more com­
diagnosed with cancer are now expected to mon childhood ailments. A careful family
be cured of their disease. That being said, history should be elicited and include famil­
cancer remains a devastating diagnosis. The ial cancers (though rare in childhood cancer)
initial approach to the child and family must and immune deficiency syndromes. Certain
be with a heightened sensitivity to the emo­ conditions can predispose to malignancy
tional impact. Once a diagnosis of cancer is such as genetic diseases (e.g., Down syn­
suspected, an immediate and thorough eval­ drome, Beckwith–Wiedemann syndrome,
uation should proceed. neurofibromatosis), prior history of a malig­
Initial symptoms of cancer may be some­ nancy (survivor of childhood cancer), or
what elusive, given the subtle and overlap­ radiation therapy.
ping symptoms and signs that may be Timely diagnosis is critical, though can
present in both malignant and nonmalig­ be difficult due to the nonspecific symptoms
nant diseases. Many pediatricians and clini­ and rarity of the diseases. Consideration
cians may only see a new case of childhood should be given to the nature of the
cancer every 5–7 years, and each case may complaint by the child and family, potential
be so unique as to not allow for the increased explanations for the complaint (or lack
awareness of this possibility. Early detection thereof), persistence of symptoms, and lack
and treatment may reduce disease‐related of response to common interventions (see
morbidity and complications. Table 13.1). Delays in diagnosis may be

Handbook of Pediatric Hematology and Oncology – Children’s Hospital & Research Center Oakland,
Third Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
© 2021 John Wiley & Sons Ltd. Published 2021 by John Wiley & Sons Ltd.
158 Chapter 13

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 vomiting Migraine, sinusitis Brain tumor
Lymphadenopathy Infection Lymphoma, leukemia
Bone pain/limping Infection, trauma, Bone tumor, leukemia,
growing pains neuroblastoma
Abdominal mass Constipation, kidney Wilms tumor,
cyst, full bladder neuroblastoma
Extremity mass Cyst, infection, trauma Bone tumor, soft tissue
sarcoma
Mediastinal mass Infection, cyst Lymphoma, leukemia
Pancytopenia Infection Leukemia
Bleeding Coagulation disorders, Leukemia (APL),
platelet disorders, ITP neuroblastoma
Back pain Trauma Leukemia, lymphoma, CNS
tumor, extension of
abdominal tumor
compressing spinal cord
Chronic ear drainage Otitis media/externa Langerhans cell histiocytosis
Feminizing/masculinizing Precocious puberty Adrenocortical carcinoma,
symptoms brain tumor, germ cell
tumor

Abbreviations: APL, acute promyelocytic leukemia (AML M3); ITP, immune thrombocytopenic
purpura; CNS, central nervous system.

related to age of the child (with older chil­ it is the association of other factors, and
dren having an increased risk of delay), type length and severity of these symptoms, that
of cancer, location of tumor, stage, clinical will often provide the most significant
presentation, and experience of the first clues. For example, children that experience
medical consultant. Referral should be made weight loss due to an underlying malig­
to a pediatric oncologist early if there is a nancy may likely have associated findings
suspicion of a cancer diagnosis. including night sweats, pallor, bruising,
Many types of cancer can develop in fevers, or findings on exam such as adenop­
children, though certain types are more athy, hepatosplenomegaly, or a mass/swell­
common in certain age groups. ing. Consideration of the history, physical,
Neuroblastoma and Wilms tumor are more and possible laboratory and imaging find­
often seen in infancy to age 4 years, acute ings are all critical in the initial diagnostic
lymphoblastic leukemia (ALL) in the 1‐ to evaluation and should be performed in a
4‐year age group, and lymphoma and sar­ timely manner.
coma in children aged 10 years and older. Fever is common in childhood, most
As common signs and symptoms may be commonly in association with an infec­
associated with common childhood illness, tious etiology and associated signs and
Evaluation of the Child with a Suspected Malignancy 159

symptoms. Fever is also a common present­ to evaluate for the presence of an intracranial
ing symptom in acute leukemia, lymphoma, mass in children with symptoms suggestive
as well as certain solid tumors (possibly due of a brain tumor. CT may be appropriate as
to tumor necrosis) such as neuroblastoma an initial test in certain settings depending
and Wilms tumor. Malignancy should be on the availability of MRI. CT is a good,
considered if fever is persistent despite rapid tool for the assessment of acute hemor­
appropriate intervention or seen in asso­ rhage or increased intracranial pressure (see
ciation with abnormal findings on the Table 13.2).
complete blood count (CBC) (i.e., cytope­ Lymphadenopathy is a common finding
nias, elevated white count, or circulating on examination in children. Enlarged lymph
blasts), chemistry panel (i.e., elevated lactate nodes are a frequent presenting sign in
dehydrogenase [LDH], liver transaminases, association with malignancies or infection,
uric acid), negative blood cultures, or in and differentiation requires a meticulous
association with clinical findings on exam and systematic approach. Lymph nodes
of organomegaly or adenopathy. become large in response to infection or
Headache is one of the most common infiltration. Generally, a lymph node is con­
complaints in the pediatric population, the sidered enlarged if it measures greater than
majority of which are attributable to nonma­ 10 mm, although inguinal nodes may be
lignant conditions. Although few headaches 15 mm or greater before being considered
are caused by intracranial masses, primary worrisome and epitrochlear nodes are con­
brain tumors or metastases must be ruled cerning once greater than 5 mm. In addition
out when dealing with a patient with to size, other factors to consider in establish­
repeated or persistent headaches. Headaches ing a differential between infection and
are often the first symptom of a brain tumor malignancy include persistence or rate of
and, dependent on location, are frequently
accompanied by subtle findings on neuro­ Table 13.2 Conditions suggesting need
logic examination. History should include for brain imaging in children with headache.
questions to characterize the headache
Presence or onset of neurologic abnormality
including worrisome signs such as recurrent Ocular findings such as papilledema,
morning headache, headache that awakens decreased visual acuity, or loss of vision
the child, intense incapacitating headache, Vomiting that is persistent, increasing in
associated vomiting, or changes in the qual­ frequency, or preceded by recurrent
ity, frequency, and pattern of the headaches. headaches
Associated neurologic symptoms or signs Change in character of headache such as
should warrant an emergent evaluation for a increased severity and frequency
tumor in the central nervous system. These Recurrent morning headaches or headaches
that repeatedly awaken child from sleep
include, but are not limited to, localizing
Short stature or deceleration of linear growth
symptoms such as focal weakness, cranial
Precocious puberty
nerve palsies, ataxia, failure to thrive, and Diabetes insipidus
developmental delays or regression. A thor­ Age 3 years or less
ough neurologic examination, including Neurofibromatosis
evaluation for papilledema, should precede History of acute lymphoblastic leukemia with
radiographic imaging. Other important his­ irradiation of central nervous system
torical points include an assessment of Loss of milestones or an abnormal increase in
changes in visual acuity, behavior, and aca­ head circumference in an infant or toddler
demic performance. MRI is recommended
160 Chapter 13

growth, quality of the node, location and diagnosis of head and neck lumps includes
distribution (i.e., adenopathy that is sym­ lymphadenopathy and congenital malfor­
metric, localized, regional, or disseminated), mations, such as cystic hygroma, thyroglos­
and presence of other signs or symptoms of sal duct cyst, branchial cleft cyst,
infection. Adenopathy in the supraclavicu­ epidermoid cyst, and neonatal torticollis.
lar, axillary, or epitrochlear areas is consid­ Localized adenopathy may be infectious in
ered an abnormal finding. Adenopathy that origin including bacterial causes such as
persists longer than 6 weeks is concerning Staphylococcus aureus, beta‐hemolytic strep,
for malignancy as is a unilateral location. cat‐scratch disease (Bartonella henselae),
Nodes that are hard, nonmobile, and non­ and tuberculous and nontuberculous myco­
tender are worrisome for malignancy. In bacteria, in addition to nonbacterial causes,
certain malignancies, nodes may become such as HIV, Epstein–Barr virus (EBV),
“matted” due to fixation with adjacent tissue cytomegalovirus (CMV), and toxoplasmo­
and each other and are palpated as an sis. The site of lymphadenopathy and age of
enlarged group of nodes in one mass. Nodes the child may also provide clues as to con­
associated with fluctuance, tenderness, cern for underlying malignancy. Young chil­
warmth, or overlying erythema are more dren with head and neck adenopathy may
consistent with infection. However, these have neuroblastoma, rhabdomyosarcoma
are only generalizations, as rapidly growing (RMS), non‐Hodgkin lymphoma, or ALL.
malignant nodes may be tender and infec­ Older children with Hodgkin and non‐
tious nodes may be quite firm and Hodgkin lymphoma may present with iso­
nonmobile. lated adenopathy in this area.
Important historical points to elicit The physical examination of the child
include: with lymphadenopathy includes an
●● Duration of lymphadenopathy or local­ assessment of the size, location, and quality
ized mass of the node(s). A full examination should
●● Presence of a recent infectious illness include assessment of the skin and
●● Skin lesions, cuts, or abrasions (and rela­ mucocutaneous tissues draining to the
tionship to nodal drainage patterns) particular node in question, and evidence of
●● Recent immunizations other signs related to an underlying disease
●● Medications process such as hepatomegaly and
●● Animal contact (e.g., cat scratch, rodent splenomegaly. Nodes should be measured in
bite, tick bite) largest diameter, the quality should be
●● Recent transfusion assessed as to mobility, firmness, tenderness,
●● Travel and overlying skin changes, and these
●● Possible sexually transmitted infection findings should be documented. Children
●● Presence of symptoms such as fever, with mildly enlarged nodes should be
arthralgias, weight loss, or night sweats monitored with frequent examinations, and
when not associated with malignancy, most
The differential diagnosis of lymphadenop­ of these nodes will revert to normal size.
athy includes infectious etiologies (viral, If infection is considered the cause of the
bacterial, spirochetal, and protozoan), adenopathy, as in localized cervical adenitis,
­connective tissue disease, hypersensitivity it is reasonable to treat the patient with a
states, lymphoproliferative disorders, 2‐week course of antibiotics. Dependent on
immunodeficiency states, storage diseases, the history, physical, and clinical suspicion, a
and malignancy. In infants, the differential particular antibiotic regimen can be chosen
Evaluation of the Child with a Suspected Malignancy 161

in addition to sending serology or further or as a result of antibiotic treatment (see


testing for likely organisms. A baseline CBC Figure 13.1).
with differential and peripheral blood smear For the large node (i.e., ≥2.5 cm in size) or
should be done if there is any concern for a in the patient without response to antibiot­
diagnosis other than infection. The patient ics, further evaluation is required. Patients
should be seen again following completion with adenopathy in the posterior neck or
of the antibiotics and if the node has not supraclavicular areas, especially if there is no
changed or has increased in size, considera­ history of recent infection, also warrant a
tion should then be given to excisional more immediate evaluation. Additional
biopsy. Many cases of infection‐related ade­ studies should include a purified protein
nopathy resolve spontaneously in 2–6 weeks derivative (PPD) skin test (or QuantiFERON

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
Growing in size or Infection confirmed or likely
Node ≥ 2.5 cm or Node < 2.5 cm
Hard, nonmobile or Empiric antibiotic therapy
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).
162 Chapter 13

gold), chest radiograph, CBC with differen­ Storage disease: Gaucher, Neimann–Pick,
tial (if not already done), chemistries includ­ acid sphingomyelinase deficiency, and
ing LDH and uric acid, and serologies as mucopolysaccharidoses.
indicated by history and examination. An Malignancy: leukemias as well as Hodgkin
excisional biopsy should be done on enlarg­ and non‐Hodgkin lymphoma.
ing, matted, or persistently large nodes or if
adenopathy is also seen on chest radiogra­ A detailed history should be obtained to
phy. It is recommended that excisional gain clues as to the etiology of the enlarged
biopsies (i.e., removal of intact node) be per­ spleen. The patient should be questioned
formed when malignancy is suspected to regarding current or recent infectious symp­
evaluate the architecture of the node in addi­ toms, fevers or rigors (e.g., in subacute bacte­
tion to the cellular infiltrate. The largest rial endocarditis, infectious mononucleosis,
node should be biopsied when possible, with and malaria), jaundice (with hemolytic
avoidance of the upper cervical and inguinal anemia or liver disease), abnormal bleeding
areas. Studies to be performed on the tissue or bruising (malignancy), travel to endemic
include Gram stain and culture (bacterial, areas (malaria), trauma (splenic hematoma),
mycobacterial, viral, and fungal); histology and family history (hemoglobinopathies,
and immunohistochemistry for suspected thalassemia, and hereditary spherocytosis
malignancy; and if malignancy is confirmed, with prior splenectomy or cholecystectomy).
flow cytometry and specific cytogenetic test­ The physical examination should include a
ing for further classification. measurement of the spleen size (centimeters
Splenomegaly is the finding of a palpable below the mid‐costal margin), consistency,
spleen edge on examination. A 1–2‐cm and presence of tenderness (which suggests
splenic tip is found in 30% of full‐term neo­ rapid increase in size) in addition to the
nates and in as many as 10% of healthy chil­ presence of adenopathy or hepatomegaly.
dren. Approximately 3% of healthy college The vitals should be reviewed for evidence of
students have palpable spleens. Therefore, fever or hypotension and the skin assessed
this finding on a routine physical examina­ for cutaneous bleeding. Other considera­
tion of an otherwise healthy child should tions include evaluation for stigmata of
not create great concern. Children with specific disease states including jaundice,
other signs of systemic disease, however, cardiac murmurs, arthritis, as well as spe­
should have their splenomegaly evaluated. cific findings of endocarditis including Roth
Splenomegaly is associated with many spots (retinal hemorrhages), Janeway lesions
disease states, both congenital and acquired: (nontender hemorrhagic lesions on the
palms/soles), Osler nodes (tender microem­
Hemolytic anemia: hereditary spherocytosis, boli on the fingers and toes), and presence of
thalassemia, splenic sequestration in neurologic or cognitive concerns (metabolic
sickle cell disease, and autoimmunity. storage diease).
Immunological disease: common variable The laboratory and imaging assessment
immune deficiency, connective tissue dis­ is determined based on suspicion of the
orders, and autoimmune lymphoprolif­ underlying etiology of splenomegaly.
erative disease. Consideration should also be given to a
Infection: viral (EBV, CMV, HIV, hepatitis), secondary effect of hypersplenism in which
bacterial (tularemia, abscesses, tubercu­ the child may have cytopenias due to a large
losis, infective endocarditis), spirochetal, spleen. Persistent splenomegaly should be
protozoan, and fungal. fully investigated as follows:
Evaluation of the Child with a Suspected Malignancy 163

CBC: evaluate red cell indices, reticulocyte lower extremity pain occurring in the after­
count, platelet count, white blood cell noon, evening, or night, and affect children
count, and review peripheral blood between 3 and 14 years of age. These can
smear (all to help rule out hematologic occur most commonly on a weekly or
disorders such as hemolytic anemia; monthly basis (and, much less frequently
membrane disorders with spherocytes or daily), and are relieved with massage in the
elliptocytes; increased red cell indices majority of cases. Pain is otherwise usually
and anemia in thalassemia; atypical lym­ due to bone, bone marrow, or nerve infiltra­
phocytes as in EBV; leukemic blasts; and tion. Back pain in young people is pathologic
cytopenias secondary to leukemia or and may be due to a tumor in the spinal cord
hypersplenism). or one causing external compression such as
Infection: blood culture, viral studies, PPD neuroblastoma, lymphoma, Ewing sarcoma,
(or QuantiFERON gold), thick and thin RMS, or a leukemic chloroma. This finding
smear, and serologies to rule out EBV, should prompt an MRI of the spine, as these
CMV, HIV, histoplasmosis, tuberculosis, conditions typically result in no abnormali­
and malaria. ties on plain film.
Hemolytic evaluation: CBC, reticulocyte Patients with primary bone tumors often
count, direct antiglobulin test (Coombs), present with localized pain, frequently in
haptoglobin, serum bilirubin, LDH, red association with a growing mass. Pain may
cell enzyme assays (G6PD, pyruvate be attributed to recent mild trauma or
kinase deficiency), osmotic fragility growing pains. In approximately 5–10% of
testing or ektacytometry, and urinalysis. cases, patients sustain a pathological fracture
Liver disease: complete metabolic panel, after seemingly mild trauma due to
coagulation studies, hepatitis panel, α‐1 infiltration of the periosteum and weakening
antitrypsin, ceruloplasmin (Wilson dis­ of the bone. Bone pain is typically a
ease), and 24‐hour urine copper. presenting symptom in patients with
Connective tissue disease: erythrocyte osteogenic sarcoma and Ewing sarcoma.
sedimentation rate (ESR), complement Langerhans cell histiocytosis very often
(C3, C4, CH50), antinuclear antibody, involves bone and may present with
and rheumatoid factor. localized bone pain anywhere in the body,
Infiltrative diseases: bone marrow aspirate often with overlying soft tissue swelling.
and biopsy (looking for blasts, storage Diffuse or multifocal bone pain is a
cells) and enzyme study for Gaucher common presenting symptom in acute leu­
(glucocerebrosidase). kemia. It is reported in more than 25% of
Lymph node biopsy: can be done if performed patients diagnosed with ALL due to mar­
with coexistent lymphadenopathy. row crowding by leukemic cells and is seen
Imaging: volumetric and heterogeneity less commonly in acute myelogenous leuke­
assessment with ultrasound, CT, or MRI; mia (AML). Patients will often complain of
liver–spleen scan with 99mTc‐sulfur col­ back or leg pain that is persistent and
loid for functional analysis; can also send increasing in intensity and very young chil­
pit count for pitted red blood cells. dren may become irritable with refusal to
walk or to participate in normal activities.
Bone or joint pain is unusual in children Musculoskeletal pain in children is often
and adolescents except when associated with diagnosed as arthritis or bone or joint
trauma. Growing pains are a common com­ infection. Pain may be asymmetric and
plaint and consist of bilateral (80% of cases) often children present with a limp with pain
164 Chapter 13

seemingly disproportionate to the findings Cytopenias or abnormalities on the


on examination. In contrast to children with peripheral blood smear are suspicious for
arthritis, children with leukemia will have a marrow infiltrative process, primarily
worsened pain at night, severe pain that may ­leukemia. Isolated anemia, leukopenia, or
shift to other locations, no morning stiffness thrombocytopenia can occur in leukemia
or swelling, and may have associated consti­ but much more commonly the patient will
tutional symptoms such as weight loss and present with bicytopenia or pancytopenia.
night sweats. Associated laboratory findings Pancytopenia may also indicate the lack of
that may suggest malignancy include an ele­ hematopoietic cell production as in aplastic
vated ESR, elevated serum LDH or uric acid anemia or as caused by the proliferation of
anemia, thrombocytopenia, neutropenia, malignant cells in the marrow with result­
leukopenia, or leukocytosis. Any of these ant crowding. The anemia is frequently
findings should prompt an examination of characterized as one of chronic disease (i.e.,
the bone marrow. normochromic with low reticulocyte count).
Bone pain may also be a direct result of Leukocyte counts are variable at presentation
bony metastatic disease or marrow infiltra­ in acute leukemia and may be normal,
tion secondary to other tumors including decreased, or elevated. The cell differential,
neuroblastoma, RMS, Ewing sarcoma, and however, is likely to show neutropenia and
non‐Hodgkin lymphoma. Localized bone the peripheral blood smear will likely,
pain warrants radiographic evaluation (two‐ though not always, demonstrate blasts or
view plain radiographs) for the assessment of immature cells (see Table 13.3). Unless
a lesion or leukemic changes. Patients with metastasis to the marrow has occurred,
leukemia and bone pain may show evidence leukopenia and thrombocytopenia are
of osteopenia, lytic lesions, metaphyseal rarely associated with extramedullary
bands, periosteal new bone or sclerotic malignancies such as neuroblastoma, Ewing
lesions. Malignant bone tumors can show sarcoma, or RMS.
characteristic onion skinning or a sunburst Patients with bicytopenia and pancytope­
phenomenon. The onion skin appearance is nia require bone marrow aspiration, and
a result of repetitive periosteal reactions, possibly bone marrow biopsy, for a suspected
with layers of calcium accumulating with diagnosis of a marrow infiltrative process
growth beyond the periosteum and is classi­ such as acute leukemia or aplastic anemia.
cally seen in Ewing sarcoma. The sunburst The situation can be more complex in the
phenomenon is common in osteosarcoma case of a single depressed lineage. Children
and occurs as a result of deposition of cal­ with persistent or worsening normocytic,
cium in blood vessels by malignant osteo­ normochromic anemia without manifesta­
blasts that radiate perpendicular from the tions of hemolysis, clinical suspicion of tran­
tumor. MRI is the imaging of choice for eval­ sient erythroblastopenia of childhood, or
uation of a suspected bone tumor and can renal disease with low erythropoietin pro­
show the extent of medullary and extramed­ duction should have diagnostic bone mar­
ullary soft tissue involvement, as well as skip row studies performed.
lesions (disease in the same bone not con­ Children with leukemia may also present
tiguous with the primary lesion). Children with an elevated white blood cell count, some­
with bone tumors also need whole body times with marked elevation over 100 × 109/l
technetium bone scanning to screen for dis­ (i.e., hyperleukocytosis). Leukemoid reac­
tant lesions as well as CT imaging of the tions (a white blood cell count >50 × 109/l
chest to evaluate for metastatic involvement. often associated with immature forms [i.e.,
Evaluation of the Child with a Suspected Malignancy 165

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, aspartate aminotransferase,
alanine aminotransferase (AST, ALT), alkaline phosphatase, 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 and possibly bone marrow biopsy
Specimens for morphology, immunophenotyping, and cytogenetics
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 or per institutional protocol for initial study)
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 thrombo­
plastin 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 simplex virus.

bands] or lymphocytosis) can be seen in Any cytopenia associated with unexplained


infants with Down syndrome (i.e., transient lymphadenopathy, splenomegaly, hepatomeg­
myeloproliferative disorder) or in cases of aly, anterior mediastinal mass, or bone pain.
severe bacterial infection such as pertussis in
the young child. Bleeding as a presenting sign in cancer is
Bone marrow examination with aspirate usually related to severe thrombocytopenia
and/or biopsy is indicated in the following and occurs commonly in children with
situations: acute leukemia. Manifestations typically
Significant depression of one or more involve mucocutaneous tissues with clinical
peripheral blood lineages (white cells, red signs including petechiae, purpura, epistaxis,
cells, platelets) without an obvious infec­ and menorrhagia. Patients with APL (AML
tious cause. M3) are at risk for severe bleeding at pres­
Presence of circulating blasts on the entation due to underlying coagulation
peripheral blood smear. abnormalities and DIC, whereas Wilms
166 Chapter 13

tumor patients may have acquired von hernia. Malignant tumors, especially lym­
Willebrand disease (aVWD) at presentation phoma and leukemia, may have rapid growth
increasing bleeding risk. Coagulopathy has rates and quickly lead to compromise with
also been seen at presentation in T‐cell acute presentation of orthopnea, superior vena
lymphoblastic leukemia, lymphoma, and cava/superior mediastinal syndrome, airway
neuroblastoma. Bleeding may also be due to obstruction, dysphagia, and symptoms of
defective platelet function and a medical increased intracranial pressure due to
cause of impairment (such as use of ibu­ decreased cerebral venous return. The mass
profen or other NSAIDs) should be consid­ may cause a pericardial effusion or directly
ered. Evaluation should consist of a CBC, obstruct cardiac outflow leading to cardiac
reticulocyte count, review of the peripheral compromise. See Chapter 14 for assessment
blood smear, coagulation studies with PT, and management.
international normalized ratio (INR), PTT, Middle mediastinal masses are likely
and fibrinogen, as well as a bone marrow malignant. Infections should be in the dif­
evaluation. ferential diagnosis with consideration for
Mediastinal masses may lead to com­ tuberculosis or histoplasmosis in addition to
pression of respiratory, vascular, or other structural anomalies such as pericardial
structures and can range from an asympto­ cysts, bronchogenic cysts, or esophageal
matic incidental finding to significant com­ lesions. Malignant tumors common in this
promise and an emergent situation. location include Hodgkin lymphoma and
Common symptoms include orthopnea (i.e., nodal masses of neuroblastoma, RMS, and
inability to lie flat), cough, shortness of germ cell tumors. Direct extension of an
breath, fatigue, hoarseness, and wheezing. abdominal mass may also present in the
Most mediastinal masses in children are middle mediastinum.
malignant and often are associated with Posterior mediastinal masses are
adenopathy, abnormal cell counts, and pos­ generally neurogenic in origin and include
sibly neck swelling. Imaging with chest radi­ benign and malignant tumors. These
ography and chest CT yields information include ganglioneuroma, neurofibroma,
with respect to location in the mediastinum lymphoma, Ewing sarcoma, and
and potential for cardiac compromise. There neuroblastoma. Most of these lesions are
are three anatomic compartments of the asymptomatic but may present with
mediastinum, and location may provide symptoms of spinal cord compression such
clues to etiology. Evaluation of the child as pain or focal neurological signs.
should include laboratory studies (i.e., CBC, A palpable abdominal mass is one of the
complete metabolic panel, coagulation most common presenting findings of a
panel), and the physical status and stability malignant solid tumor in children.
of the patient will help determine next steps Nonmalignant etiologies include impacted
such as feasibility of safely performing a stool, intussusception, abdominal aorta, a
bone marrow aspirate and lymph node distended bladder, hydronephrotic kidneys,
biopsy. and pregnancy.
Anterior mediastinal masses are more The age of the child can provide a clue
typically seen in older children and adoles­ to diagnosis. In the newborn, an abdomi­
cents and frequently are associated with lym­ nal mass is most likely to be a congenital
phoma, T‐cell leukemia, thymic tumors, abnormality of renal origin. The most
thyroid tumors, and some benign tumors common malignant tumors in young chil­
(teratomas, lipomas, and angiomas) or dren are neuroblastoma and Wilms tumor.
Evaluation of the Child with a Suspected Malignancy 167

Children with Wilms tumor most often examination). Care should be taken to pal­
are well‐appearing and the mass is an inci­ pate the mass gently and limit the number of
dental finding noted by a family member examiners. Imaging will also help determine
or during a well‐child check. Unlike size and location. A careful general physical
Wilms tumor, neuroblastoma will often examination is vital, as many tumors may
present with evidence of spread and sys­ have associated signs and symptoms or
temic symptoms including weight loss, underlying syndromes. Aniridia, hemihy­
fever, and bone pain. In older patients, the pertrophy, and genitourinary abnormalities
mass may be related to leukemia or lym­ have been reported in association with
phoma with enlargement of the spleen Wilms tumor. Subcutaneous nodules (typi­
and liver. The most common lymphoma cally bluish), periorbital ecchymoses, opso­
in children is Burkitt lymphoma which clonus–myoclonus, and presence of
may present as a rapidly enlarging abdom­ organomegaly are seen with neuroblastoma.
inal mass leading to pain and obstructive Signs of precocious puberty may be seen
symptoms (gastrointestinal and urinary with tumors involving the liver, adrenal
tracts) in association with metabolic glands, or gonads (i.e., germ cell tumors).
derangements from tumor lysis. Burkitt The neurological examination may show
and other lymphoma, as well as primary evidence of a Horner’s syndrome with apical
gastrointestinal tumors, may also occur in tumors (often neuroblastoma) or spine
the ileocecal area and serve as a lead point compression with large abdominal masses.
for intussusception. See Table 13.4 for the workup of a child with
The history is important to determine if an abdominal mass. A surgical consultation
the symptoms are related to the mass. A should be obtained and the decision made
careful genitourinary history should be whether to obtain a biopsy or perform a
obtained to determine if the mass may be of resection of the mass. Surgical staging is
renal origin. Historical points may provide done by assessing tumor margins, nodal
suspicion of catecholamine production such involvement, presence of locally invasive or
as flushing, palpitations, diarrhea, and distant disease, and for possible tumor spill­
sweating (very rare). Constitutional age (see Chapters 18 and 19).
symptoms such as failure to thrive, fever,
night sweats, and sudden weight loss should
lead one to suspect a disseminated process Case study for review
such as neuroblastoma in young children or
lymphoma in older children and adolescents. A 14‐year‐old female presents with progres­
A meticulous and careful physical exam­ sive bilateral neck swelling in the setting of
ination of the child should be done by first anemia and fatigue.
attempting to have the child relax. When Six months prior to presentation, her
examining the abdomen, keep in mind the physician sent labs for a complaint of fatigue
normal structures that may be present such and noted the following: hgb 8.3 g/dl, WBC
as the liver or spleen edges, kidneys, aorta, 7.8 × 109/l, platelets 517 × 109/l, mean cor­
sigmoid colon, stool, or spine. A rectal puscular volume (MCV) 81, neutrophils
examination and pelvic/vaginal examina­ 74%, lymph 10%, serum iron 14, transferrin
tion may be indicated but should be per­ saturation 7%, ferritin 441, fibrinogen 788,
formed only by an experienced practitioner INR 1.3, PTT 42. Due to suspected iron
and after obtaining laboratory studies deficiency anemia, she was placed on an oral
(should not be neutropenic for a rectal iron supplement.
168 Chapter 13

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/pelvic 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, α‐fetoprotein, β‐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, metaiodo­
benzylguanidine; CBC, complete blood count; LDH, lactate dehydrogenase; BUN, blood urea
nitrogen; VMA, vanillylmandelic acid; HVA, homovanillic acid; HCG, human chorionic
gonadotropin; ESR, erythrocyte sedimentation rate.

Six weeks prior to presentation, she Past history included travel to Mexico
noted increasing bilateral neck swelling, 4 years previously. Her brother is in the
without tenderness or overlying erythema. army and recently returned home after
Two weeks prior to presentation, she 3 years in Germany.
traveled with her family to Mexico and after a. What elements of the history and
several days felt dizzy and more fatigued. evaluation to date are concerning and
She was seen in a local hospital and a CBC what additional information should you
was essentially unchanged. CT imaging seek?
showed hepatosplenomegaly and diffuse The patient has a normocytic anemia, mild
adenopathy involving anterior cervical, elevation of the platelet count, a predomi­
mediastinal, and retroperitoneal nodes. She nance of neutrophils on the differential, and
was given a blood transfusion and her symp­ elevated ferritin and fibrinogen. These are
toms improved. She also underwent a fine suggestive of a chronic inflammatory state.
needle aspiration of an enlarged right ante­ There is evidence of iron deficiency based
rior cervical node, and the pathology on the low transferrin saturation, but this
showed follicular hyperplasia. is more likely secondary to macrophage
Evaluation of the Child with a Suspected Malignancy 169

trapping secondary to inflammation as in palpation with many small (<1 cm), non­
chronic disease states. The diffuse adenopa­ tender, mobile, rubbery nodes detected in
thy and hepatosplenomegaly suggest this is a the anterior and posterior cervical chains
systemic illness and could possibly represent extending to the supraclavicular regions, left
infection, malignancy, immune deficiency, side > right. The largest palpable node meas­
or autoimmune disease. Of note, a fine nee­ ures 1.5 cm in the left supraclavicular fossa
dle aspirate is not an ideal method to obtain (which the patient notes to be the first lump
tissue for diagnostic purposes, as nodal she felt). Axillary and inguinal adenopathy
architecture often provides important clues are also noted. There is mild diffuse tender­
and diagnostic information (also in more ness of the abdomen in the bilateral upper
sufficient quantity). quadrants. You are unable to appreciate
The history should seek information on organomegaly due to the obese abdomen,
travel, ill contacts (including TB exposure), even with percussion. She has a tender drain­
the presence of fever or infectious symp­ ing lesion (serosanguinous) on the upper
toms, respiratory symptoms, weight loss, inner right thigh.
night sweats, and familial diseases. Labs: UA neg, complete metabolic panel
Given the diffuse nature of her adenopa­ (CMP) wnl, uric acid nl, LDH 708, INR 1.2,
thy, imaging with a CXR should be obtained APTT 38.2, hgb 9.5 g/dl, WBC 6.8 × 109/l,
to determine if mediastinal adenopathy (or a platelets 368 × 109/l, MCV 81, polymorpho­
mass) may be present with imminent danger nuclear neutrophils (PMNs) 62%, bands 6%,
(i.e., compromise to heart, trachea, or great lymphs 14%, retic 0.6%; DAT neg, beta‐
vessels). human chorionic gonadotropin (βHCG)
Upon questioning, she has an intermit­ neg.
tent nonproductive cough several times a CXR: prominent anterior mediastinal
day. She has not had fever or infectious and right paratracheal lymphadenopathy,
symptoms and she has not noted a change in no mediastinal mass.
breathing pattern or endurance. She gets b. What considerations are in the differ­
sweaty at night but not drenching or need­ ential diagnosis and what studies should
ing to change her linens. No known sick you seek?
contacts, GI symptoms, pain, or arthralgia. Consideration should be given to an infec­
Over the past 5–6 months she has lost 20 lbs tious etiology that could explain the chro­
unintentionally. Her appetite and food nicity and progressive nature of the illness
intake have not changed. Her menses are and include (with appropriate testing
regular. No unusual bleeding or easy bruis­ modality): HIV (ab), CMV by polymerase
ing. She has been attending school, missing chain reaction (PCR), EBV (serology and
only the days her family traveled to Mexico. PCR), coccidioides (IgM, IgG), and TB
Several days prior to presentation, she also (PCR, acid fast bacillus‐AFB). The prior
noted a small tender bump on the inner negative PPD and no history of ill contacts
aspect of her right thigh and noted it bled (i.e., coughing contacts) suggest TB is less
the day before. likely but should give consideration to
Immunizations are up to date; four sib­ obtaining a QuantiFERON gold test.
lings and parents are healthy without Malignancy is a major consideration
known autoimmune disease. Recent PPD is given the degree of enlarged nodes, organo­
negative. megaly, as well as weight loss. The elevations
On exam she is well‐appearing and of ferritin and fibrinogen, both acute phase
comfortable. She is obese. Neck swelling is reactants, can be elevated with malignancy.
not obvious but adenopathy is noted on An elevated LDH can support this diagnosis
170 Chapter 13

as well. A diagnosis of lymphoma especially cancer in those receiving radiation to the


Hodgkin disease should be ruled out (given mediastinum.
the chronicity). An excisional lymph node
biopsy should be performed. The left supra­
clavicular node appears to be the ideal lesion Multiple choice questions
to remove (in its entirety) given the patho­
logic site, and that it was the first lump felt 1. You are seeing a 6-month-old male
and is the largest. This is a surgically acces­ patient in your clinic and you palpate an
sible site with an expected short healing enlarged liver/mass. There is no hyperten­
process. Night sweats, fevers, and/or unin­ sion or hematuria. Follow up CBC reveals a
tentional weight loss are all considered “B” microcytic anemia with thrombocytosis.
symptoms of lymphoma, and therefore her The most likely diagnosis is:
unintentional weight loss is suggestive. a. Neuroblastoma
The physical exam is less suggestive of an b. Hepatoblastoma
autoimmune process and this evaluation c. Wilms tumor
can be deferred until after the biopsy. d. Embryonal sarcoma of the liver
The patient had an infectious evaluation e. Abdominal lymphoma
with negative studies for HIV, CMV, EBV, TB, Explanation: Hepatoblastoma, although rare,
and cocci. Additionally, the draining lesion is the most common liver tumor of infancy
on the left thigh was incised and drained and early childhood. Hepatoblastoma has an
and grew mixed anaerobic bacteria. It was increased risk in premature, very low birth
packed and the patient received a 7‐day weight infants. Hemihypertrophy and
course of cephalexin with full resolution. overgrowth syndromes are a risk factor for
The pathology on the excised lymph hepatoblastoma as well as Wilms tumor
node reveals nodular sclerosing classical (less likely neuroblastoma and sarcoma and
Hodgkin disease, with effaced nodal archi­ rarely adrenocortical carcinoma). Elevation
tecture, replaced by histiocytic cells sepa­ of alpha‐fetoprotein is diagnostic for hepa­
rated by thick fibrous bands. This type of toblastoma. Thrombocytosis is commonly
pathology is best appreciated in a fully seen due to thrombopoietin (TPO) produc­
excised node as opposed to a fine needle tion by the hepatoblastoma tumor cells and
core specimen. Occasional Reed–Sternberg can be a clue to diagnosis. The answer is b.
cells are noted as well.
c. What are the next steps for the patient 2. You are seeing a 4-year-old female admit­
at this time? ted for work up of an abdominal mass. She
Given the diffuse adenopathy, organomeg­ had a CT of the chest, abdomen and pelvis
aly and pathology findings, as well as weight which showed her disease was limited to
loss, she is at a minimum stage IIIB. the abdomen/pelvis. She underwent open
Complete staging will include further imag­ biopsy and has been having some oozing at
ing with positron emission tomography‐ the biopsy site. You are concerned about
computer tomography (PET‐CT) (full acquired von Willebrand disease. Her most
body) and bilateral bone marrow aspirate likely underlying tumor is:
and biopsy. A number of chemotherapeutic a. Neuroblastoma
regimens offer excellent outcomes and b. Hepatoblastoma
potential radiation sparing, which must be a c. Wilms tumor
significant consideration especially in a d. Rhabdomyosarcoma
female patient given the high risk of breast e. Osteosarcoma
Evaluation of the Child with a Suspected Malignancy 171

Explanation: Although acquired von in a basilar skull fracture. Opsoclonus myo­


Willebrand disease (AVWD) has been clonus ataxia syndrome (OMAS) and VIP
reported in other tumor types, it has been hypersecretion are both paraneoplastic syn­
most commonly reported in Wilms tumor. dromes seen with neuroblastoma. In OMAS,
AVWD has also been reported in other it is thought that antibodies to the tumor
miscellaneous conditions including car­ cells attack the cerebellum. Brain and CSF
diovascular disorders, autoimmunity and involvement generally do not occur in
hypothyroidisim. AVWD can be similar to neuroblastoma although the tumor may
type I or type IIA VWD and therefore the wrap around or even impinge upon the
suspected patient should have testing to spinal column. The answer is a.
include von Willebrand factor (VWF)
antigen, ristocetin cofactor assay and high 4. Prognosis of neuroblastoma is based
molecular weight multimers. Initiation of on multiple factors. All of the following
therapy usually rapidly resolves the AVWD, are important baseline prognostic factors
thought to be due to absorption of VWF EXCEPT:
antigen by tumor cells or, specifically in the a. Age
case of Wilms tumor, due to secretion of b. Stage of disease
hyaluronic acid. The answer is c. c. N‐Myc status
d. Urine HVA and VMA
3. Neuroblastoma can present with many e. Histology
different clinical findings as well as paraneo­ Explanation: Although urine HVA and
plastic syndromes. All of the following are VMA are important diagnostic modalities,
potentially consistent with neuroblastoma they are not baseline prognostic factors.
EXCEPT: They are important as markers of response
a. Seizure disorder in addition to imaging response. Patient age
b. Opsoclonus myoclonus ataxia is a very important prognostic marker as
syndrome younger children do better and those <1
c. Secretory diarrhea secondary to VIP year of age may be diagnosed with stage IVS
hypersecretion neuroblastoma which can often resolve
d. Horner’s syndrome without treatment. Stage of disease is also an
e. Raccoon eyes important prognostic marker as stage III
Explanation: Neuroblastoma occurs due to and IV patients (based on the older INSS
lack of maturation of neural crest cells and staging system) are harder to treat and
can therefore occur anywhere along the require more intensive therapy. Generally
sympathetic chain. Infants may present with these patients also harbor the N‐Myc genetic
an incidental finding of neuroblastoma mutation which is also correlated with poor
which often regresses spontaneously and prognosis. Finally, histology as well as DNA
does not require any therapy. Older infants index are utilized as prognostic markers.
and children require therapy dependent on The answer is d.
the extent of disease (i.e., stage) as well as
presence of N‐Myc which is a poor prognos­ 5. You are seeing a patient with rhabdomyo­
tic genetic marker. Low stage neuroblastoma sarcoma. All of the following are important
may present along the cervical chain leading baseline prognostic factors EXCEPT:
to Horner’s syndrome or heterochromia. a. Extent of disease (i.e. disease stage)
Retrobulbar involvement can lead to prop­ b. Loss of heterozygosity (LOH) at 1p
tosis or raccoon eyes, similar to what is seen and 16q
172 Chapter 13

c. Alveolar vs. embryonal histology disease. Certain sites of disease including


d. Presence of the fusion protein PAX3 the head and neck as well as the genito­
and FOXO1 urinary (GU) (non‐bladder, non‐prostate)
e. Site and resectability of primary regions do better. LOH of 1p and 16q is a
tumor prognostic marker in Wilms tumor. The
Explanation: Younger patients with rhab­ answer is b.
domyosarcoma (RMS) tend to have the
embryonal histologic subtype which has
a better prognosis than the alveolar sub­ Suggested reading
type. Alveolar RMS is more likely in older
patients and is more likely to present with Allen‐Rhoades, W. and Steuber, C.P. (2015).
metastatic disease. Alveolar histology is Clinical assessment and differential diagnosis
confirmed with fusion of PAX3 (or rarely of the child with suspected cancer. In:
Principles and Practice of Pediatric Oncology,
PAX7) with FOXO1. Patients with alveolar
7e (eds. P.A. Pizzo and D.G. Poplack), 101–
histology but without the PAX3/FOXO1
112. Wolters Kluwer.
fusion protein do better due to some overlap Siegel, D.A., King, J., Tai, E. et al. (2014). Cancer
in histology with the embryonal subtype. incidence rates and trends among children
Patients with smaller, more easily resectable and adolescents in the United States, 2001‐9.
tumors do better as do patients with localized Pediatrics 134: e945–e955.
14 Oncologic
Emergencies
The prognosis for children diagnosed with tumor involvement and compress the SVC.
cancer has steadily improved over the past Adjacent coronary and collateral vessels can
50 years, and it is now estimated that more become clotted. Compression, clotting, and
than 80% of children newly diagnosed with edema lead to diminished blood flow and
cancer will ultimately be cured of their compromised cardiac return. Additionally,
disease. Given this positive outlook, it the trachea and right mainstem bronchus
becomes even more important that life‐ are less rigid in children as compared to
threatening complications arising either as a adults potentially impacting air flow as well.
result of the patient’s cancer diagnosis or the Superior vena cava syndrome (SVCS)
treatment being provided be promptly refers to the signs and symptoms resulting
recognized and appropriately treated. from the compression or obstruction of the
Oncologic emergencies can be SVC caused by an anterior mediastinal
categorized based on their pathogenesis, mass. These include orthopnea, headache,
including emergencies caused by space‐ facial swelling, dizziness or fainting, sudden
occupying lesions, those caused by pallor, and exacerbation of symptoms with
abnormalities of blood and blood vessels, the Valsalva maneuver. The physical
and metabolic emergencies. examination often reveals a plethoric,
edematous face and neck, jugular venous
distension, papilledema, and pulsus
Emergencies caused by space‐ paradoxus. Blood pressure changes, pallor,
occupying lesions and even cardiac arrest can result from
postural changes. Superior mediastinal
Superior vena cava syndrome syndrome (SMS) is the combination of
and superior mediastinal SVCS and tracheal compression that leads to
syndrome symptoms of cough, dyspnea, air hunger,
The superior vena cava (SVC) is a thin‐ and wheezing. These names are often used
walled vessel with low intraluminal pressure, interchangeably. Examination often reveals
prone to thrombosis, and surrounded by decreased breath sounds, wheezing, stridor,
thymus and nodes that drain the right side or cyanosis. Affected children are often
and lower left side of the chest. Part of the incredibly anxious as well.
SVC is also in the pericardial reflection. Malignant tumors are the most common
Lymph nodes, thymus, and the pericardium primary cause of SVCS. The most common
may become enlarged from infection or cause is non‐Hodgkin lymphoma (NHL;

Handbook of Pediatric Hematology and Oncology – Children’s Hospital & Research Center Oakland,
Third Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
© 2021 John Wiley & Sons Ltd. Published 2021 by John Wiley & Sons Ltd.
174 Chapter 14

usually lymphoblastic lymphoma or diffuse widening, tracheal deviation or compression,


large B‐cell lymphoma). Other malignant and possibly pleural effusions.
causes include Hodgkin lymphoma, T‐cell 4. CT scan of the chest: This helps delineate
acute lymphoblastic leukemia (T‐cell ALL), distortion of normal anatomy, identify
malignant teratoma or germ cell tumor, thy­ tracheal compression, and allow assessment
moma, neuroblastoma, rhabdomyosarcoma, of mediastinal mass width. This may be
or Ewing sarcoma. A secondary cause can be done in the prone or somewhat sitting
thrombosis of the major vessels caused by position if the patient is unable to lie supine.
the presence of a central venous catheter. 5. Tissue diagnosis with least invasive
Nonmalignant causes include mediastinal procedures, with no sedation, and local
granulomas (histoplasmosis), aortic aneu­ anesthesia only: A biopsy is often essential
rysms, vascular thrombosis complicating for diagnosis, but must only be performed if
cardiovascular surgery for congenital heart safe to do so. General anesthesia may
disease, shunting for hydrocephalus, cathe­ cause cardiovascular and/or respiratory
terizations, and infections (e.g., tuberculosis compromise by increasing abdominal tone
and syphilis). and decreasing respiratory muscle tone and
lung volume. Sedatives can also decrease
Clinical findings and evaluation venous return and should be used cautiously.
1. History and physical examination: The 6. Bone marrow aspiration under local
history is nonspecific, with a typically short anesthesia: If the bone marrow is involved,
period of progressively worsening this test will allow the diagnosis to be made
symptoms. Patients with ALL may have a without a biopsy of the mass itself.
short history of fever, bone pain, bruising, 7. Examination of pleural fluid/pericardial
petechiae, or other signs of marrow fluid/ascitic fluid: In addition to relieving
dysfunction. Presenting symptoms and distress, removal of fluid and subsequent
signs include cough and dyspnea cytological examination may allow a
(commonest), closely followed by dysphagia, definitive diagnosis to be made, especially in
orthopnea, hoarseness, chest pain, facial the case of NHL.
edema, wheezing, pleural effusion, 8. Echocardiography: This is necessary to
pericardial effusion, features of carbon look for pericardial effusion and cardiac
dioxide retention like anxiety, confusion, tamponade and to evaluate myocardial
lethargy, headache, distorted vision, altered function.
mental status, and syncope, and occasionally 9. Pulmonary function tests: The patient’s
other symptoms like conjunctival suffusion peak expiratory flow rate and the shape of
and cyanosis. The respiratory symptoms are the flow volume loop can reliably predict the
often much worse in a lying position and the patient’s ability to tolerate various diagnostic
patient will often assume a position of being and therapeutic procedures.
upright for comfort. Engorgement of 10. Measurement of β‐HCG (β‐subunit of
collateral veins may be apparent. human chorionic gonadotropin) and AFP (α‐
2. Laboratory assessment: Screening labs fetoprotein). Elevations in these markers are
including a CBC, chemistries, LDH, and diagnostic of a malignant germ cell tumor.
uric acid should be done for diagnostic clues
as well as determination of general well‐ Management
being of the patient. 1. Children should be under close observa­
3. Chest radiography including a lateral tion in the intensive care unit with comfort­
view: This usually shows mediastinal able positioning with elevation of the head,
Oncologic Emergencies 175

continuous cardiovascular and respiratory diagnosis. Radiation may not provide any
monitoring, and pulse oximetry. quicker response than steroids or other
2. Extreme care to ensure oxygenation, air­ chemotherapy and may require moving the
way patency with emergency staff readily patient outside the critical care setting and
available is critical. Management of pain, possibly positioning or sedating the patient.
anxiety, and stress with procedures should All risks and benefits need to be carefully
consider the possibility of an emergent considered before entertaining this
decline in status. Anesthesiology and criti­ intervention. Tracheal swelling and further
cal care staff should monitor the child. airway compromise can occur as a result of
Avoid the use of medications that may radiation.
cause relaxation of the intercostal muscles 5. Patients that have symptomatic venous
and decrease intravascular resistance. thrombosis as the etiology for SVCS should
Diagnostic procedures that cause pain or be treated with unfractionated heparin,
require special positioning or sedation may 75 U/kg as a loading dose followed by 18 U/
not be possible. kg/h in children or 28 U/kg/h in infants to a
3. Empiric therapy for suspected malig­ goal unfractionated anti‐Xa level of 0.3–
nancy may need to be initiated due to the 0.7 U/ml. Low‐molecular‐weight heparin is
life‐threatening situation. Steroids, cyclo­ a suitable therapeutic alternative at a dose of
phosphamide, vincristine, or anthracyclines 1 mg/kg every 12 hours with a goal
have been given in this situation to children fractionated anti‐Xa level of 0.5–1.0 U/ml.
with suspected leukemia or lymphoma.
Prednisone is commonly used, as most of Spinal cord compression
these children are ultimately diagnosed Acute spinal cord (or cauda equina)
with lymphoma or leukemia, especially in compression occurs in <5% of children with
those with typical laboratory abnormalities cancer. Prolonged cord compression leads
or clinical findings (i.e., organomegaly, gen­ to irreversible neurological injury with
eralized adenopathy, evidence of tumor paralysis, sensory loss, and loss of bowel and
lysis syndrome [TLS], elevated WBC [white bladder control. Once a neurological deficit
blood cell] counts). Intravenous methyl­ occurs, it often progresses to paraplegia
prednisolone is started at a dose of 50 mg/ within days or even hours. The most
m2/day divided twice daily (BID). Hydration frequent cause of cord compression is
should be given and the child monitored for external compression caused by extension
TLS, which may be exacerbated by the ini­ of a paravertebral tumor through an
tiation of therapy (discussed later). If the intervertebral foramen into the epidural
patient does not respond to steroids within space, most typically a sarcoma. The tumor
24–48 hours and the patient is less stable, compresses the vertebral venous plexus
then additional chemotherapeutic agents leading to cord edema, venous hemorrhage,
should be started, although other diagnos­ myelopathy, and ischemia. The most
tic conditions must be considered. common tumors leading to spinal cord
4. Children with impending or actual air­ compression include neuroblastoma, Ewing
way obstruction may be candidates for sarcoma, germ cell tumors, NHL, Hodgkin
emergent radiation therapy, given in 1–2 Gy lymphoma, as well as drop metastases from
fractions for 1–4 days. A small area of the central nervous system tumors. Tumor
tumor should be shielded to prevent masses cause compression of the epidural or
radiation‐induced changes if a biopsy still subarachnoid space, and metastases cause
needs to be performed to establish the compression typically within the cord
176 Chapter 14

parenchyma. Intraspinous chloromas in Dexamethasone 0.1–0.2 mg/kg (loading


patients with acute myelogenous leukemia dose) should be given IV even before imag­
(AML) are a less common cause. Spinal cord ing is performed, and an MRI should be
astrocytomas and ependymomas can also obtained immediately. Dosing should con­
cause cord compression, more commonly tinue at 0.1 mg/kg every 6–8 hours. Patients
from an intramedullary location. with suspicious findings but without spe­
cific neurologic deficits can be started on a
Clinical findings and evaluation lower dose of oral dexamethasone (0.025–
1. Localized back pain or radicular pain 0.05 mg/kg every 6 hours) with MRI per­
extending down the leg occurs in up to 80% formed within 24 hours.
of children with cord compression. Any 2. If imaging reveals a tumor with spinal
child with known cancer and back pain cord compression, this must be promptly
should be presumed to have spinal cord relieved. Surgical decompression with a
compression and an evaluation is warranted laminectomy quickly relieves pressure and
emergently. The duration of symptoms is allows tumor to be removed for diagnosis. It
variable but generally short; one series should be performed if the diagnosis is
reported a range of 5 days to 4 weeks. unknown. If the diagnosis is known and the
Characteristically, straight leg raising, neck tumor is radiosensitive, emergent radiation
flexion, or the Valsalva maneuver aggravates therapy should be promptly initiated.
the pain. Pain is almost always the first Chemotherapy is an alternative for
presenting symptom, with weakness and chemotherapy‐responsive tumors such as
bowel or bladder dysfunction occurring neuroblastoma, Ewing sarcoma, Hodgkin
later. Sensory loss may occur but is often lymphoma, and NHL; the onset of action in
difficult to identify, especially in young these tumors is similar to radiation therapy.
children. Any objective neurological deficit The prognosis for recovery of function is
requires further immediate evaluation. directly related to the degree of disability at
2. Plain radiographs are generally not diagnosis. This is associated with the
helpful in identifying abnormalities in duration of symptoms and the length of
children, as cord compression typically time required to make the diagnosis.
occurs via extension of tumor through the Patients who are ambulatory at diagnosis
intervertebral foramina without bony typically remain ambulatory; approximately
erosion or injury. MRI is the best study for 50% of children who are nonambulatory at
demonstrating intraspinous extension. While diagnosis regain the ability to ambulate.
CT is less sensitive in identifying extension
of disease into the spinal canal, it will Increased intracranial pressure
effectively identify paraspinous disease. and brain herniation
3. Examination of the cerebrospinal fluid Brain tumors are the most common solid
(CSF) is rarely helpful, and patients may expe­ tumor in childhood, and the majority of
rience rapid neurological deterioration fol­ patients present with signs and symptoms of
lowing a lumbar puncture (spinal coning). increased intracranial pressure (ICP).
Fortunately, few of these patients progress to
Management actual brain herniation. Most pediatric
1. Patients with rapidly progressing spinal brain tumors are infratentorial and cause
cord dysfunction require immediate inter­ increased ICP by obstructing the third or
vention following a thorough history and fourth ventricle with resultant obstructive
meticulous neurologic examination. hydrocephalus. They can also cause
Oncologic Emergencies 177

increased ICP simply by mass effect. This Management


most commonly occurs with astrocytomas, If increased ICP or impending brain hernia­
as well as with primitive neuroectodermal tion is suspected, appropriate management
tumors including medulloblastoma. must be initiated prior to more definitive
The presentation can vary significantly testing to hopefully avoid death or perma­
depending on patient age. Common nent neurological injury. The following
symptoms in infants include vomiting, steps should be initiated:
lethargy, personality changes, loss of 1. Fluid intake should be limited to no
developmental milestones, and increased more than 75% maintenance. Hypertonic
head circumference. Older children saline and mannitol are often utilized.
commonly complain of headache. This is 2. A loading dose of dexamethasone
frequently intermittent at first, and then 0.1 mg/kg should be given IV followed by
becomes more frequent and severe. It 0.05 mg/kg every 6 hours.
characteristically occurs in the morning, 3. An emergent CT scan of the head should
and is often but not always accompanied by be performed. A noncontrast scan will
vomiting without diarrhea. Other signs and effectively identify bleeding, most tumors,
symptoms include diplopia, ataxia, cerebral edema, and hydrocephalus.
hemiparesis, speech disturbance, neck 4. If the diagnosis of increased ICP is
stiffness, dizziness, lethargy, and coma. confirmed, the patient should be admitted
Some tumors may also cause focal neuro­ to the ICU. Neurosurgical consultation
logical changes. Cerebellar astrocytomas should be immediately obtained if a tumor
may lead to ipsilateral weakness, hypotonia, is identified or the patient has hydrocephalus.
and ataxia. Herniation of a cerebellar tonsil 5. Acetazolamide 5 mg/kg/dose every 6
often causes head tilt and neck stiffness. hours can be given to decrease CSF
Tumors near the third ventricle (craniophar­ production.
yngiomas, germ cell tumors, optic gliomas, 6. Intubation and hyperventilation to
and hypothalamic and pituitary tumors) decrease the pCO2 to 20–25 mmHg will
may cause visual loss, increased ICP, and decrease cerebral perfusion. Care must be
hydrocephalus. A pineal tumor may lead to taken not to cause cerebral ischemia.
Parinaud’s syndrome (impairment of upward 7. Surgical options (external ventricular
gaze, convergence nystagmus, and altera­ drainage, third ventriculostomy) should be
tions in pupillary response). considered.
8. Prophylaxis with antiseizure medication
Clinical findings and evaluation should also be considered.
1. Assessment of vital signs is critical. 9. Intracranial pressure monitoring may be
Increased ICP can result in Cushing’s triad: beneficial.
bradycardia, hypertension, and apnea. 10. An MRI should be obtained as soon as
2. A careful physical examination looking possible to look for or confirm the presence
for signs of increased ICP or impending her­ of a tumor.
niation must be done quickly. In addition to
the findings mentioned in the preceding text, Massive hepatomegaly
impending brain herniation may cause Massive hepatomegaly is the most emergent
changes in respiratory pattern, pupil size and complication of stage 4S neuroblastoma in
reactivity, extraocular movements, spontane­ infants, especially those under 4 weeks of
ous motor function, and responsiveness to age. While 4S neuroblastoma often regresses
verbal and physical stimuli. spontaneously, when it occurs during early
178 Chapter 14

infancy the resulting hepatomegaly can leukemia), as well as during therapy with
cause death due to respiratory insufficiency chemotherapy and/or radiation. Patients are
or hepatic failure. Patients with hepato­ immune suppressed and at risk for infection
blastoma and transient myeloproliferative with bacteria, viruses, fungi, and protozoa.
disorder may also present with massive Fever in a patient with neutropenia is a
hepatomegaly, while those with Wilms medical emergency and standards of
tumor may present with a massive abdomi­ practice should be in place at institutions
nal tumor with similar clinical presentation. caring for these patients to immediately
assess and provide treatment for these
Clinical findings and evaluation patients. Details of this management are
1. The patient should be examined for evi­ provided in Chapter 27.
dence of gastrointestinal dysfunction, res­
piratory compromise (respiratory rate >60/ Neutropenic colitis
min or oxygen requirement), poor venous Patients experiencing severe neutropenia,
return (leg edema), renal dysfunction (poor particularly those in treatment for leukemia
urine output and azotemia), hypertensive or stem cell transplant, are at particular risk
urgency or emergency, or disseminated to develop colitis. Acute inflammation of the
intravascular coagulation (DIC). cecum in particular is termed typhlitis,
2. Baseline laboratory and imaging studies though neutropenic colitis is generally the
should be performed urgently to help more accepted term, since colitis can occur
determine underlying etiology to allow for beyond the cecum. Chemotherapy is usually
prompt initiation of appropriate therapy. the initial inciter of mucosal injury,
predisposing the patient to inflammation
Treatment and infection. Patients presenting with signs
1. If the patient is stable and does not and symptoms of an acute abdomen with
exhibit any of the findings mentioned in the sudden onset of severe pain (often localized
earlier text, careful observation and sup­ to the right lower quadrant) suggestive of an
portive care should be provided. Positive obstruction (functional or physical) should
pressure can be helpful given the low lung be assumed to have neutropenic colitis until
volumes secondary to the large abdominal proven otherwise. This condition is the
tumor. Coagulation studies may be altered if result of mucosal invasion by bacteria or
there is significant liver involvement. fungi and can quickly progress to full
2. If the patient is unstable, treatment thickness mucosal involvement, perforation,
with chemotherapy or radiation therapy peritonitis, and septic shock. Pathogens
should be considered dependent on the most commonly implicated are Pseudomonas
underlying diagnosis. Definitive surgical spp., Escherichia coli, nonspecific Gram‐
resection for massive Wilms tumor may negative bacteria, Staphylococcus aureus, α‐
also be required. hemolytic streptococcus, Clostridium spp.,
Aspergillus spp., and Candida spp. Acute
appendicitis must also be a consideration.
Emergencies caused by
inflammation/infection Clinical Findings and Evaluation
1. Careful history of recent chemotherapy
Fever and neutropenia (or stem cell transplant) and presentation
Neutropenia is commonly seen in the newly with right lower quadrant pain, with
diagnosed cancer patient (primarily acute physical exam findings revealing absent
Oncologic Emergencies 179

bowel sounds, bowel distention (may lead to assessment of urine output to guide
abdominal distention), tenderness of hydration. Bolus hydration is often
palpation, or a palpable mass, are highly necessary on admission with continuation
suspicious of an acute functional bowel of one to two times maintenance intravenous
obstruction and neutropenic colitis. Serial hydration. Vasopressors may be needed if
examinations are warranted. hypotension is present. Sepsis protocols
2. Imaging studies may prove diagnostic should be followed for any patient presenting
and include: plain radiograph of the with clinical evidence of sepsis.
abdomen which may reveal pneumatosis 3. Surgical consultation is considered
intestinalis, free air in the peritoneum or standard of care upon admission, though
bowel wall thickening; abdominal surgical intervention is rarely required with
ultrasound may show thickening of the current supportive care measures. The
cecum or affected large bowel (most patient should be monitored for a clinical
common initial imaging assessment); CT change (i.e., drop in blood pressure,
scan is the definitive imaging technique and worsening pain, evidence on imaging of free
should be done in all patients with the air in the peritoneum) suggesting rupture of
suspected diagnosis. It may demonstrate the mucosa or suspicion for necrotic bowel,
diffuse thickening of the colonic wall and both indications for acute surgical
suspicion of necrotic bowel. Mortality has intervention. Surgery consists of resection
decreased significantly in the past decade of infarcted bowel and diversion via
due to early recognition and intervention colostomy.
with bowel rest and antimicrobials.
However, patients may progress to bowel
necrosis, perforation, and sepsis in a short Emergencies caused by
period of time and require close monitoring abnormalities of blood
and quick intervention. and blood vessels
3. Laboratory assessment should include
blood cultures (from all central catheter Hyperleukocytosis
lumens), CBC, and complete metabolic Hyperleukocytosis is defined as a peripheral
panel. Patients should be tested for WBC count >100 × 109/l. It is most
Clostridium difficile toxin. commonly seen at presentation or relapse of
AML, ALL, or chronic myelogenous
Treatment leukemia (CML). Using this definition, the
1. Supportive medical management is the incidence of hyperleukocytosis at
mainstay of treatment and includes: nothing presentation is 9–13% of children with ALL,
by mouth (NPO) status, a nasogastric tube 5–22% with AML, and almost all children in
(NG) in place to suction (if tolerated); chronic phase of CML. However, clinically
initiation of broad‐spectrum antibiotics to significant hyperleukocytosis occurs when
ensure coverage for anaerobic and Gram‐ the WBC counts are >200 × 109/l in patients
negative organisms; intravenous fluids with AML or >300 × 109/l in patients with
(IVF) and electrolyte replacement. ALL or CML. The most common
Transfusion support may be indicated as complication of hyperleukocytosis in AML
often patients present with severe and CML is stroke, whereas in ALL it is TLS.
pancytopenia. Hyperleukocytosis is more common in
2. The patient should be placed on infantile ALL and AML, T‐cell ALL, and in
cardiovascular monitoring with meticulous any phase of CML.
180 Chapter 14

Hyperleukocytosis leads to occlusion of (inclusive of phosphorus), and uric acid, to


small veins in the brain, lung, and other follow the rate of rise of the WBC or
organs through the formation of WBC response to therapy, as well as monitor for
aggregates (white thrombi). The rheology of evidence of TLS and renal dysfunction (see
blasts (large, non‐deformable cells, the section “Tumor Lysis Syndrome”).
adherence to each other and the vascular 4. Assess risk for hemorrhage and DIC.
endothelium, elevated leukocrit) leads to Patients with AML (especially acute
sludging and resultant tissue ischemia. A promyelocytic leukemia, APL) and those
vicious cycle of worsening leukostasis, with ALL and WBC >400 × 109/l are at
thrombosis, and secondary hemorrhage particular risk for coagulopathy and
may ensue. As myeloblasts and monoblasts intracranial hemorrhage and therefore
are less deformable than lymphoblasts or coagulation studies, platelets and fibrinogen
granulocytes, leukostasis is far more likely to should be monitored closely and replenished
occur in AML than in ALL or CML, whereas as necessary.
patients with ALL are more likely to develop
metabolic derangements from TLS. Other Treatment
factors that can increase the risk of 1. Therapeutic intervention should be
leukostasis include dehydration and an immediate.
elevated hemoglobin level. 2. If the child is being transported, the fol­
Poor perfusion and anaerobic lowing simple measures are to be initiated
metabolism in the microcirculation lead to prior to transport. The mainstay of therapy is
lactic acidosis. Thrombi develop in the aggressive hydration, typically two to three
circulation leading to vascular damage and times maintenance IVF. Alkalinization of
parenchymal ischemia, and resultant edema IVF promotes excretion of uric acid by con­
and hemorrhage in the small vessels of the verting it to a more soluble urate salt. A fluid
lungs and central nervous system initially such as D5W 1/4NS with 40 mEq/l NaHCO3
(and then widespread in larger vasculature). is quite suitable. Allopurinol or urate oxidase
Clinical evidence of small vessel occlusion is (rasburicase) should also be started to pre­
associated with early morbidity and vent or decrease the severity of hyperurice­
mortality. mia. If urate oxidase is given, alkalinization
is not required (or recommended), as the
Clinical findings and evaluation uric acid level falls precipitously and uric
1. Signs and symptoms of leukemia are acid solubility is no longer an issue.
usually present (pallor, fatigue, fever, Alkalinization should also not be given with
bleeding, bone pain, adenopathy, underlying hyperphosphatemia due to
organomegaly, anemia, thrombocytopenia). increased propensity for calcium phosphate
2. Signs and symptoms of leukostasis in the precipitation. This topic is discussed more
lungs or brain include tachypnea, hypoxia, fully in the section on TLS.
acidosis, dyspnea, cyanosis, blurred vision, 3. Therapy for the underlying malignancy
papilledema, stupor/confusion, coma, or should begin as soon as possible and is the
ataxia. Oliguria, anuria, and priapism may most important intervention for
also occur in this setting. Intracranial hyperleukocytosis. Prompt initiation of
hemorrhage may also occur. therapy can mitigate the potential risks of
3. The laboratory assessment should hyperleukocytosis. Diagnosis can be made
include frequent monitoring (i.e., every using peripheral blood flow cytometry given
4–12 hours) of CBC, renal function panel the high peripheral WBC count.
Oncologic Emergencies 181

4. In situations where prompt initiation of 2. Several authors of textbooks on pediatric


chemotherapy cannot be initiated or in the oncology recommend that children with
patient with rapidly rising WBC count or profound anemia (hemoglobin <5 g/dl) that
clinical deterioration (pulmonary or CNS has developed over an extended period of
leukostasis), leukapheresis can be time be transfused more slowly (1 vs. 3 ml/
considered, though impact on mortality is kg over 4 hours) to avoid causing congestive
controversial. Leukapheresis should not be heart failure. This concern is based on an
initiated if it will delay start of chemotherapy. extrapolation of the Starling law of cardiac
5. Emergency cranial radiation therapy has physiology, but there are few data supporting
not been demonstrated to improve outcome this concern. Two published studies as well
and is not recommended. as experience in our own institution suggest
6. Platelet transfusions can be given safely, that this practice is unnecessary, especially if
as platelets have not been shown to increase these children are being cared for in the ICU
viscosity or worsen leukostasis and may where more careful monitoring is being
decrease risk of hemorrhage. Platelets performed. If there are signs or symptoms of
should be given if the child is bleeding or the congestive heart failure or cardiomegaly on
platelet count is <50 × 109/l. Red cell chest X‐ray, furosemide can mitigate
transfusions should be given with extreme circulatory overload. More aggressive
caution due to the potential to increase total transfusion can avoid potentially exposing
blood viscosity and worsen leukostasis. The patients to additional donor units and
main indications are cardiovascular prevent delay in diagnostic procedures.
compromise caused by anemia as well as 3. All blood products given to known or
threshold requirements for anesthesia. The potential oncology patients should be
hemoglobin level should be kept <10 g/dl. leukoreduced to decrease the risk of CMV
transmission and irradiated to prevent
Anemia transfusion‐associated graft‐versus‐host
Anemia is a common finding in children disease (TA‐GVHD).
presenting with malignancy. Up to 80% of 4. Erythropoietin is not used in children at
children presenting with ALL will be anemic this time to increase the hemoglobin and
at diagnosis. This can result from bleeding, decrease the need for transfusion due to
inflammation, or marrow infiltration by data from adult studies that suggest adult
tumor. It is rarely an emergency; children patients receiving erythropoietin have
can tolerate a hemoglobin level as low as increased risk of thrombosis and subsequent
2–3 g/dl if it develops slowly from decreased mortality.
production. Conversely, levels as low as 5 g/
dl can be life‐threatening if they occur as a
result of sudden hemorrhage. Metabolic emergencies

Treatment Tumor lysis syndrome


1. Anemia in this situation is treated with TLS is a pattern of specific metabolic
packed red blood cell (PRBC) transfusions. abnormalities that occurs as a result of
As a general rule, 10 ml/kg of PRBCs will extremely rapid cellular turnover. It is
increase the hemoglobin level by 2–3 g/dl. In almost always encountered in the context of
practice, children with acute anemia due to malignant tumors that either have a rapid
blood loss may need 15–20 ml/kg to reach rate of apoptosis or after initiation of therapy
this level. in bulky tumors that are exquisitely sensitive
182 Chapter 14

to the therapy being provided. The rapid doubling time (38–116 hours in Burkitt
lysis of tumor cells in either situation causes lymphoma), poor urine output (often
large amounts of potassium, phosphate, and secondary to tumor infiltration in the
nucleic acids to be released into the kidneys), and elevated uric acid. TLS has
circulation, resulting in hyperuricemia, also been rarely reported in children with
hyperphosphatemia (often associated with hepatoblastoma and advanced stage
hypocalcemia), and hyperkalemia. Cell lysis neuroblastoma.
and release of intracellular contents often
exceed the excretory capacity of the kidneys Clinical findings and evaluation
further compromising renal function. 1. Evaluate for signs and symptoms of met­
Tumor infiltration of the kidney and abolic abnormalities, specifically hyper­
decreased intravascular volume can further kalemia and hyperphosphatemia. These
impair renal function and accelerate the include lethargy, nausea, vomiting, hypo­
development of TLS. When uric acid tension, muscle spasm, and cardiac
concentration in the renal tubule exceeds its dysrhythmia.
solubility coefficient, urate crystals form in 2. Perform a metabolic panel in all children
the tubule causing obstruction. Lactic with suspected malignancy to assess for
acidosis, secondary to poor tissue TLS. Frequent reassessments should be
oxygenation in patients with high WBC performed early in therapy as well,
counts, may contribute to uric acid particularly in patients with high tumor
deposition. In addition, phosphates are burdens and those expected to respond
released when tumor cells lyse. When the rapidly to treatment. The timing of serial
calcium–phosphorus product exceeds 60, assessments will depend on the underlying
precipitation occurs in the microvasculature, disease, evidence of preexisting renal
particularly in an alkaline environment, dysfunction, timing of initiation of therapy,
further obstructing the renal tubules and and the risk of TLS. A uric acid level of
resulting in hypocalcemia. Lymphoblasts ≥8 mg/dl, potassium of ≥6 mEq/dl, or
are particularly rich in phosphate, phosphate of ≥2.1 mmol/l is of concern, as
containing up to four times the amount well as a creatinine that is above the normal
found in normal lymphocytes. Potassium is range for age (or known for the patient).
also released from tumor cells, is secondarily 3. Carefully monitor vital signs, intake and
elevated with poor renal function, and can output for evidence of renal insufficiency,
lead to fatal arrhythmia. The complete and possible symptoms of hypocalcemia
syndrome includes all these components, (anorexia, vomiting, cramps, spasms, tetany,
with progressively worsening renal failure. and seizures). A renal ultrasound may be
Insufficiently treated TLS can lead to cardiac indicated in suspected renal dysfunction.
arrhythmia, renal failure, seizure, coma,
DIC, or death. Prevention and intervention
In children, TLS develops most 1. Aggressive hydration should be provided
frequently in association with malignancies at a rate of two to three times (up to four
presenting with a high tumor burden or times) maintenance to promote excretion of
rapid rate of malignant cell proliferation uric acid and phosphorus. Diuretics
such as Burkitt lymphoma and other NHL (furosemide, mannitol) may also be needed
as well as pre‐B‐cell and T‐cell ALL. These to promote good urine output (>3 ml/kg/h).
malignancies typically present with a large Alkalinization with D5W 1/4NS + 40 mEq/l
tumor burden (i.e., elevated LDH), short NaHCO3 can be utilized to aid with uric acid
Oncologic Emergencies 183

excretion, but will worsen calcium over 30 minutes (though even lower doses of
phosphate precipitation so should be not 0.03–0.05 mg/kg may be sufficient in
ordered in the patient with consideration for vial size/usage). This dose
hyperphosphatemia. The bicarbonate can be repeated daily for up to 5 days;
should be titrated to keep the urine pH however, a single dose will often keep the
between 7 and 7.5, which will optimize the uric acid level low for 48 hours or more, and
excretion of uric acid without leading to may be sufficient to prevent clinical TLS for
clinically significant metabolic alkalosis. the entire course. Patients treated with
2. An agent to prevent or reverse rasburicase do not require alkalinization,
hyperuricemia should also be initiated which is an advantage when managing other
immediately. Allopurinol (xanthine analog electrolyte abnormalities. It can potentiate
that blocks the conversion of xanthine and hemolysis in patients with G6PD deficiency
hypoxanthine to uric acid by competitively and has caused methemoglobinemia in
inhibiting xanthine oxidase) is the most susceptible individuals. Routine screening
commonly used agent for this purpose, and for G6PD deficiency is not recommended
it can be given at presentation and as needed before use, but families should be asked
during the early phase of therapy for the about this possibility. It also has rarely
prevention or treatment of TLS. Allopurinol caused anaphylaxis.
does not eliminate existing uric acid and 3. Potassium should not be added to IV
therefore may not be the optimal choice in fluids until it is clear that the potassium is
the patient presenting with a markedly stable and not increasing due to TLS or renal
elevated uric acid and high tumor burden. insufficiency.
The urine is typically alkalinized when 4. The patient’s intake and output must be
allopurinol is used, as it often does not cause closely monitored to ensure it remains
a significant decrease in the uric acid level balanced, taking into account insensible
until the rate of tumor lysis slows. It is given losses.
at a dose of 300 mg/m2/day divided every 8 5. Careful metabolic monitoring should be
hours (800 mg/day maximum). Another performed with assessment of renal function
agent, rasburicase (urate oxidase), is a (blood urea nitrogen (BUN), creatinine),
recombinant enzyme that acts as a catalyst electrolytes, calcium, phosphorus, and uric
in the degradation of uric acid into allantoin, acid every 4–12 hours depending on rapidity
a highly soluble product compared with uric of change, degree of TLS or renal
acid. It works quickly (within 30 minutes of dysfunction, and timing of chemotherapy.
IV infusion) and keeps the uric acid level 6. Hyperkalemia (<6 mEq/l and asympto­
low for 12–24 hours or longer, depending on matic) can typically be treated with hydration
the underlying rate of cell turnover. and diuretics as described in the earlier text.
Rasburicase has been studied in children Sodium polystyrene sulfonate (kayexalate) at
with cancer who have hyperuricemia as a a dose of 1 g/kg every 6 hours with sorbitol
result of tumor lysis and has been found to 50–150 ml will remove 1 mEq of potassium
be safe and effective. Rasburicase is the per liter per gram of resin over 24 hours.
preferred agent in the setting of Patients with potassium ≥6 mEq/l ± symp­
hyperuricemia and any degree of renal toms should have cardiac monitoring and
insufficiency, concern for rapidly increasing receive directed therapy with rapid infusions
tumor burden, or massive lysis. Rasburicase of insulin (0.1 U/kg/h) and glucose (dextrose
is given at a dose of 0.15–0.2 mg/kg/day in 0.5 g/kg/h) with close glucose monitoring.
50 ml preservative‐free normal saline IV The electrocardiogram (ECG) may show
184 Chapter 14

widened QRS complexes and peaked T waves and alveolar rhabdomyosarcoma, but has
in hyperkalemia. Symptomatic hyperkalemia also been reported in children diagnosed
requires immediate therapy with calcium with rhabdoid tumor, hepatoblastoma,
gluconate, albuterol, and glucose and insulin, Hodgkin lymphoma, NHL, AML, brain
along with kayexalate to remove excess potas­ tumors, and neuroblastoma. The cause of
sium from the body. In emergent situations, MAH can be increased bone resorption
50% dextrose can be given at a dose of 1 ml/ from skeletal metastases or production of
kg through a central line. Patients who calcium‐mobilizing substances such as
develop life‐threatening arrhythmias should parathyroid hormone (PTH)‐related pep­
receive calcium gluconate (100–200 mg/kg) tide or osteoclast‐activating factor by the
or calcium chloride (10 mg/kg) slow IV infu­ tumor. MAH in turn adversely affects the
sion. Calcium infusions cannot be given in ability of the kidney to concentrate the
the same line as NaHCO3. NaHCO3 stabilizes urine, leading to dehydration, decreased
cardiac cell membranes and is used to reverse glomerular filtration rate (GFR), a further
acidosis (1–2 mEq/kg IV). These interven­ reduction in calcium excretion, and wors­
tions should be done in the critical care set­ ening hypercalcemia.
ting. Dialysis should be considered in patients
in which these emergent interventions are Evaluation
necessary, especially in those patients at high 1. Symptoms of mild hypercalcemia (12–
risk for continuation or worsening of TLS. 15 mg/dl) include generalized weakness,
7. If hyperphosphatemia is present, treat­ poor appetite, nausea, vomiting,
ment is forced saline diuresis with furosem­ constipation, abdominal or back pain,
ide and dietary phosphate restriction. Oral polyuria, and drowsiness. More severe
phosphate binders are typically not effective, hypercalcemia (>15 mg/dl) results in
as they do nothing to remove phosphate from profound muscle weakness, severe nausea
the circulation. Persistent hyperphos­ and vomiting, coma, and bradydysrhythmias
phatemia may require dialysis. with broad T waves and prolonged PR
8. Hypocalcemia may develop in the face of interval on the ECG.
hyperphosphatemia. As the calcium‐ 2. Patients exhibiting any of these
phosphate product increases, compensatory symptoms should have a serum Ca2+ level
hypocalcemia may develop with resultant measured immediately. Measurement of an
clinical symptoms (e.g., tetany). Treatment ionized Ca2+ should be done if there is any
of symptomatic hypocalcemia is with question about the validity of the total
administration of elemental calcium, such ­calcium measurement or to rule out primary
as 10% calcium gluconate 0.5–1.0 ml/kg (10 hyperparathyroidism or pseudohyper­
mg/kg) until symptoms abate. Dosing calcemia due to increased plasma protein
should be judicious to treat symptoms but binding capacity.
not exacerbate calcium phosphate 3. Measurement of the intact PTH level
precipitation. Calcium should not be given should be performed to exclude concomitant
in the same line as NaHCO3. hyperparathyroidism. PTH levels are usually
low, normal, or suppressed in MAH. The
Hypercalcemia serum concentration of calcitriol should be
Malignancy‐associated hypercalcemia (MAH) measured when the hypercalcemia is
is a rare complication of childhood cancer, suspected to be due to Hodgkin lymphoma
occurring in 0.4–1.3% of newly diagnosed or NHL, as it has been implicated as a key
cases. It is seen most frequently in ALL mediator of hypercalcemia in almost all
Oncologic Emergencies 185

cases of Hodgkin lymphoma and 30–40% of breathing pattern. The X‐ray shows a large
cases of NHL. anterior mediastinal mass occupying over
two‐thirds of the chest volume and severely
Treatment compressing the trachea, which appears
1. The goals of treatment are to increase flattened on the lateral view. The heart size
renal clearance of calcium and decrease appears large as well. After reviewing this
bone resorption. film and recognizing the threat for
2. Aggressive hydration will help increase immediate airway compromise, you locate
renal excretion of calcium. Forced diuresis the patient in the waiting area with his
with normal saline at two to three times mother. He is quiet and appears to be
maintenance and furosemide 2–3 mg/kg making significant efforts to breath and is
every 2 hours was previously recommended. cautious in his movements. He is
Furosemide blocks calcium reabsorption by immediately taken into the CT scanner
the kidney, and can decrease the calcium (performed with the patient in a semi‐
level in 24–48 hours. However, a recent upright position due to discomfort and
meta‐analysis suggests the data to support desaturations with lying down). The CT
this approach are limited, and it can also confirms pressure on the entire trachea by
cause severe loss of sodium, potassium, the mass, as well as the lower airways,
and magnesium. At present, the use of furo­ confirming the suspicion that an
semide is recommended only for patients endotracheal (ET) tube would not secure
experiencing fluid overload following the airway. Additionally, a large pleural
aggressive saline diuresis. effusion is present.
3. If the patient is hypophosphatemic, a. What are the immediate next steps?
phosphate replacement at a dose of 10 mg/ The patient is in imminent danger of los­
kg/dose 2–3 times per day may inhibit ing his airway due to compression along
osteoclastic activity and promote calcium the entire airway including below the
deposition into bone. bifurcation. Potential emergencies the
4. Bisphosphonates have become the patient is at risk for include: airway com­
treatment of choice for adults with MAH, pression/obstruction, cardiac compro­
but these have not been widely studied in mise, TLS, and SVC syndrome. The patient
children. The most published experience is should be positioned in a comfortable
with pamidronate at a dose of 0.5–2 mg/kg position, likely upright and often bending
given IV over 2–24 hours. This has been forward. Additional immediate steps
highly successful in correcting MAH within should include: initiation of oxygen by
1–4 days of treatment. When MAH is nasal cannula, cardiovascular and pulse
associated with ALL, rapid initiation of oximetry moni­toring, IV access, alerting
induction therapy has been shown to be anesthesia of the patient’s critical airway,
very important in reversing the and moving the patient to the critical care
hypercalcemia. unit. It is important to keep the patient
calm and comfortable and avoid sedation,
which would lead to decreased vascular
Case study for review tone, decreased cardiac return, and poten­
tial cardiovascular decompensation. Pain
You are called by the radiologist to review a medications should be given with caution
chest X‐ray obtained on a 4‐year‐old boy given the potential for smooth muscle
referred for evaluation of an unusual relaxation.
186 Chapter 14

Upon further questioning, the patient has is too tenuous to risk anesthesia. Diagnosis
a history of 1 month of intermittent fevers of by flow cytometry from peripheral blood
unknown etiology, increasing fatigue, and a can be done if sufficient blasts are present.
1‐lb weight loss. Over the past 1–2 weeks, he Depending on the lab findings, other
has complained of headaches and has an odd interventions may include transfusion sup­
cough or grunting sound. He was thought to port and correction of metabolic
have croup or a viral upper respiratory infec­ derangements.
tion (URI) and received oral antibiotics and The patient’s lab findings are as follows:
a single dose of prednisone with an albuterol WBC 25.1 × 109/l, hgb 14.0 g/dl, platelets
aerosol treatment, without improvement. 480 × 109/l
The parent denies bone pain, night sweats, Na+ 149, K+ 5.2, Cl− 108, CO2 22, Ca++
bruising, or overt bleeding. He has not been 11.2, Phos 5.6, Cr 0.4, LDH 4998 (nl 425–
eating much, but is drinking well and urinat­ 975), uric acid 8.7 (nl 2.3–6.1)
ing normally. Review of the peripheral blood smear shows
On exam, his temp is 37.4 °C, HR 142, a large population of abnormal WBCs with
RR 30, BP 123/30, SaO2 97% on 10 l/min clumped chromatin, high nuclear:cytoplasmic
non‐rebreather (NRB) facemask. Prior to ratio, and an increased number of eosinophils
the oxygen and pulse oximetry, he was noted and basophils.
to have perioral cyanosis with movement as Germ cell tumor markers: βHCG <2
well as with placement of the peripheral mIU/ml (nl); AFP 1.0 ng/ml (nl).
intravenous catheter (PIV). His skin is clear, c. What intervention(s) would you
with mild pallor and no petechiae or ecchy­ start? What are the potential
moses. He has no adenopathy, lungs are consequences?
clear with diminished breath sounds bilater­ IV hydration should be initiated, as the
ally, noted systolic murmur, and no gallop or patient has evidence of rapid cell turnover
friction rub. The patient has no hepatosple­ with an elevated uric acid and LDH as
nomegaly or testicular abnormality. well as elevated Na+ and K+. Ideally start at
b. What is the most likely diagnosis and 2× maintenance IVF, though patients may
what studies would you obtain? require bolus fluids if dehydrated or renal
Most likely the patient has acute lympho­ function is compromised. Avoid K+ or phos
blastic leukemia or lymphoma. T‐cell dis­ in the IVF. This patient must be monitored
ease is more likely to present in this manner. for signs of fluid overload given the pleural
Another consideration for a large mediasti­ effusions and potential vascular congestion
nal mass is a germ cell tumor. The patient from tumor compression.
should have a CBC, comprehensive meta­ Rasburicase is indicated given the bulky
bolic panel (CMP), and labs to evaluate for disease, elevated uric acid, and likely contin­
TLS. Large mediastinal masses may be asso­ ued rise when therapy is initiated (assuming
ciated with rapid cell turnover and the this is a lymphoblastic leukemia/lymphoma
patient may have hyperleukocytosis and that would be highly sensitive to chemother­
metabolic derangements associated with apy). This patient’s calcium‐phosphorus
TLS including hyperkalemia, hyperphos­ product exceeds 60 (11.2 × 5.6 = 62.72),
phatemia, and hyper­ uricemia. Ideally, a putting the patient at risk for renal compro­
bone marrow aspirate (as well as lumbar mise. This may improve initially with IVF
puncture to evaluate CSF) would be done to but it is important to anticipate worsening
confirm or rule out the diagnosis of ALL TLS and potential interventions to avoid
or lymphoma. However, the patient’s airway renal­­compromise. The patient should not
Oncologic Emergencies 187

be alkalinized given the use of rasburicase cyclophosphamide, vincristine, and dauno­


as well as elevated ­ calcium‐phosphorus mycin. If the diagnosis is uncertain, other
product. diagnostic considerations must be enter­
d. The patient is worsening after tained including germ cell tumor, Ewing
24 hours of steroid treatment and maxi­ sarcoma, undifferentiated sarcoma, thy­
mal supportive care. What are the next moma, and thyroid cancer.
steps to be considered? The patient continued on steroids and
Given the patient is likely immune sup­ cyclophosphamide 1000 mg/m2 was initiated.
pressed and has a history of fever, blood cul­ The patient continued to worsen. Repeat
tures should be drawn and broad‐spectrum echocardiogram showed an increasing peri­
antibiotics initiated. Continuous assess­ cardial effusion and evidence of pulsus para­
ment of cardiac and respiratory status is doxus due to right outflow obstruction by the
vital with an echocardiogram to evaluate mass. With the continued deterioration, the
cardiac function. Oxygen delivered in a patient was put on extra‐corporeal mem­
comfortable manner via NRB mask or with brane oxygenation (ECMO) for heart–lung
positive pressure (given severe airway com­ bypass, so that diagnostic procedures could
pression) should be given. He was placed on be performed, and for more time for treat­
BiPAP and did not fight this, suggesting he ment to work and shrink the mass. The
was more comfortable with the mask and patient was carefully transitioned to the oper­
pressure. ating room (OR) and while in the upright
The electrolytes normalized with IV position and with mild sedation and meticu­
hydration and the uric acid dropped to lous care, cannulation of the vessels was per­
<0.5 ng/dl with rasburicase. The WBC also formed. The patient then had the pericardial
dropped to 10.5 × 109/l with stable hgb and effusion drained which immediately dropped
platelets. his BP (due to the weight of the mass now
Steroids (methylprednisolone) 1 mg/kg fully compressing the heart, which had been
IV q12 hours is the treatment of choice for protected by the effusion) and the patient was
life‐threatening presumed leukemia (given immediately placed on ECMO. In quick suc­
the chest mass, circulating abnormal cells, cession, flow cytometry of the peripheral
and evidence of TLS) prior to definitive blood resulted with confirmation of T‐cell
diagnostic studies. The benefits of shrinking ALL and the pericardial fluid also showed
the mass and relieving airway pressure out­ malignant cells of T‐cell origin. Four‐drug
weigh the need for obtaining a bone marrow induction with prednisone, daunomycin,
aspirate. Emergent radiation is also a con­ vincristine, and PEG‐asparaginase was
sideration to shrink the mass, though does immediately initiated. The patient showed
not confer an added benefit over steroids. significant response to treatment by hour 96
The patient therefore received steroids and was able to come off ECMO after
for 24 hours, but unfortunately with worsen­ 48 hours and was completely neurocogni­
ing clinical status and no improvement on tively intact. The diagnostic lumbar puncture
CXR. Response to treatment in this short was delayed several weeks, and the patient
period of time is not predictable and may was treated for presumed CNS involvement
not be a clue to diagnosis. T‐cell leukemia/ given the lack of diagnostic studies in lieu of
lymphoma may not respond this rapidly to the patient’s precarious status. IVF hydration
steroids. and management/prevention of TLS contin­
Consideration should be given to adding ued for 4 more days without significant fur­
another chemotherapeutic agent including ther abnormality.
188 Chapter 14

a. Placing the patient on a CR monitor


Multiple choice questions
and giving calcium gluconate
b. Giving albuterol
1. You are seeing a 12-year-old male patient
c. Giving insulin/glucose
in the emergency department with the
d. Consulting the oncology fellow and
complaint of trouble breathing while lying
the PICU
flat (orthopnea). All of the following are
e. All of the above
appropriate next steps EXCEPT:
Explanation: Tumor lysis syndrome occurs
a. Imaging to include CXR and CT scan
most commonly in non‐Hodgkin lymphoma
of chest, abdomen, pelvis
and ALL, especially in patients with bulky dis­
b. General anesthesia for bone marrow
ease and after the commencement of chemo­
aspirate and lumbar puncture in the am
therapy. Hyperuricemia, hyperphosphatemia
c. Checking baseline labs to include
and hyperkalemia can all occur. Hydration is
CBC/diff, CMP, phos, LDH, uric acid
key to preventing tumor lysis and for the
d. Starting IVF, D5 1/2NS without KCl
patient with hyperkalemia sudden cardiac
at 2x maintenance
arrest can occur, especially as the potassium
e. Starting allopurinol
increases above 6.5–7.0 mEq/l. Therefore the
Explanation: Given the symptomatology
patient should be placed on a CR monitor to
this patient likely has an anterior mediastinal
check for peaked T waves as well as giving
mass, most likely T‐cell lymphoblastic lym­
medications to shift potassium intracellular,
phoma (the other terrible Ts include thymoma,
namely with insulin and albuterol. These are
thyroid, teratoma are much rarer). Cardiac
temporizing mechanisms and such patients
return is compromised with an anterior medi­
may require dialysis so the PICU should be
astinal mass which is best determined by
contacted and the patient transferred.
orthopnea and CT of the chest. General
Kayexalate should also be started although
anesthesia can decrease cardiac return and
does take some time to work. The answer is e.
lead to cardiac arrest in these patients and is
contraindicated. Diagnostic procedures are
3. For the above patient you recheck the
still vital and are done with mild conscious
potassium and it is again now 4.0 mEq as it
sedation. In the emergent situation patients
turns out that the previous sample was
can receive steroids or radiation to treat the
hemolyzed. You do note that the uric acid
likely anterior mediastinal lymphoma. Tumor
has risen from 6 mg/dl to 7.5 mg/dl and the
lysis can occur so it is important to hydrate the
phosphorus has increased from 5 mg/dl
patient without potassium and start allopu­
to 7 mg/dl. The calcium has concomitantly
rinol to prevent formation of additional uric
dropped to 5.7 mg/dl. The patient is com­
acid. The answer is b.
plaining of mild paresthesias in the fingers
but otherwise is stable. Which of the follow­
2. You are following the above patient after ing is the best treatment at this point?
they have been diagnosed with T‐lympho­ a. Start alkalinizing fluids, D5 1/4NS
blastic lymphoma and chemotherapy has with 40 mEq/l NaHCO3
commenced. You are checking tumor lysis b. Increase fluids from 2x maintenance
labs every 6 hours and note that the potas­ to 3x maintenance
sium has increased from 4 mEq/l to now 6.7 c. Give a calcium bolus for the paresthesias
mEq/l. The patient is on 2x maintenance d. Give rasburicase (urate oxidase)
IVF without KCl. Which of the following e. No changes required, continue to moni­
interventions should be considered? tor labs q6 hourly
Oncologic Emergencies 189

Explanation: Similar to hyperkalemia, there upon using the central line, or if there is a
are no significant beneficial treatments for history of chills with flushing the line, a
hyperphosphatemia beyond renagel, a phos­ larger gauge peripheral IV should be placed
phate binder which decreases GI absorption for IVF and antibiotics rather than using the
of phosphate, increased IVF and dialysis. central line. For septic appearing patients,
Alkalinization increases renal excretion of administer meropenem and vancomycin for
uric acid but also leads to increased calcium broad coverage for potential GNR and GPC
phosphate precipitation so should not be bacteremia/sepsis, respectively. For the
used with hyperphosphatemia. Giving non‐septic appearing patient, monotherapy
calcium increases the calcium phosphate with cefepime is generally sufficient. The
product and also leads to increased calcium answer is b.
phosphate precipitation in the kidneys.
Urate oxidase is an effective treatment for 5. You are seeing a 13-year-old male patient
hyperuricemia but is generally reserved for who developed altered mental status and a
uric acid continuing to rise over 10 mg/dl seizure at home and is rushed to the ED. In
and should be given as a dose of 0.05 mg/kg the ED his mentation is slowed and he has
to a max of 3 mg. Similar to question 2., right sided weakness. A STAT head CT shows
the oncology fellow and PICU should be a left sided intracranial hemorrhage. Of note,
consulted as this patient may require dialy­ his CBC shows a hemoglobin of 8, platelets of
sis if the phosphorus continues to rise. The 24, fibrinogen of 75 mg/dl with a prolonged
answer is b. PT and PTT. The WBC count is 2.4 × 109/l
and the lab reports out blasts. Of the follow­
4. You are seeing an expected arrival to the ing, the most likely diagnosis is:
ED, a 4-year-old female with rhabdomyo­ a. Acute lymphoblastic leukemia
sarcoma who had a fever at home. She is tri­ b. Immune thrombocytopenic purpura
aged immediately to a room where she is c. DIC due to sepsis
noted to be tachycardic and hypotensive. d. Acute promyelocytic leukemia
There is no history of chills with line flush­ e. Chronic myelogenous leukemia
ing. All of the following next considerations Explanation: Patients with acute leukemia
are correct EXCEPT: can have a bleeding diathesis, especially
a. First assure that her airway and with thrombocytopenia. Patients with ALL
breathing are reassuring and severe hyperleukocytosis (generally
b. Give a 20 ml/kg IV bolus with a WBC >200‐400 × 109/l) may present with
peripheral IV the acute onset of intracranial bleeding.
c. Give a 20 ml/kg bolus through her Patients with acute promyelocytic leukemia
central line (APL; M3 on the FAB classification for
d. Start meropenem AML) are at particular risk of coagulopathy.
e. Start vancomycin Although all patients with AML may have
Explanation: This patient is having distribu­ bleeding and coagulopathy, the promyelo­
tive shock, most likely from sepsis. First cytes in APL characteristically have intra­
always ensure that you check the A and B of cellular granules which when release due
ABCs prior to treating the circulatory issues. to cell lysis lead to coagulopathy. These
Given the hypotension, a fluid bolus should patients may often present with severe
be given and can be given through the cen­ bleeding in addition to labs consistent with
tral line. If there are problems accessing the DIC. Choice a. is incorrect as patients with
central line, if the hypotension worsens ALL will generally not have significant
190 Chapter 14

bleeding with a low WBC count. Choice b.


Suggested reading
is not supported by the total lab findings.
Choice c. could be correct although there is
Henry, M. and Sung, L. (2015). Supportive care in
nothing in the history that supports sepsis pediatric oncology: oncologic emergencies
as a cause for DIC. Choice e. will often lead and management of fever and neutropenia.
to hyperleukocytosis but generally the Pediatr. Clin. North Am. 62: 27–46.
platelet count will be normal and there will Prusakowski, M.K. and Cannone, D. (2017).
not be an increased risk of hemorrhage. The Pediatric oncologic emergencies. Hematol.
answer is d. Oncol. Clin. North Am. 31: 959–980.
15 Acute Leukemias

Acute leukemia (AL) is the most common increased incidence of developing AL with
type of malignancy in children, accounting certain genetic conditions including: agam­
for approximately 25% of newly diagnosed maglobulinemia, ataxia telangiectasia,
cancers in patients less than 15 years of age. Shwachman–Diamond, Li–Fraumeni, neu­
Approximately 2500–3000 new cases are rofibromatosis, Diamond–Blackfan anemia,
diagnosed each year in the United States and Kostmann disease/severe congenital
with a peak incidence between 2 and 5 years neutropenia. When a child is affected, the
of age. Acute lymphoblastic leukemia (ALL) siblings have a risk up to four times the gen­
accounts for 75% of cases of AL, followed by eral population of developing AL, with this
acute myelogenous leukemia (AML) in 20%, risk markedly increased in identical twins
and the remainder being undifferentiated or (noting though that the baseline risk is
mixed phenotypic ALs (MPALs). The ALs extremely low and therefore the risk in sib­
are biologically classified by blast morphol­ lings remains extremely low). Though
ogy, surface proteins, cytogenetic abnor­ unclear at this time, inciting events in the
malities, and cytochemical staining. This development of leukemia in children and
information, in addition to clinical features, adolescents involves complex relationships
is utilized for risk stratification into treat­ between host genetic polymorphisms, envi­
ment groups. Risk‐based therapy optimizes ronmental exposures, and infections.
curative potential while minimizing risks
and side effects.
The etiology of AL is not known, though Acute lymphoblastic leukemia
most cases are likely associated with somatic
genetic alterations. Ionizing radiation and The majority of cases of ALL arise from B‐cell
exposure to benzene have been associated committed progenitors (pre‐B‐ALL). T‐cell
with an increased risk of developing leuke­ acute lymphoblastic leukemia (T‐ALL) repre­
mia. Exposure to alkylating agents, with or sents approximately 15% of ALL cases. T‐ALL
without radiation, has also been associated is associated with certain features at diagnosis
with an increased risk of developing AML. including older age, higher white blood cell
Certain syndromes with unique chromo­ (WBC) count, and presence of extramedul­
somal abnormalities are associated with a lary (i.e., outside the bone marrow) disease.
high incidence of developing leukemia such Until recently, outcomes for T‐ALL were sub­
as Down syndrome (DS), Fanconi anemia, optimal, but with tailored high‐risk therapies,
and Bloom syndrome. There is also an outcomes between pre‐B‐ALL and T‐ALL are

Handbook of Pediatric Hematology and Oncology – Children’s Hospital & Research Center Oakland,
Third Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
© 2021 John Wiley & Sons Ltd. Published 2021 by John Wiley & Sons Ltd.
192 Chapter 15

similar. Most patients (75%) have evidence of helped to identify and refine biologically
chromosomal changes in the malignant cell distinct ALL subsets. This technology helps
population. The molecular characteristics determine targeted therapy by identifying
have important biologic and prognostic sig­ unique genes and pathways. Patients with
nificance and have contributed to a sophisti­ “Ph‐like” (Philadelphia‐like) ALL comprise
cated understanding of the interplay between up to 15% of patients and have a uniquely
these features and treatment stratification. devastating prognosis. Recent discoveries
Hyperdiploidy (an excess of chromosomes; have categorized this group into JAK path­
e.g., >50) is present in approximately one‐ way and CRLF mutations, with now an
third of children with pre‐B‐ALL. opportunity to study directed therapy with
Hyperdiploidy and specific trisomies (chro­ JAK1/2 inhibitor ruxolitinib. Identification
mosomes 4 and 10) are associated with good of other high‐risk genes such as IKZF1 is
outcomes. The ETV6–RUNX1 fusion gene further refining risk stratification and
(formerly TEL–AML1) translocation t(12;21) treatment algorithms.
(p13;q22) detected by fluorescent in situ Another area of active discovery is related
hybridization (FISH) is present in 25% of to host pharmacogenomics and identifying
cases and also associated with an excellent gene polymorphisms that encode for drug‐
prognosis. Hypodiploidy (i.e., ≤44 chromo­ metabolizing enzymes which influence the
somes) is present in about 6% of cases with efficacy and toxicity of certain chemothera­
extremely poor prognosis, especially if with peutic agents. For example, patients that
<40 chromosomes in the malignant clone. carry the polymorphism for thiopurine
The MLL rearrangement (now KMT2A gene methyltransferase catalyze the inactivation of
mutations) at chromosome band 11q23 mercaptopurine. Up to 10% of the population
affects 80% of infants, 3% of ALL cases, and carry these genes, resulting in high levels of
85% of secondary AML cases, and is associ­ active metabolites with increased side effects
ated with a very poor prognosis despite inten­ and the need for marked reduction in dosing
sive therapy. The BCR–ABL fusion gene of 6‐mercaptopurine (6MP) during treat­
t(9;22)(q34;q11) (Philadelphia chromosome) ment. NUDT15 is a recently discovered
occurs in 3% of childhood ALL in contrast to genetic polymorphism which also impacts
adult ALL (25%) and chronic myelogenous 6MP metabolism.
leukemia (CML) (95%). The breakpoint in
pediatric ALL produces a 190‐kDa fusion Clinical presentation
protein in contrast to the classic 210‐kDa The clinical manifestations of leukemia are a
fusion protein in CML. This fusion gene has direct result of marrow invasion with result­
previously been associated with exceptionally ant cytopenias (i.e., anemia, thrombocytope­
poor outcomes, but now with the addition of nia, leukopenia, neutropenia) and, rarely,
tyrosine kinase inhibitors (i.e., imatinib), out­ extramedullary (outside the marrow)
comes are approaching other high‐risk (HR) involvement (see Table 15.1). Children typi­
ALL patients. The intrachromosomal amplifi­ cally present with nonspecific symptoms
cation of chromosome 21 (iAMP21) present related to these cytopenias. Symptoms of
in 2% of ALL cases has been associated with fatigue, irritability, and anorexia as well as
inferior outcomes, but patients treated on clinical signs of pallor, tachycardia, and more
more intensive regimens are seeing improved rarely evidence of congestive heart failure
outcomes. are a result of the anemia. Low‐grade fever
Gene expression profiling and next‐ may be present due to leukemia‐related cyto­
generation sequencing technology have kine release or secondary to infection with
Acute Leukemias 193

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

underlying neutropenia and immunosup­ count >100 × 109/l (30–50%). The presence
pression. Bleeding secondary to thrombocy­ of an anterior mediastinal mass may cause
topenia is usually mild with petechiae, airway or cardiovascular compromise (see
bruising, gingival oozing, or epistaxis. Life‐ Chapter 14).
threatening hemorrhage is very rare. Bone Laboratory abnormalities frequently pre­
pain (typically long bones) is common and sent at diagnosis include elevated liver
may result in refusal to walk or irritability in enzymes, uric acid (UA), and lactate dehydro­
young children. The pain is usually due to genase (LDH). Evidence of tumor lysis related
leukemic infiltration of the periosteum or to a large tumor burden and rapid cell turno­
expansion of the marrow cavity by leukemic ver may result in hyperuricemia, hyper­
cells. Pathologic fractures may also be pre­ kalemia, and hyperphosphatemia (with
sent at diagnosis and cause acute severe resultant hypocalcemia due to maintenance of
pain (see Table 15.2). Patients with T‐ALL the calcium phosphate product). Tumor lysis
may present with some distinctive clinical syndrome (TLS) may also occur with the ini­
features. T‐ALL is more frequent in males tiation of treatment and should be anticipated
and associated with a higher incidence of to become clinically significant in those with
central nervous system (CNS) leukemia at elevated WBC (i.e., >100 × 109/l) or bulky dis­
diagnosis compared to pre‐B‐ALL (10–15% ease (organomegaly, adenopathy, mediastinal
vs. 2–5%). T‐ALL patients are more likely to mass) (see Chapter 14).
present with a mediastinal mass (nearly CNS involvement is usually detected in
50%), bulky lymphadenopathy, or a WBC an asymptomatic child with analysis of the
194 Chapter 15

Table 15.2 Radiographic changes Table 15.3 Differential diagnosis of acute


of the bones in leukemia. lymphoblastic leukemia (ALL).

Osteolytic lesions involving the medullary Nonmalignant disorders


cavity or cortex Aplastic anemia
Subperiosteal new bone formation; Immune thrombocytopenia
pathologic fracture Infections: EBV, CMV, pertussis,
Transverse metaphyseal radiolucent bands parapertussis
Transverse metaphyseal lines of increased Malignant disorders (round blue cell
density (growth arrest lines) tumors with marrow involvement)
Lymphoma
Neuroblastoma
cerebrospinal fluid (CSF). Rarely, a patient Retinoblastoma
may present with signs or symptoms of Medulloblastoma
Rhabdomyosarcoma
increased intracranial pressure (e.g., cranial
nerve VI palsy with resultant esotropia and Abbreviations: EBV, Epstein–Barr virus;
diplopia, papilledema, visual changes, CMV, cytomegalovirus.
morning headache, vomiting, lethargy, irri­
tability, possible seizures) or evidence of
parenchymal involvement, hypothalamic Laboratory and imaging studies indi­
syndrome, or diabetes insipidus. Other rare cated are:
●● Complete blood count (CBC), differen­
complications such as chloromas (mass of
malignant cells) causing compression of the tial, review of peripheral blood smear by an
spinal cord or CNS hemorrhage (related to experienced individual.
●● Bone marrow aspirate (consider biopsy
leukostasis and coagulopathy) are more
likely to be associated with AML. for dry tap or inadequate specimen) for
Leukemic involvement of the testes morphology, blast count, immunopheno­
occurs in 2–5% of boys at diagnosis and pre­ typing by flow cytometry, karyotype, and
sents as painless enlargement, either unilat­ molecular studies.
●● Metabolic panel, to include liver function
erally or bilaterally. Early testicular
involvement is also associated with T‐cell studies, electrolytes, LDH, UA, phosphorus,
disease, elevated WBC count, and lympho­ calcium, blood urea nitrogen, and
matous features. creatinine.
●● Blood culture if febrile; cultures of any
Infants with ALL may present with skin
infiltration known as leukemia cutis. Skin suspected sites of infection.
●● Chest radiography to evaluate for the
lesions are a result of dermal infiltration by
malignant cells and may appear skin‐colored presence of a mediastinal mass.
●● Plain films of the long bones for patients
or violaceous. They may also be seen in chil­
dren with AML. presenting with pain to evaluate for patho­
logic fracture or evidence of leukemic
Diagnostic evaluation changes.
Children presenting with more than one Typically, the bone marrow is 80–100%
cytopenia and clinical symptomatology sug­ replaced with lymphoblasts at diagnosis, and
gestive of bone marrow infiltration should there is a paucity of normal cellular differen­
undergo a diagnostic evaluation to investi­ tiation. Megakaryocytes tend to be absent. At
gate for the possibility of AL. Other diagno­ times, due to the packed condition of the mar­
ses to be considered are in Table 15.3. row, it may be difficult to aspirate a sample
Acute Leukemias 195

(dry tap) and a bone marrow biopsy can pro­ status) requires the presence of five or more
vide diagnostic samples. The diagnosis of AL WBCs/μl and identification of blasts on the
requires the presence of 25% or more blasts CSF cytocentrifuge examination. CNS leu­
(M3 bone marrow); however, the diagnosis is kemia is classified as follows:
suspected when the marrow contains 5% or ●● CNS1: no detectable blasts.

more blasts (M2 bone marrow, with M1 being ●● CNS2a,b,c: <5 WBCs/μL, blasts present

a normal bone marrow with <5% blasts) (a: <10 RBC; b: ≥10 RBC; c: ≥ 10 RBC, ≥5
(Table 15.3). Lymphoblasts have a typical WBC, but negative by Steinherz/Bleyer
morphology with a high nuclear to cytoplas­ algorithm, see below).
mic ratio, fine nuclear chromatin, and the ●● CNS3a,b,c: ≥5 WBCs/μL and blasts pre­

presence of nucleoli. In addition, the cells sent (a: <10 RBC; b: ≥10 RBC, positive by
tend to have a uniform size and appearance. Steinherz/Bleyer algorithm, see below; c:
Immunophenotyping and immunohisto­ signs of CNS leukemia [i.e., facial nerve
chemistry are used to differentiate ALL and palsy, hypothalamic syndrome, brain/eye
AML and identify T‐ALL and pre‐B‐cell involvement].
ALL. Lymphoid malignancies should lack
A traumatic LP is defined as the presence of
myeloid cell surface markers (i.e., myeloper­
≥10 red blood cells/μl. Formulas are avail­
oxidase [MPO], CD13 and CD33; CD [clus­
able to assist with interpretation of CNS
ter of differentiation]). Pre‐B‐ALL should
status in the event of a traumatic initial tap
have B‐cell surface markers such as CD10,
with blasts on cytospin (Steinherz/Bleyer
CD19, kappa, and lambda, whereas T‐cell
algorithm in which CNS disease is present
ALL should have T‐cell surface markers
[positive] if CSF WBC/CSF RBC >2× blood
including CD2, CD5, and CD7. ALL cells
WBC/blood RBC).
should also stain for ­terminal deoxynucle­
otidyl transferase (TdT), an immature lym­
phoid marker. Other assessments prior
After the diagnosis is confirmed, evalua­ to initiation of therapy
tion of the CSF by lumbar puncture (LP) Echocardiogram and electrocardiogram are
should be done to determine CNS involve­ done as baseline studies in all patients who
ment. CNS status is defined based on the will receive anthracyclines. These drugs are
WBC chamber count and the presence of frequently given in induction for high‐risk
leukemic blasts on a centrifuged specimen. patients and in the delayed intensification
Many patients require a platelet transfusion (DI) phase for all patients. Acute and delayed
prior to the initial LP, which should be per­ cardiotoxicity may occur, though late com­
formed by an experienced clinician to plications are more often related to high
decrease the chance of a traumatic tap, which cumulative doses (i.e., >300 mg/m2) given at
may theoretically predispose the patient to a young age (<4 years) and asso­ciated with
CSF seeding with leukemic blasts. There is other risk factors for cardiac disease such as
evidence that a platelet count ≥100 × 109/l mediastinal radiation, obesity, family history
for the diagnostic LP may decrease this risk. of early cardiovascular disease, and abnormal
Subsequent LPs should be done with platelet lipid profiles.
counts of ≥20–50 × 109/l, per institutional Viral serologies for hepatitis B and C,
guidelines. A cell count and cytocentrifuge cytomegalovirus (CMV), herpes simplex
examination for cell morphology is done virus (HSV), and varicella zoster virus
on the fresh CSF (i.e., within 1 hour). The (VZV) are obtained as baseline information
diagnosis of overt CNS leukemia (CNS3 (see Chapter 25). This aids in determining if
196 Chapter 15

later infection with one of these viral agents therapy based on flow cytometric response
could be related to transfusion (potentially to therapy (as measured by day 8 periph­
in the case of hepatitis B and C), and whether eral blood and day 29 [end of induction]
the infection is primary or reactivation (in bone marrow aspirate) and cytogenetic/
the case of CMV, HSV, and VZV). Baseline molecular features identified from the diag­
knowledge of previous exposure may impact nostic marrow. Patients with negative bone
later treatment choices. Other screening marrow minimal residual disease (MRD)
includes immunoglobulin G in patients with measured by multiparameter flow cytometry
increased risk of infection such as infants at end of induction have an excellent prog­
and those with DS. nosis. Other considerations include presence
of extramedullary disease such as testicular
Risk group classification or CNS disease.
The concept of risk stratification allows
patients at comparatively higher risk of Treatment
relapse to be treated with more intensive, Treatment of childhood leukemia is one of
molecularly guided, and potentially more the success stories in pediatric oncology,
toxic therapies, whereas patients at lower risk though much remains to be accomplished.
of relapse are treated with lower intensity Patients with SR ALL have a 4‐year event‐
regimens that maintain the same low risk of free survival (EFS) of over 90%. High‐risk
recurrence while minimizing exposure to patients have an EFS of 75%, though certain
unnecessary agents, decreasing toxicity. subgroups including infants and those with
Classification into risk groups previously very‐high‐risk molecular features may be
was based on clinically apparent prognostic at 50% or less. Remarkable advances have
factors identified by retrospective analysis occurred by the identification of more effec­
and then verified by prospective studies. tive drug combinations, recognition of drug
Age, WBC, and immunophenotype (i.e., sanctuary sites with routine presympto­
AML vs. ALL, T‐ALL vs. B‐cell ALL) are uti­ matic CNS‐directed therapy, response‐based
lized for initial classification while awaiting intensification of therapy in early phases,
cytogenetic results from the leukemia clone and identification of clinical and biological
to determine high‐ and low‐risk features and variables predictive of outcome. Certain fea­
subsequently treatment response. tures, such as CNS2 involvement, continue
The National Cancer Institute (NCI) crite­ to be analyzed and treatment refined to
ria classify standard risk (SR) ALL as children ensure the best possible outcomes. Most
1 to <10 years of age with a WBC <50 × 109/l children with cancer in the United States are
and HR ALL as children with age ≥10 years treated at specialized pediatric oncology
and/or WBC ≥50 × 109/l. Utilizing these crite­ centers and have access to participation in
ria, approximately two‐thirds of B‐ALL and clinical trials. These studies have accelerated
one‐third of T‐ALL patients are classified as the advances made in the diagnosis and
SR. Children under 1 year of age are catego­ treatment of pediatric cancers and clearly are
rized as infant ALL, with a worse prognosis. a major reason for many of the successes.
Overall, better outcomes are observed in Pediatric oncology centers provide expertise
younger children (over the age of 1), females, in the management of pediatric oncology
pre‐B‐ALL, and those with more favorable patients with consi­deration of the unique
cytogenetic features. aspects of protocol management and sup­
Further stratification into treatment portive care that are critical to optimizing
groups is done within the first few weeks of survival and quality of life. The Children’s
Acute Leukemias 197

Oncology Group (COG) is a national col­ an attempt to improve outcomes and dimin­
laborative group of multidisciplinary profes­ ish acute and late toxicity. Some examples
sionals involved in the treatment of children may include adding a new agent in certain
with cancer. Most pediatric cancer centers phases, altering chemotherapy dosing or
(over 200 nationally) register patients on timing in specific phases, or possibly asking
COG trials. Families are asked to give a radiation‐related question. The addition of
informed consent to participate in these new agents such as molecularly targeted and
trials, which may include epidemiological, immunotherapeutics is becoming more fre­
clinical, and biological questions. For fami­ quent and can be expected to continue to be
lies not wishing to participate, the standard integrated into phase 3 trials.
of care is offered (best published clinical Conventional induction therapy includes
regimen). Many centers also participate in prednisone or dexamethasone, PEG‐aspara­
other smaller multicenter collaborative ginase, and vincristine in addition to intrath­
studies or have sufficient numbers of patients ecal cytarabine and methotrexate. Patients
to support their own clinical protocols. with HR ALL also receive daunorubicin for a
The purpose of therapy in ALL is to induce four‐drug systemic induction. Consolidation
a permanent biologic and clinical remission for HR patients incorporates cyclophospha­
(no evidence of disease on laboratory and mide, cytarabine, PEG‐asparaginase, 6‐mer­
physical assessment). Overall, approx­imately captopurine (6‐MP), and intensified therapy
85% of children with ALL will be cured of directed toward the CNS (either intrathecal
their disease. Treatment regimens are classi­ methotrexate alone or with radiation for
cally divided into phases of therapy: patients with CNS3 disease and certain
●● Induction: refers to the first 28–35 days of T‐ALL subgroups). SR patients receive
therapy, after which the child should be in a intensified CNS therapy with intrathecal
morphologic remission. Remission induc­ chemotherapy. The DI phase has led to sig­
tion rate in SR ALL is 98%. nificantly improved outcomes as has further
●● Consolidation: further systemic chemo­ intensification of the IM phase. DI is a re‐
therapy is given, in addition to a focus on intensification of therapy following attain­
CNS prophylaxis. ment of remission with some alteration in
●● Interim Maintenance (IM): similar drugs drugs utilized previously in the induction
as maintenance, but in a more intensified and consolidation phases. Maintenance
manner. classically consists of pulses of steroid (pred­
●● Delayed Intensification (DI): refers to nisone or dexamethasone) and vincristine in
re‐induction and reconsolidation with many addition to oral methotrexate and 6‐MP.
of the same medications as these phases. Individualized drug dosing is required to
●● Maintenance: continuation of therapy; this maintain certain hematologic levels with
phase lasts 2-3 years, timed from start of IM differences in metabolism and tolerability
(possibly longer in males due to increased related to individual polymorphisms (e.g.,
risk for relapse, per institutional guidelines). thiopurine methyltransferase (TMPT) with
●● CNS‐directed therapy: intrathecal cytara­ 6‐MP metabolism).
bine (Ara-C), methotrexate; cranial radiation CNS prophylaxis continues with intra­
(treatment or prophylaxis) in select groups thecal methotrexate throughout therapy.
(i.e., CNS3, certain T‐ALL subgroups). Children with CNS3 disease receive augmented
therapy with 1800 cGy cranial radiation, in
Open clinical trials will likely alter some addition to intrathecal methotrexate. Sub­
aspect of therapy for certain risk groups in groups of patients with T‐ALL may require
198 Chapter 15

cranial radiation prophylaxis to 1200 cGy, These anticipated side effects should also be
though this continues to be studied within discussed in detail with the patient and fam­
newer protocols. Treatment of children with ily in the consent process and as part of
CNS2 disease has been a focus of recent ongoing care, to help with earlier recognition
review and these patients now receive addi­ and supportive care initiation as needed.
tional intrathecal therapy. CNS‐directed Potential late toxicities are monitored
therapy includes intrathecal drug delivery as throughout the course of treatment and in
well as systemic medications that penetrate off‐therapy follow‐up.
the blood–brain barrier in cytotoxic concen­
trations. Substituting dexamethasone for
Relapse
prednisone in some phases of treatment as
Despite the dramatic improvements in our
well as using high‐dose methotrexate in lieu
knowledge of the biology of ALL, prognostic
of lower doses has led to decreased CNS
factors, and more targeted therapy, up to 20%
relapses. Patients that are CNS1 (no overt
of children with ALL will relapse, with only
CNS disease) also benefit from CNS‐directed
about one‐third of those patients going on to
treatment. Without CNS prophylaxis, it is
have a long‐term remission with intensified
estimated that 60–70% of children would
therapy. Due to the frequency of ALL as com­
relapse in the CNS (based on historical data).
pared to other pediatric malignancies,
relapsed ALL is the fourth most common
Complications of therapy oncologic diagnosis in children. Timing and
Due to the myelosuppressive nature of site of relapse are prognostic and important
therapy, and the underlying disease state, determinants for future therapy. Sites of
children undergoing chemotherapy or relapse include the bone marrow or an
radiation should expect to experience com­ extramedullary site, such as the CNS or testes,
plications during the course of their treat­ or some combination of sites. Early relapse
ment. Many of these complications can be (<18 months since therapy initiation) is asso­
anticipated and prevented or ameliorated ciated with a particularly grim prognosis.
(see Chapter 25). Most patients will require MRD status at the end of the first block of re‐
transfusion support with packed red blood induction therapy also strongly correlates
cells and/or platelets (see Chapter 5). with outcome, with improved survival in
Children with neutropenia are instructed to those who are negative. Novel agents such as
take special precautions to decrease the risk blinatumomab (anti‐CD19 antibody conju­
of infection exposure. Any child receiving gated to CD3) and inotuzumab (anti‐CD22
chemotherapy who develops a fever should monoclonal antibody) are examples of tar­
have emergent medical attention, especially geted therapies being studied in this setting.
during phases of anticipated neutropenia Concepts of therapy include systemic retreat­
(i.e., induction, consolidation, DI; see ment and radiation therapy to sites of
Chapter 27). Certain therapies are associated extramedullary relapse, though emerging tar­
with specific toxicities: anthracyclines with geted therapies and immunotherapies may
cardiotoxicity, vincristine with peripheral become more important in this group of
neurotoxicity, intrathecal methotrexate with patients. Hematopoietic stem cell transplant
neurotoxicity, steroids (especially dexameth­ (HSCT) may improve the outcome of patients
asone) with avascular necrosis of bone, and with bone marrow or combined relapse.
radiation with acute and late effects depend­ Overall survival after relapse is also impacted
ing on site and dosage (see Chapter 30). by cumulative toxicity from prior therapy and
Acute Leukemias 199

high infection rates due to the increased increasingly important and effective therapies
intensity of therapy. In general, children with in the treatment of conditions such as ALL.
late bone marrow (i.e., >36 months from ther­
apy initiation) or isolated relapse (>18 months
from therapy initiation) fare better.
Acute myelogenous leukemia

Novel agents Approximately 20% of acute childhood


Various novel agents are under investigation leukemia is myelogenous, equating to about
to improve outcomes for high‐risk and 500 new cases per year in the United States.
relapsed patients. Previous such targeted Unlike ALL, AML does not have a peak age
agents included the tyrosine kinase inhi­ incidence in children, as there are only subtle
bitors (imatinib, dasatinib and others) for increases in incidence from infancy to ado­
Philadelphia‐chromosome‐positive ALL lescence. AML is associated with a poorer
and now JAK1/2 inhibition in “Ph‐like” ALL prognosis than ALL, with long‐term survival
(i.e., CRLF2‐rearranged and JAK pathway‐ expected to be 50–60%. Additionally, therapy
mutant). Finding agents with favorable tox­ for AML, which includes multiagent chemo­
icity profiles for use in highly treated relapse therapy and HSCT, is one of the most toxic
patients is a challenge. Targeted antibodies therapies in childhood cancer. Hispanic
such as blinatumomab and inotuzumab are children are noted to have a higher incidence
attractive due to their efficacy and decreased of AML, mostly accounted for by the acute
off‐target effects. Immunologic therapies promyelocytic leukemia (APL) subtype.
are of great interest at this time and offer tre­ The etiology of AML is mostly sporadic,
mendous hope. Rituximab, an anti‐CD20 although several inherited and acquired risk
monoclonal antibody utilized in many con­ factors may significantly predispose to this
ditions such as non‐Hodgkin lymphoma, disease. Children with DS have a 10–20‐fold
is being investigated in mature B‐cell ALL increased risk of developing AL compared
(a notoriously poor prognostic group). to other children. In early life, children
Blinatumomab, a bispecific T‐cell engager with DS have a particularly high risk of the
anti‐CD19 antibody, which acts by directing megakaryoblastic subtype of AML (AMKL).
the patient’s cytotoxic CD3‐postive T‐cells Overall, children with DS constitute approx­
to CD19‐positive leukemic cells and thereby imately 10% of children with AML. In addi­
activating T‐cells to destroy the malignant tion, approximately 10% of neonates with
cells, has shown benefit for relapsed and DS may develop a transient myeloprolifera­
refractory pre‐B‐ALL. Engineering autolo­ tive disorder (TMD) that mimics congenital
gous T‐cells to express chimeric antigen AML but typically improves spontaneously
receptors targeting leukemia cells (CAR‐T by 4–6 weeks of age. However, these chil­
cells) has been an extremely exciting, poten­ dren have a 20–30% risk of developing AML
tially curative therapy in refractory or multi­ before 4 years of age. Children with TMD or
ply relapsed CD19‐positive pre‐B‐ALL with presumed congenital AML who do not
recent FDA approval of a Novartis product display phenotypic features of DS should
called tisagenlecleucel. Harnessing other have a karyotype performed to look for
cell populations which are important for possible trisomy 21 mosaicism.
tumor cell surveillance and destruction Patients with inherited bone marrow
such as natural killer cells, macrophages, failure syndromes such as Fanconi anemia,
and dendritic cells are likely to become severe congenital neutropenia or Kostmann
200 Chapter 15

syndrome, and Shwachman–Diamond marrow and other body organs. Depending on


syndrome have very high rates of deve­
­ the total WBC and percentage of circulating
loping AML (30–50% risk). Other genetic myeloblasts, some of the presenting mani­
­conditions that predispose to development of festations may be life‐threatening. Chil­dren
AML (and some other malignancies) include: with extreme leukocytosis (i.e., WBC >200
Bloom syndrome, dyskeratosis congenita, × 109/l) may present with metabolic abnor­
congenital amegakaryocytic thrombocyto­ malities and leukostasis (see Chapter 14) and
penia, Li–Fraumeni syndrome, neurofibro­ be at risk for sudden events. Clinically, they
matosis‐1, and Diamond–Blackfan anemia. are at risk for hypoxia in the small vessels of
Acquired conditions with an increased risk the brain and lungs as a result of increased
of AML include aplastic anemia, acquired blood viscosity related to the rheology of the
paroxysmal nocturnal hemoglobinuria, myeloblasts (large, adherent, and nondeform­
myeloproliferative syndrome, and myelod­ able cells) and decreased flow through small
ysplastic syndrome. Twins and siblings of vessels. This may translate to sludging in the
AML patients have an increased risk of small vasculature of the retinal, pulmonary,
AML, though this is much more significant and CNSs with resultant decreased oxygen
in monozygotic twins within 6 years of initial delivery, hypoxia, and stroke (Table 15.4).
diagnosis. The incidence of secondary AML Patients typically present with signs and
following treatment of childhood cancer is symptoms related to pancytopenia with pal­
rising, due in part to increased survival after lor, fatigue, anorexia, and bleeding. Fever is
primary malignancy as well as increased very common at presentation, often with no
use of agents that cause direct DNA damage apparent infectious etiology. Bone pain is
(e.g., alkalytors [cyclophosphamide, ifosfa­ also common, though not typically associ­
mide], anthracyclines [doxorubicin, dauno­ ated with fractures. Some unique and sug­
rubicin], radiation therapy), and those that gestive features of AML at presentation
inhibit topoisomerase II (e.g., epidophyllo­ include leukemia cutis in infants (blueberry
toxins [etoposide] and anthracyclines). muffin rash thought secondary to extramed­
Exposure to ionizing radiation has also been ullary hematopoiesis) and myeloid sarcomas
convincingly implicated, especially following (also known as chloromas) which are
the atomic bombs in World War II. extramedullary collections of myeloblasts.
Extramedullary disease occurs in 10–20% of
Clinical presentation patients with AML, much higher than seen
AML and ALL are indistinguishable at pres­ in ALL. Myeloid sarcomas may occur in up
entation, with the majority of signs and to 10% of children with AML and present
symptoms being related to infiltration of the most commonly in the orbit, periorbital

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 (increasing risk for tumor lysis syndrome)

Abbreviation: CNS, central nervous system.


Acute Leukemias 201

areas, bones, skin, soft tissues, and lymph fibrinogen, and D‐dimers to evaluate for the
nodes, but may occur anywhere in the body. possibility of disseminated intravascular
Gingival hypertrophy is a unique finding in coagulation (most typically seen in M3).
AML secondary to gum infiltration by Decreased factors VII, IX, and X may be seen
malignant cells. A particularly rare yet related to consumption. Other necessary
important presentation is spinal cord com­ assessments are similar to ALL, as previously
pression due to myeloid sarcoma in the par­ outlined.
aspinal/epidural region. AML may present
with only evidence of extramedullary dis­ Risk group classification
ease, but is treated in the same manner as As with ALL, classification of AML is based on
with marrow involvement. morphologic features, as well as immunohis­
Life‐threatening bleeding is another tochemical, biologic, and molecular character­
complication somewhat unique to AML. istics. Several morphologic classification
Although the majority of patients will be systems are available for AML, including the
thrombocytopenic secondary to marrow classic French–American–British (FAB) clas­
infiltration with myeloblasts, patients with sification of M0 to M7 (Table 15.5) and the
concomitant coagulopathy due to dissemi­ World Health Organization (WHO) catego­
nated intravascular coagulation are at par­ rization (now the preferred classification
ticularly high risk of bleeding, a finding seen method). The WHO classification system was
most commonly in APL. This association is developed to include cytogenetic, disease biol­
thought to be secondary to the release of ogy, and clinical history. Categories include: (i)
thromboplastin from cytoplasmic granules AML with recurrent genetic abnormalities; (ii)
in the promyelocytic blasts. AML with myelodysplasia‐related changes;
TLS is unlikely in AML. See Chapter 14 for (iii) therapy‐related myeloid neoplasms; (iv)
management of hyperleukocytosis and TLS. AML not otherwise specified; (v) myeloid sar­
coma; (vi) myeloid proliferations related to DS
Diagnostic evaluation (transient abnormal myelopoiesis and myeloid
The same studies suggested for the evalua­ leukemia associated with DS); and (vii) blastic
tion of ALL should be obtained in the evalu­ plasmacytoid dendritic cell neoplasm.
ation of the child with suspected AML. It is Chromosome analysis is performed to
common to see severe anemia and thrombo­ obtain important diagnostic and prognostic
cytopenia in association with an elevated information. Approximately 60% of children
total WBC count. The definitive diagnosis is with primary AML demonstrate clonal abnor­
made with the evaluation of the bone mar­ malities in the blast lineage prior to therapy
row, with at least 20% of the cellular elements initiation. The t(8;21)(q22;q22) RUNX1–
identified as myeloblasts based on immu­ RUNX1T1 translocation is the most common
nophenotyping and cytochemistry and con­ chromosomal translocation and is associated
firmed by flow cytometric assessment. with the FAB M2 subgroup and a more favora­
Morphologic review may also reveal Auer ble prognosis. The t(15;17)(q22;q12) produces
rods (clumps of azurophilic granules shaped a balanced translocation of the retinoic acid
into elongated needles and located in the receptor‐alpha (RARα gene at 17q12) and the
cytoplasm), classically only seen in some PML gene at 15q22 leading to APL (FAB M3).
subtypes of AML (i.e., M2 and M3). A LP Defect in RARα induces an arrest in myeloid
should be done to evaluate for CNS involve­ differentiation at the promyelocytic (PML)
ment. A coagulation evaluation should be stage. All‐trans retinoic acid (ATRA) over­
performed consisting of PT, INR, PTT, comes this blockage at the RAR site, allowing it
202 Chapter 15

Table 15.5 Morphologic classification for acute myelogenous leukemia (AML).

AML Subtype Clinical/biologic features

M0 Acute undifferentiated Similar to ALL, requires CD13, CD33, CD117, and


leukemia absence of lymphoid markers
M1 Acute myeloblastic MPO+ by special stains/flow cytometry
leukemia without
differentiation
M2 Acute myeloblastic Auer rods, t(8;21)(q22;q22), AML1–ETO fusion gene
leukemia with common, chloromas, good prognosis
differentiation
M3 Acute promyelocytic Auer rods, DIC/bleeding, t(15;17), good prognosis
leukemia with ATRA/arsenic therapy
M4 Acute myelomonoblastic Mixed myeloblasts (20%) and monoblasts, peripheral
leukemia monocytosis, inv(16), t(16;16) producing chimeric
protein CBFB–MYH11, favorable prognosis
M5 Acute monocytic ≥80% marrow nonerythroid cells are monocytic, KMT2A
leukemia (11q23) rearrangements in infants, CNS disease more
common, chloromas (e.g., gingival hyperplasia)
M6 Erythroleukemia (Di Rare in children
Guglielmo disease)
M7 Acute megakaryoblastic Classically seen in Down syndrome with GATA1
leukemia mutation, good prognosis; rare in non‐DS with
t(1;22), poor prognosis; associated with myelofibrosis

Abbreviations: ALL, acute lymphoblastic leukemia; MPO, myeloperoxidase; DIC, disseminated


intravascular coagulation; ATRA, all‐trans retinoic acid; CNS, central nervous system; DS, Down
syndrome.

to function normally while arsenic clears the monosomy 5 or 5q deletion, and complex
PML molecule. Inversion of chromosome 16, abnormalities (≥3 or more unrelated
inv(16)(p13.1;q22) or t(16;16)(p13.1;q22), changes) all impart a worse prognosis.
CBFB–MYH11, is associated with acute Monosomy 5 and 7 are also highly associated
myelomonoblastic leukemia (FAB M4) and with cases of secondary myeloproliferative
also portends a favorable prognosis. disorders and AML that develop following
Megakaryoblastic AML (FAB M7) is associ­ treatment with chemotherapy for a primary
ated with t(1;22)(p13;q13), RBM15–MKL1. malignancy. Gene expression profiling by
FMS‐like tyrosine kinase mutations internal microarray technology has identified distinct
tandem duplications (FLT3‐ITD) or point expression profiles of AML based on many
mutations are identified in up to 30% of chil­ genetic mutations, and will likely become
dren with AML. ITDs result in constitutive more important in the future to connect
activation of the kinase and uncontrolled molecular signatures with targeted therapies
proliferation of malignant blasts. FLT3‐ITD is and clinical outcomes.
associated with a high WBC at diagnosis Host factors such as age, sex, race, and
and poorer prognosis. Chromosomal aber­ constitutional abnormalities all have prog­
rations including monosomy 7 or 7q deletion, nostic influence. Females do slightly better
Acute Leukemias 203

than males, older children do better than etoposide, 6‐thioguanine, and L‐asparagi­
infants (though outcomes in infants are nase. Gemtuzumab, a monoclonal antibody
improving with current therapies), and to CD33, continues to be studied but appears
Caucasians fare better than non‐white beneficial in inducing remission, especially
patients. Children with DS, particularly those in those patients with increased CD33
under the age of 4 years at diagnosis (driven expression. Patients with FLT3‐ITD should
primarily by GATA1 mutation), have an be treated with a tyrosine kinase inhibitor
improved outcome with less intensive ther­ such as sorafenib, with more specific agents
apy (overall survival 80%) but also show such as quazartinib and gilteritinib being
increased toxicity with therapy. Children studied for improved efficacy. Prior studies
with an initial WBC <20 × 109/l have an have shown no added benefit for a main­
improved outcome and those with hyperleu­ tenance phase following intensification.
kocytosis (i.e., WBC >100 × 109/l) have a Allogeneic HSCT is recommended for
worse outcome. Risk features have been sim­ high‐risk groups and those without induc­
plified to include cytogenetics and MRD tion remission. Patients with favorable risk
response at the end of induction. cytogenetics and good early responses to
induction therapy receive allogeneic HSCT
Treatment only in the case of relapse and if a second
As with ALL, the goal of induction therapy in remission has been achieved.
AML is to induce a clinical and biological The morphologic subtypes of AML with
remission by rapidly reducing the number of highest involvement of the CNS are FAB M4
malignant cells. Approximately 80% of chil­ and M5. Overall, the frequency of CNS
dren achieve remission with induction ther­ involvement is between 15 and 20% at diag­
apy, and approximately 60–70% achieve a nosis. Unlike ALL, CNS disease in AML
long‐term remission. Two distinct subsets of does not affect survival, so is not included in
AML do particularly well: APL (survival risk stratification. Intrathecal chemotherapy
>95%) and children with DS (survival >80%). is given to all patients, regardless of detection
Treatment failure is due to relapse or treat­ of blasts in the CSF. All patients receive CNS
ment‐related mortality. Improvements in consolidative therapy with intrathecal cyta­
survival rates are due to more intensified rabine in addition to systemically adminis­
treatment regimens based on risk stratifica­ tered high‐dose cytarabine, which has good
tion, early identification of complications and blood–brain barrier penetration. AML
aggressive supportive care, and better treat­ patients with myeloblasts in the CNS receive
ment options for relapsed/refractory disease. intrathecal cytarabine on a biweekly basis in
The most effective induction regimens induction until the CSF is clear. Cranial irra­
use a combination of cytarabine and an diation is reserved for those children with
anthracycline, typically daunorubicin. refractory CNS involvement. Children with
Individual trials have built upon this back­ DS rarely have CNS involvement, and due to
bone with changes in dosing and timing, their increased risk of toxicity, CNS prophy­
alternative anthracyclines, and inclusion laxis has been greatly reduced without nega­
of aggressive supportive care strategies to tive consequence in these patients.
better manage potential adverse effects at APL is distinctly unique from the other
presentation or with therapy including AML subtypes, comprises approximately
coagulopathy, side effects of hyperleukocy­ 5–10% of pediatric AML cases, and has a
tosis and infection. Other important agents particularly good prognosis due to the devel­
utilized in continuation therapy include opment of targeted therapies. Approximately
204 Chapter 15

3% of patients with APL die in the early ommended in addition to antibacterial


phases of therapy as a result of the unique (i.e., fluoroquinolone) and PCP prophylaxis.
hemorrhagic complications of this disease. Streptococcus viridans sepsis is associated with
Patients receive treatment with a combina­ the use of high‐dose cytarabine and resultant
tion of ATRA and arsenic trioxide (ATO). altered integrity of the gastrointestinal
ATRA induces the differentiation of imma­ mucosa. Sepsis with this organism can be rap­
ture promyelocytes into mature granulocytes, idly fatal. Empiric therapy for fever and neu­
followed by restoration of normal hemat­ tropenia should include coverage for S.
opoiesis. ATRA does not cross the blood– viridans (i.e., vancomycin) and be started
brain barrier and is therefore ineffective in immediately with the first fever. Prolonged
treating CNS involvement with APL. A WBC hospitalization is expected during the course
≥10 × 109/l is associated with a higher risk of of treatment for AML and these patients
relapse, and those with a WBC <10 × 109/l are should be in protective environments.
considered SR. Higher WBC count is also Utilization of protocols to minimize central‐
associated with the microgranular variant line‐associated bloodstream infections is vital.
(M3v), presence of the FLT3‐ITD muta­ Toxicities with ATRA may be significant,
tion, and the bcr3 isoform. Treatment with specifically ATRA induced differentiation
ATRA should be started immediately when syndrome in 2–6% of patients. This syn­
APL is suspected to quickly drive the malig­ drome should be suspected in the patient
nant blasts into terminal differentiation and with fever, respiratory distress, weight gain,
prevent further proliferation. Treatment with edema, and importantly, hyperleukocytosis.
ATRA and anthracyclines has now led to The incidence of ATRA syndrome has
clinical remission rates of over 90% and EFS decreased due to the addition of anthracy­
of 70–80%. ATO works synergistically with cline therapy in induction and can be treated
ATRA and binds the PML moiety of the by temporary discontinuation of ATRA and
oncoprotein PML–RARα resulting in partial ATO with administration of dexamethasone
differentiation and apoptosis in promyelo­ q12 hours until the symptoms have resolved
cytic blasts. Recent studies suggest this com­ for a minimum of 3 days. Additional support­
bination will likely lead to improved long‐term ive care measures may be needed including
outcomes. HR patients receive idarubicin in diuretics, blood products, mechanical venti­
addition to ATRA and ATO. lation, or dialysis. ATRA may cause other
side effects including dryness of the lips and
Complications of therapy mucosa, fever, transaminitis, and headache
Given the intensity of therapy for AML, associated with pseudotumor cerebri. ATO
children can expect to experience a number can lead to prolongation of the QTc interval
of complications that include cytopenias and therefore electrocardiograms (EKGs)
requiring frequent blood product support, must be regularly followed while on therapy.
mucositis, liver toxicity, need for parenteral Coagulopathy at diagnosis is not uncom­
hyperalimentation, and severe, prolonged mon, especially in the child with APL.
marrow suppression. Infection is common Coagulation studies should be done rou­
and should be anticipated. Once neutropenic, tinely. In general, platelet counts should be
these children are at particularly high risk for maintained above 10 × 109/l (>50 × 109/l for
the development of sepsis with Gram‐nega­ the APL patient at diagnosis). The use of
tive organisms in addition to invasive fungal fresh frozen plasma, cryoprecipitate, and
disease. Routine antifungal prophylaxis with platelets are important supportive care meas­
fluconazole or an extended‐spectrum azole ures due to the frequently observed coagu­
(i.e., voriconazole or posaconazole) is rec­ lopathy, hypofibrinogenemia (fibrinogen
Acute Leukemias 205

<100 mg/dl), and thrombocytopenia, respec­ lymph node. You get a CBC with differen­
tively. The total anthracycline dose used in tial, which shows a normal white blood cell
AML protocols is quite high (i.e., approxi­ count, absolute neutrophil count (ANC) of
mately 450 mg/m2 of doxorubicin equiva­ 750 × 106/L, hgb 5.9 g/dl, and platelet count
lents), therefore left ventricular ejection of 23 × 109/l.
fraction must be followed closely during and a. What is the differential diagnosis and
after therapy. Utilization of a cardioprotect­ how will definitive diagnosis be made?
ant (i.e., dexrazoxane) should also be consid­ Lymph nodes that are very firm, matted, or
ered routinely. larger than 1.5–2 cm in diameter are con­
cerning for neoplasm. Lymph nodes may
Relapse be tender with infection or rapid growth.
Prognosis for AML patients in relapse remains Additionally, certain locations such as
poor. Chemotherapy alone results in <10% epitrochlear or supraclavicular, as well as
1‐year disease‐free survival (DFS). For those lack of association with mild or transient
patients that can again obtain MRD remission infectious illness, may indicate a more
after re‐induction chemotherapy, HSCT severe illness such as chronic infection,
improves survival. Re‐induction regimens rheumatologic disease, or malignancy.
include FLAG therapy (G‐CSF, fludarabine, Cytopenias affecting two or more cell lines
cytarabine, with or without daunorubicin), (in this case all three, as the ANC is low)
CPX‐351 (liposomal daunorubicin and cyta­ suggest a bone marrow infiltrative process
rabine), and TVTC (thiotepa, vinorelbine, (such as AL) or decreased production
topotecan, and clofarabine). Additional agents (such as aplastic anemia). Definitive diag­
that may have benefit alone or in combination nosis is made via bone marrow aspiration
include demethylating agents (azacitidine and (BMA; bone marrow biopsy should be
decitabine), histone deacetylase inhibitors done as well if aplastic anemia is a consid­
(vorinostat), and bcl2 inhibitors (venetoclax). eration) unless there are adequate abnor­
Chimeric antigen receptor T‐cell immuno­ mal circulating cells to assess by flow
therapy to target CD33 or CD123 is currently cytometry (typically requires an elevated
being investigated, but has not shown simi­ white cell count with a large percentage of
lar benefit of CAR‐T cell therapy as in ALL, abnormal peripheral blasts).
to date. Traditionally, the diagnosis of ALL is
defined as a blast (malignant cell line) popu­
lation represented by at least 25% of the
Case study for review bone marrow cells (AML is defined by the
presence of ≥20% myeloblasts by BMA),
You are seeing a 12‐year‐old girl who pre­ often seen in association with a markedly
sents to the emergency department for eval­ hypercellular marrow (monotonous cell
uation of “hard bumps” in her neck. She had population) and crowding with little evi­
initially thought they were related to her dence of normal cell production of red cells,
“cold,” since she has cough, tactile fever, and white cells, or platelets. In addition to this
looked pale and tired to her parents. On exam, morphologic assessment of the bone marrow
vital signs are notable for a temperature of in patients with evidence of leukemia, a
39 °C and respiratory rate of 24 breaths sample is sent to assess for specific known
per minute. She has pale conjunctiva, dry cytogenetic anomalies (which may be
cracked lips with mild bleeding, multiple confirmative of a malignant process if the
firm cervical lymph nodes bilaterally 1–3 cm marrow has fewer than 20–25% blasts).
in diameter, and a 0.5‐cm left epitrochlear Immunophenotyping is done to differentiate
206 Chapter 15

AML and ALL, and subsequently a B‐ or T‐ related to cytokine release. However, risk of
cell lineage in ALL. This information in infection is also increased in the setting of a
combination with the cytogenetic results low white blood cell count or neutropenia.
yields important diagnostic and prognostic Patients should always be considered to
information, and is the basis of the initial have infection and have blood cultures
treatment plan. drawn and started on empiric broad‐spec­
b. What additional workup should be trum antibiotics.
done from the emergency department? c. What additional studies should be
Additional laboratory evaluations are helpful done to confirm the diagnosis of AL once
to determine if an oncologic emergency may the patient is admitted to the hospital?
be eminent, such as TLS or a mediastinal As mentioned in the preceding text, the
mass compromising heart function. A com­ definitive diagnostic study is BMA which
plete metabolic panel with electrolytes and includes a morphologic interpretation, as well
renal and hepatic function assessments are as immunophenotyping and cytogenetic
important to assess the degree of cellular studies. FISH for known mutations with prog­
turnover and impact on end‐organ function. nostic and therapeutic significance should be
Particularly important to assess are evi­ sent in conjunction with chromosomal analy­
dence of hyperkalemia, hyperphosphatemia, sis. About 2-5% of patients with AL will have
hypocalcemia, and hyperuricemia in addi­ overt evidence of CNS involvement at diagno­
tion to creatinine. LDH is a marker of cell sis. In ALL, this is defined as an elevated white
turnover and is often markedly elevated in blood cell count (chamber count of five or
the presence of ALL. Transaminases, blood more cells) and presence of blasts on the cyto­
urea nitrogen (BUN), and creatinine are centrifuge slide. Additionally, ALL patients
helpful in supporting the diagnosis and are may have presence of rare blasts in the face of
often elevated at diagnosis. Coagulation stud­ normal chamber count and be classified as
ies should be sent to determine appropriate CNS2. No overt evidence of CNS involvement
intervention if AML is suspected (especially is referred to as CNS1. Patients may also rarely
APL), or if with clinically significant bleeding manifest neurological signs of CNS involve­
with a possible underlying coagulopathy. ment. Additional interpretation of CNS
However, in general the PT/INR, PTT and involvement of a traumatic (bloody) tap (CSF
fibrinogen are likely to be normal. contaminated by many red blood cells sugges­
Any patient with suspected leukemia tive of peripheral blood contamination) is
should have a CXR to evaluate for the required. Classification of the leukemia with
presence of a mediastinal mass. The patient all these components is critical for precise
with a mediastinal mass may be asympto­ diagnosis and determining a therapeutic plan,
matic or present with mild symptoms like with possible eligibility to participate in a clin­
cough, or have evidence of airway com­ ical trial.
promise (hypoxia, positional discomfort BMA reveals 84% lymphoblasts which is
[orthopnea]) or cardiovascular compromise confirmed on flow cytometry by presence of
(poor pulses, hypotension, or superior vena TdT. Additionally, flow is positive for CD2,
cava/mediastinal syndrome). This informa­ CD3, CD4, CD5 and CD7, and negative for
tion is vital prior to any anesthesia due to CD10, CD19 and CD33. CNS studies are
risk of sedation‐associated decompensation negative.
(i.e., decreased cardiac return). d. Does this confirm your suspected
Fever is common at presentation of leu­ diagnosis? What can you tell the family
kemia and may be caused by inflammation generally about treatment options?
Acute Leukemias 207

These bone marrow and immunohisto­ developed off and on low grade fever with­
chemistry results are consistent with T‐ALL out URI symptoms. She also appears more
without evidence of CNS disease. Given her pale and tired to them. All of the following
age above 10 years, she is considered to be should prompt a follow up CBC to rule out
“high risk” and therefore would be treated acute leukemia EXCEPT:
on a protocol for HR T‐ALL. She will receive a. Presence of bruising or petechiae
systemic chemotherapy in addition to CNS b. Notable diffuse cervical lymphade­
prophylaxis with intrathecal chemotherapy. nopathy on exam
Without CNS-directed therapy, the majority c. Fever to 101°C with pharyngeal
of patients would have a CNS relapse as the exudates
CNS (in addition to the testes in males) is a d. Refusal to walk
sanctuary site for leukemia given limited e. Pallor
chemotherapy penetration due to the blood- Explanation: For the general pediatrician,
brain barrier. T‐ALL patients have a higher acute leukemia will be a rare diagnosis and
CNS relapse rate compared to B‐ALL and many symptoms will overlap with viral
many will receive prophylactic cranial radia­ illnesses, even without URI symptoms.
tion as well. Generally, treatment for lymph­ Non‐specific symptoms including fever and
oblastic leukemia is about two and a half fatigue will not help rule in or out leukemia.
years in length, the majority of which is Specific questions that are helpful include a
delivered in an outpatient setting. The initial description of bone pain or refusal to walk
8–10 months however are more rigorous, in a young child due to the bone marrow
require frequent clinic visits and periodic being packed with lymphoblasts. Similarly,
hospitalization for treatment or complica­ signs or symptoms of anemia, thrombocyto­
tions of treatment. Additionally, many sup­ penia or neutropenia can be helpful such as
portive care measures will be in place to with pallor, petechiae or bruising. Focal
anticipate and treat adverse effects of treat­ symptoms related to a viral or bacterial pro­
ment and include transfusion therapy, pain cess may be reassuring as long as symptoms
management, infection prophylaxis and resolve—close follow up is warranted and
treatment, as well as monitoring for specific there should be a low threshold to check a
adverse effects of the chemotherapeutic CBC/diff. The answer is c.
agents (such as cardiac dysfunction from
anthracyclines). Current EFS (event free 2. You end up performing a CBC on the
relapse) is approximately 80–85%. Early above child with findings concerning for
response to treatment (as measured by end acute lymphoblastic leukemia. All of the
of induction bone marrow MRD detection) following are high‐risk features EXCEPT:
is of prognostic significance. Identification a. Age between 1 and 10 years
of cytogenetic risk factors more precisely b. WBC >50 × 109/l
categorize the pre-B-ALL and AML patient c. Disease in the cerebrospinal fluid
but are not as useful in the T-ALL patient. (CNS3)
d. KMT2A rearrangement (11q23)
e. Positive minimal residual disease
Multiple choice questions (MRD) at the end of induction therapy
Explanation: Early clinical findings showed
1. You are seeing a 4‐year‐old female in that infants and those 10 years of age of greater
clinic. She had been well appearing until did more poorly than younger children.
2 weeks ago when the family noted that she Additionally, patients presenting with a high
208 Chapter 15

WBC count also did more poorly in earlier should always be without any potassium
studies. With more sophistication of testing, it though. It is important to check a CXR to
has been found that older children and those rule out an associated mediastinal mass
with a high WBC count are more likely to especially prior to anesthesia for diagnostic
harbor a genetic mutation which makes their bone marrow aspirate and lumbar puncture.
leukemia more difficult to treat, such as The answer is a.
KMT2A rearrangement or Philadelphia chro­
mosome t(9;22). Similarly, these patients are 4. In the above scenario, this 4‐year‐old
more likely to have residual leukemia at the female would be standard risk at the outset
end of induction, the most prognostic high‐ given her age and presenting WBC count.
risk features. CNS disease is another inde­ Per the hematology fellow the blasts look
pendent high‐risk feature. The answer is a. consistent with acute lymphoblastic leuke­
mia (ALL). The family has many questions
3. You are seeing the above patient in the ED for you about the diagnosis and plan of care.
after the primary physician sends the patient All of the following are reasonable state­
in for admission. The CBC is notable for a ments EXCEPT:
WBC of 22 × 10 9/l, hemoglobin is 5.5 g/dl a. Definitive diagnosis will require bone
and platelets are 15 × 109/l. Manual differen­ marrow aspiration
tial reports 58% blasts. All of the following b. Based on what we know now she
are initial considerations EXCEPT: would be standard‐risk ALL but this
a. Initiation of fluids at 2x maintenance, depends on the findings on bone mar­
D5 1/2NS with 10 mEq of KCl/l row aspirate and lumbar puncture
b. Checking a CXR to rule out a medias­ c. If this is standard risk ALL, she will
tinal mass definitely be cured
c. Checking tumor lysis labs to include d. If this is standard risk ALL, the total
potassium, phosphorus, uric acid treatment time is about 2 and a half years
d. Planning for platelets prior to diag­ e. If this is standard risk ALL, the major­
nostic lumbar puncture ity of treatment will be in the clinic rather
e. Planning for PRBCs prior to anesthesia than in the hospital
and to keep hgb >7 g/dl Explanation: When delivering bad news it
Explanation: Multiple supportive care is important to be concise, avoid medical
measures are vital to stabilize a new diagno­ jargon, provide accurate information, be
sis with leukemia. Fever due to leukemia is reassuring and hopeful but without misrep­
often present and these patients should be resenting the truth, and to spend time
started on cefepime for functional neutro­ concentrating on the next steps. In this case
penia. Anemia and thrombocytopenia are it is okay to be hopeful as the majority of
common and transfusion is often indicated, children with standard risk ALL will be
often emergently depending on the level as cured. In general practice it is best to avoid
well as underlying signs and symptoms. giving percentages unless specifically asked.
Elevation in potassium, phosphorus, and/or Definitive diagnosis requires a bone marrow
uric acid can be seen, especially with higher aspirate with immunophenotyping to deter­
WBC counts and should be followed closely, mine if this is ALL or AML. Also a lumbar
especially after the initiation of therapy. puncture is necessary to rule out CNS
Twice maintenance fluids are vital to start disease. Cytogenetics will help determine
immediately to help prevent symptoms of underlying genetic risk. For standard risk
tumor lysis syndrome (TLS); these fluids leukemia the treatment is about 2 and a half
Acute Leukemias 209

years for girls. Due to worse outcomes, boys similar findings and has been rarely reported
may be treated with a longer maintenance in DS. Finally, JMML is a rare condition
though this is decreasingly a standard prac­ found in young children with increased
tice. For standard risk leukemia the large monocytes and a small percentage of
majority of treatment is outside the hospital peripheral blasts and elevation in hemo­
setting. The answer is c. globin F usually due to an underlying
mutation in the RAS pathway or with NF1.
5. You are seeing a 3‐year‐old male with The answer is d.
Down syndrome in the ED due to easy bruis­
ing and some pallor. His CBC is concerning
for a WBC of 1.8 × 109/l, hemoglobin 6.2 g/dl
and platelets of 17 × 109/l. Of note, he had Suggested reading
transient myeloproliferative disorder when
Bhojwani, D., Yang, J.J., and Pui, C.H. (2015).
born with a high WBC count that self‐
Biology of childhood acute lymphoblastic leu­
resolved over several days after birth. The
kemia. Pediatr. Clin. North Am. 62: 47–60.
most likely underlying diagnosis is: Hunger, S.P. and Mullighan, C.G. (2015). Acute
a. ALL lymphoblastic leukemia in children. N. Engl. J.
b. Aplastic anemia Med. 373: 1541–1552.
c. Acute myelomonoblastic leukemia Karol, S.E., Coustan‐Smith, E., Cao, X. et al. (2015).
(AML, M4 based on the FAB classification) Prognostic factors in children with acute mye­
d. Acute megakaryoblastic leukemia loid leukemia and excellent response to remis­
(AML, M7 based on the FAB classification) sion induction therapy. Br. J. Haematol. 168:
e. Juvenile myelomonocytic leukemia 94–101.
Kolb, E.A. and Meschinchi, S. (2015). Acute mye­
(JMML)
loid leukemia in children and adolescents:
Explanation: Children with Down syndrome
Identification of new molecular targets brings
(DS) are at increased risk of leukemia, both promise of new therapies. Hematology Am.
myelogenous and lymphoblastic. Infants Soc. Hematol. Educ. Program 2015: 507–513.
that had transient myeloproliferative disor­ Maloney, K.W. and Gore, L. (2018). Agents in
der (TMD) have about a 25% risk of devel­ development for childhood acute lymphoblas­
oping acute megakaryoblastic leukemia tic leukemia. Paediatr. Drugs 20: 111–120.
(AMKL, AML M7) and should be followed Porter, C.C., Druley, T.E., Erez, A. et al. (2017).
closely with serial CBCs. This risk is due to a Recommendations for surveillance for children
mutation in GATA1 which is unique to chil­ with leukemia‐predisposing conditions. Clin.
dren with DS. Fortunately, those children Cancer Res. 23: e14–e22.
Rubnitz, J.E. (2017). Current management of
with DS who present with AMKL prior to
childhood acute myeloid leukemia. Paediatr.
4 years of age have a very high rate of cure.
Drugs 19: 1–10.
Although ALL is possible in this child, the Vrooman, L.M. and Silverman, L.B. (2016).
history of TMD makes AML much more Treatment of childhood acute lymphoblastic
likely. Similarly, M4 AML is possible but leukemia: Prognostic factors and clinical
much less likely in DS with the history of advances. Curr. Hematol. Malig. Rep. 11:
TMD. Aplastic anemia can present with 385–394.
16 Central Nervous
System Tumors
Central nervous system (CNS) tumors are tumor classification. The version released in
the most common cancer diagnosis in chil- 2016 was the first to incorporate a tumor’s
dren after leukemia, accounting for 20% of all genomic profile into the diagnostic scheme.
pediatric malignancies. The most common This version contains 59 primary diagnoses
location is infratentorial in children up to with several sub‐diagnoses. The current
14 years of age, while supratentorial tumors structure makes it easier for a pathologist to
are more common in adolescents. Spinal cord make a diagnosis in many cases, but can lead
tumors are also more common in adolescents to a delay in diagnosis while waiting for the
than in younger children (9 vs. 3%). results of the genomic profiling. Historically,
The cause of most pediatric CNS tumors microscopic criteria used in grading the
is unknown. The majority of them are spo- aggressiveness of CNS tumors have included
radic, although a small percentage is associ- cellular pleomorphism, mitotic rate, degree
ated with genetic disorders such as of anaplasia, and presence or absence of
neurofibromatosis, tuberous sclerosis, von necrosis. While these remain relevant, the
Hippel–Lindau syndrome, and Li–Fraumeni current classification also identifies certain
syndrome (germline mutation of p53, a sup- mutations such as H3.3K27M in glial
pressor oncogene). Radiation therapy is the tumors as being sufficient to identify a
most frequently identified cause of pediatric tumor as Grade 4 regardless of its histologic
CNS tumors; these occur as second malig- appearance. A growing number of chromo-
nancies in children who have previously somal abnormalities and copy number vari-
received radiation therapy for treatment of ations have also been included in the
leukemia or primary CNS tumors. A variety classification system.
of epidemiologic studies have looked for an
association between cellular telephone use
and an increased risk of brain tumors, pri- Clinical presentation
marily gliomas. The results remain unclear;
if a risk exists, it appears not to emerge until It is often difficult to make the diagnosis of a
greater than 10 years (or >1640 hours) of CNS tumor in a child. The presenting symp-
exposure, making this an unlikely cause of toms of CNS tumors can be quite variable,
pediatric gliomas, even if the risk is real. depending on the location of the tumor, its
The World Health Organization (WHO) rate of growth, and the age of the child.
Classification System for brain tumors is the Additionally, most of the symptoms are
internationally accepted standard for brain nonspecific, and occur more commonly in

Handbook of Pediatric Hematology and Oncology – Children’s Hospital & Research Center Oakland,
Third Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
© 2021 John Wiley & Sons Ltd. Published 2021 by John Wiley & Sons Ltd.
212 Chapter 16

children with a variety of benign conditions. dorsal midbrain. Diencephalic syndrome is a


Prompt diagnosis of a CNS tumor requires rare cause of failure to thrive (FTT) in infants
an appropriate index of suspicion and a good and young children. Characteristic features
understanding of the duration of symptoms include profound emaciation with normal
that is expected with other disorders. caloric intake, absence of subcutaneous fat,
Due to the frequent association of hydro- locomotor hyperactivity, euphoria, and
cephalus with infratentorial tumors, 50% of alertness. It is almost always associated with
children with brain tumors will present with hypothalamic/suprasellar tumors, most
signs of increased intracranial pressure (ICP), commonly optic pathway gliomas.
including lethargy, vomiting, and irritability. Seizures are relatively uncommon in
These symptoms may be less prominent in younger children and occur twice as often in
infants because the cranial sutures remain adolescents (13 vs. 25%). They can be focal or
open and the skull can accommodate the generalized. When focal, they may provide a
increase in ventricular size. For this reason, clue as to the location of the tumor. An often
accurate measurement of the head size overlooked symptom in school‐age children
should be part of every well‐baby examina- is deterioration in school performance and
tion. Headache is a presenting symptom in behavioral changes. It is not uncommon for
70% of children with posterior fossa tumors these children to be referred by a psychologist
and 50% of central tumors. Features that are or psychiatrist after many months of unsuc-
helpful in distinguishing tumor‐related head- cessful therapy for these symptoms.
ache from a primary headache syndrome Symptoms of spinal cord tumors are
include nausea and vomiting (particularly in caused by compression of the spinal cord or
the morning), increasing frequency and/or nerve roots by the mass. The most common
severity over a few weeks, and abnormalities symptom in children with spinal cord
on neurologic examination. Neurologic defi- tumors is back pain. Unlike adults, the com-
cits such as ataxia, visual changes, and hemi- plaint of back pain is very rare in childhood.
paresis occur in about 25% of cases. Weakness “Benign” causes of back pain include trauma,
is sometimes subtle, but parents often relate a medical conditions such as sickle cell dis-
loss of physical function or developmental ease, or involvement in aggressive sports
milestones. Visual changes include diplopia such as football, gymnastics, skateboarding,
caused by cranial nerve dysfunction and loss or motocross bicycling. In the absence of
of visual acuity caused by optic nerve involve- such a history, there should be concern that a
ment or papilledema. tumor might be the cause. Tumor‐related
Two specific clinical syndromes are fre- pain is often described as being worse in the
quently associated with childhood brain supine position or with Valsalva’s maneuver.
tumors. Parinaud’s syndrome consists of Other symptoms associated with spinal cord
paralysis of upward gaze, accommodative tumors include sciatica, weakness, numb-
paresis, nystagmus during attempts at ness, problems with or loss of bowel and/or
upward gaze, eyelid retraction (Collier’s bladder control, and spinal deformity.
sign), and “setting‐sun sign” or conjugate Tumors within the spinal canal are either
downward gaze. It is most commonly associ- intramedullary (within the spinal cord or
ated with pineal region tumors, but it can nerve roots) or extramedullary. Intramedullary
also be caused by obstructive hydrocephalus spinal cord tumors are almost always glial in
from any cause, posterior fossa tumors, or a origin, the most common histologies being
number of infectious or vascular abnormali- astrocytoma, myxopapillary ependymoma,
ties leading to ischemia or damage to the and oligodendroglioma. Extramedullary
Central Nervous System Tumors 213

tumors are either intradural or extradural. brain and spine. It provides greater sensitivity
Extramedullary intradural tumors are most in areas that may be obscured by proximity
commonly neurofibromas, schwannomas, or to bone, such as the temporal lobe and
more rarely meningiomas. Extradural tumors posterior fossa. It also provides multiplanar
are usually not CNS tumors at all, but meta- images. Contrast MRI allows for identifica-
static tumors from other locations such as tion of tumor within areas of surrounding
neuroblastoma, lymphoma, or Ewing sar- edema or hemorrhage, detects focal areas of
coma. Tumors within the vertebral bodies blood–brain barrier (BBB) breakdown, and
such as primary bone tumors, tumors meta- improves delineation of cysts from solid
static to bone, and histiocytosis can also extend tumor.
into the spinal canal and cause the abovemen- Extensions of MRI technology include
tioned symptoms. MR spectroscopy, tractography, and func-
tional MRI scanning. MR spectroscopy
allows the determination of the chemical
Diagnostic evaluation makeup of tissue within a predefined region
of interest. Different types of tumors have
Imaging studies characteristic spectroscopic signatures,
The first step in evaluating a child with a allowing the identification of the most likely
suspected CNS tumor is neuroimaging. A tumor type before surgery takes place.
variety of imaging techniques can be useful Tractography is a software technique that
in this assessment. allows identification of the location of white
matter tracts and their associated nuclei.
Computerized tomography (CT) This has become very useful as a preopera-
CT was the first imaging modality to revolu- tive planning tool that allows the neurosur-
tionize the diagnosis of CNS tumors both in geon to avoid disruption of eloquent nuclei
children and adults. It is often still the first and fiber tracts during surgery. Functional
imaging study obtained when evaluating MRI is another technique that precisely
patients suspected of having a tumor. identifies specific motor regions, speech and
However, magnetic resonance imaging (MRI) language centers, and other eloquent areas
has largely replaced it as the definitive neuro- for preoperative planning purposes.
imaging study. Advantages of CT include
widespread availability, rapid acquisition Positron emission tomography
time, superior resolution of bone and vascular (PET)
structures including lesions of the skull base, PET imaging is a useful modality for evalu-
and utility in patients with contraindications ating a variety of tumors. It detects the rela-
to MRI (i.e., ferrous metal implants). It also tively higher rate of metabolism present in
more clearly identifies intratumoral calcifica- many neoplasms by measuring the rate of
tion. Its limitations include inferior resolution metabolism of one of a growing number of
of the brain itself, particularly within the pos- radioactively labeled compounds, most
terior fossa, and significant radiation expo- commonly 2‐deoxy‐2[18F]fluoro‐d‐glucose
sure, especially with regular monitoring. (FDG). Similar to many high‐grade tumors
elsewhere in the body, malignant brain
MRI tumors also show high FDG uptake because
An MRI, with and without gadolinium of increased glucose transport and glycoly-
enhancement, is the single best study for sis. However, images are more difficult to
evaluating the majority of tumors within the interpret because of the high and regionally
214 Chapter 16

variable glucose metabolism in normal Other studies


brain gray matter. Its utility in young chil- Lumbar puncture (LP)
dren is limited due to the need for a 30–60 Certain primary CNS tumors, such as
minute period of limited mental activity medulloblastoma, primitive neuroectoder-
between injection and scanning to optimize mal tumor (PNET) (now classified as CNS
the distinction between normal and abnor- embryonal tumor, not otherwise specified
mal structures. Other compounds have been (NOS) in the 2016 WHO CNS tumor clas-
developed in order to overcome some of sification), ependymoma, malignant germ
these limitations, particularly the large neu- cell tumors (GCTs), and, to a lesser extent,
tral amino acid (LNAA) tracers 11C‐methio- malignant gliomas, have a propensity to
nine (MET), 18F‐fluoroethyltyrosine (FET), metastasize throughout the subarachnoid
and 18F‐fluorodopa (FDOPA). MET‐PET space. As with other malignant tumors,
has been found to demonstrate presence of the presence of metastatic disease alters
tumor in non‐contrast enhancing regions of both treatment and prognosis. For this
pediatric high‐grade gliomas, and to predict reason, LP is a routine part of the diagnos-
areas of relapse. tic workup of these patients. Spinal fluid is
Traditionally, PET images have been co‐ collected and sent for cell count and differ-
registered with CT images to provide ana- ential, glucose, protein, and cytological
tomic detail. More recently, technology has examination to look for malignant cells.
been developed to allow co‐registration of Children with malignant GCTs should also
PET images with MRI scan images. This have measurements of α‐fetoprotein (AFP)
provides better definition and decreases the and the beta subunit of human chorionic
amount of radiation exposure associated gonadotropin (β‐HCG) performed. These
with the scan. Use of these compounds in markers, while not diagnostic of metastatic
the diagnosis of adult brain tumors has disease, can, if present, be used to measure
become well‐established, but experience in response to therapy. The diagnostic LP
children is still largely lacking. should be delayed for at least 10‐14 days
following tumor resection to allow any non-
Single‐photon emission viable tumor cells present in the cerebrospinal
computed tomography (SPECT) fluid (CSF) as a result of the surgery itself
SPECT is a three‐dimensional imaging tech- to be cleared.
nique introduced in the early 1980s for the
investigation of regional cerebral blood flow
that often correlates with tumor metabolic Bone marrow aspiration
activity. In this technique, radioactive thal- In the past, it was recommended that
lium and radiolabeled tyrosine have been patients with medulloblastoma undergo a
used to localize brain tumors as well as dis- bone marrow aspiration at the time of diag-
tinguish viable tumor from radiation necro- nosis to rule out metastatic disease prior to
sis. It is less expensive than PET scanning, starting therapy. However, more recent
but the resolution is less and the 20–30 minute studies have indicated that fewer than 3% of
acquisition times needed for high‐quality patients diagnosed with medulloblastoma
pictures again limit utility in young children. have extraneural metastases at diagnosis,
It has also typically been co‐registered with with fewer than 25% of these having disease
CT images to provide anatomic detail. As of in the marrow. Therefore, recent clinical tri-
2018, it was not yet possible to co‐register als have no longer recommended routine
SPECT images with MRI. examination of the bone marrow, but have
Central Nervous System Tumors 215

instead recommended this only for patients (iii) functioning ­pituitary adenomas, as the
with unexplained cytopenias. Extraneural role of surgery in children is limited, except
metastases are almost unheard of with other when they are large and causing mass effect
types of pediatric CNS tumors at diagnosis, or threatening vision through compression
making bone marrow aspiration at diagno- of the optic chiasm.
sis unnecessary. The goals of neurosurgical intervention
are to achieve the maximum tumor removal
Bone scan possible while minimizing morbidity and
While 90% of extraneural metastases in mortality and to obtain tissue for a histologic
patients with medulloblastoma are to bone, diagnosis. In the case of medulloblastoma
this remains an unlikely presenting finding, and other CNS embryonal tumors, epend-
since, as mentioned in the earlier text, fewer ymoma, and high‐grade gliomas, extent of
than 3% of patients have extraneural metas- surgical resection is one of the major predic-
tases at diagnosis. Therefore, bone scan tors of outcome. A number of surgical
should be reserved for patients who present advances have increased the likelihood of
with the underlying diagnosis of medullo- safely achieving complete tumor resections,
blastoma that is accompanied by bone pain. including use of an operating microscope
with neuronavigation; intraoperative ultra-
sound, CT and MRI; intraoperative neuro-
Treatment physiological monitoring; and use of dyes
that selectively stain tumor tissue. The latter
Children with CNS tumors are typically is not yet approved for use in children, but
treated with some combination of neurosur- clinical trials are ongoing.
gery, radiation therapy, and chemotherapy. Children with increased ICP caused by
The specific treatment chosen is dependent obstructive hydrocephalus will likely have a
on the histology, location, resectability, and CSF diversion procedure performed as well,
prognosis of the patient’s tumor. either before their definitive surgery if they
are unstable or at the time of resection. An
Neurosurgery external ventricular drain (EVD) will often
Initial neurosurgical intervention is indi- be placed initially and will be clamped at
cated in the majority of patients. Exceptions some point after surgery to determine if nor-
include: (i) diffuse intrinsic pontine glioma mal CSF flow has been restored. Children
(DIPG), due to the infiltrative nature of the who redevelop symptoms of increased ICP
tumor throughout the pons (however, tumor will likely require placement of a permanent
biopsy has become a more standard part of shunt. Most often this is a ventriculoperito-
treatment to obtain tissue for molecular neal shunt (VPS), but shunts to other sites
analysis and this innovation has led to more may be done in specific circumstances.
specific tumor‐directed therapies that will
hopefully improve outcomes for these typi- Radiation therapy
cally fatal tumors); (ii) tumors that are radio- Radiation therapy has played a pivotal role in
graphically consistent with GCTs and present the management of children with malignant
with elevation of both AFP and β‐HCG, con- brain tumors for decades. It has been used to
firming the diagnosis of nongerminomatous destroy remaining tumor in cases of incom-
germ cell tumor (NGGCT) without a biopsy plete surgical resection, and to treat micro-
for which aggressive surgery at diagnosis has scopic residual disease and metastatic disease
not been shown to improve survival; and in children who have malignant tumors with
216 Chapter 16

a high likelihood of tumor regrowth. The regarding a possible association with PBT
dose of radiation used is dependent on the and the development of intracranial vascu-
type of tumor and the location and volume lopathy. Research to further evaluate this
to be irradiated. potential risk is ongoing.
Unfortunately, the use of radiation ther- Despite these innovations, the use of
apy is associated with significant adverse radiation therapy continues to carry the risk
late effects, particularly in young children. of significant morbidity. Work continues to
Those children receiving higher doses of develop novel strategies that will allow the
radiation before 6 years of age, especially to avoidance of radiation therapy completely
the entire craniospinal axis (craniospinal in children with CNS tumors, or at least to
irradiation, CSI), can be expected to suffer delay it in very young children.
significant impairment of intellectual and
physical development as a result of this ther- Chemotherapy
apy. Another concern is the development of The utility of chemotherapy in the treatment
secondary malignancies in the radiation of CNS tumors was recognized more slowly
field. Children receiving prophylactic crani- than in other tumors. It was initially felt that
ospinal radiation for leukemia have been the BBB would prevent the delivery of effec-
shown to have a >20‐fold increase in the risk tive concentrations of chemotherapeutic
of secondary CNS tumors; over 80% of these agents to the tumor. However, it became
tumors occurred in children who were radi- apparent that many drugs do adequately
ated before 5 years of age. Children with cross the BBB, or that this barrier is abnor-
standard risk medulloblastoma who survive mally permeable in many malignant brain
5 years following conventional treatment tumors. Other factors, such as tumor hetero-
that includes CSI are more likely to die from geneity, cell kinetics, drug distribution, and
a radiation‐induced cancer than from a drug excretion may also have a significant
recurrence of their primary tumor. impact on effectiveness of chemotherapy.
A variety of innovations in radiation Lower‐grade tumors are characterized by a
therapy delivery have been developed to low mitotic index and relatively slow growth
minimize the side effects of radiation. These rate; it was long felt that these tumors would
include 3D conformal planning techniques be less sensitive to chemotherapy, while
and intensity modulated radiation therapy more malignant tumors would be more sen-
(IMRT), which minimize the dose of radia- sitive. However, several studies over the last
tion delivered to adjacent normal tissue. three decades have demonstrated that many
Another innovation is proton beam therapy low‐grade tumors are sensitive to chemo-
(PBT), which takes advantage of the shorter therapy. Commonly used drugs for the treat-
decay path of high‐energy protons com- ment of brain tumors include vincristine,
pared to gamma rays (photons). This mini- methotrexate, temozolomide, procarbazine,
mizes the exit dose delivered beyond the site lomustine (CCNU), cisplatin, carboplatin,
of the tumor. These innovations have proven cyclophosphamide, and etoposide.
very valuable in decreasing the radiation The genomic alterations that have been
exposure to heart, lung, bowel, and repro- identified in a growing number of CNS
ductive organs in children receiving total tumors have led not only to the modifica-
spine radiation. PBT has had limited availa- tion of tumor classification mentioned ear-
bility until recently due to the significant lier, but to a major evolution in the treatment
cost of building PBT centers. As it has of these tumors in both children and adults.
become more available, a concern has arisen There is a growing appreciation that tumors
Central Nervous System Tumors 217

of many different histologies can share iden- PTCH1 or SUFU. Infants harboring ­germline
tical genetic mutations (BRAF V660E in PTCH1 mutations present with Gorlin syn-
melanoma and a variety of CNS tumors), drome (nevoid basal‐cell carcinoma syn-
and that the same drugs can be equally drome). Older children have mutually
effective in treating any of these tumors. In exclusive somatic mutations in PTCH1 or
the case of CNS tumors, a major considera- germline and/or‐somatic TP53 mutations,
tion is whether the drug is able to cross the the latter of which frequently co‐occur with
BBB, especially in lower‐grade tumors amplifications of GLI2 and MYCN. TP53
where the BBB is more often intact. A grow- mutations are present in approximately 30%
ing number of these agents have shown of childhood SHH, are frequently germline,
promise and are discussed in more detail in and confer a poor prognosis. Outcomes of
the following text. SHH tumors are age‐specific. Infants have an
excellent outcome, even with chemotherapy‐
only regimens. Conversely, children with
Specific tumor types TP53‐mutant SHH tumors have a dismal
prognosis compared with that of TP53‐
Medulloblastoma wildtype tumors. Group 3 tumors are char-
Medulloblastoma is the most common CNS acterized transcriptionally by the activation
tumor in children, accounting for up to 20% of the GABAergic and photoreceptor path-
of all childhood brain tumors. It has a pro- ways. Group 3 tumors occur in younger chil-
pensity to metastasize early; in up to 35% of dren, 40–50% are metastatic at diagnosis,
cases, seeding of the subarachnoid space is have short prediagnostic intervals, and a
present. Extraneural spread is unusual at male preponderance. The most common
diagnosis but may develop later if tumor cytogenetic aberration is amplification of
recurrence occurs. MYC (10–20% of tumors). Isochromosome
Until recently, medulloblastomas had 17q (i17q) is seen in 40% of Group 3 tumors.
been considered as a single tumor entity. Patients with Group 3 tumors have a poor
However, years of highly sophisticated outcome overall, particularly nonirradiated
genomic profiling of many of these tumors infants, but likely benefit from myeloablative
led in 2010 to a consensus classification sys- treatment regimens. Group 4 tumors are
tem that divided medulloblastoma into four characterized transcriptionally by overrepre-
main molecular subgroups (wingless [WNT], sentation of neuronal and glutaminergic
sonic hedgehog [SHH], Group 3, and Group pathways. Group 4 tumors are the predomi-
4). Figure 16.1 compares the features of these nant subgroup in children 3–16 years of age,
subgroups. WNT tumors have activated have long prediagnostic intervals, and 30
WNT pathway signaling. They rarely metas- and 40% are metastatic at diagnosis. The
tasize, have a longer prediagnostic interval, most common cytogenetic aberration is
occur in older patients, and have a balanced i17q, seen in almost 80% of tumors, with less
sex ratio. SHH tumors are characterized by frequent aberrations in 8p, 7q, 11p, and 18q.
activation of the SHH pathway. SHH tumors Group 4 tumors have an intermediate prog-
are almost exclusively located within the cer- nosis, although in some series, patients with
ebellum rather than in the fourth ventricle, nonmetastatic Group 4 tumors can have an
consistent with their cells of origin (granule excellent prognosis with >90% survival. The
cerebellar progenitors). molecular classification of medulloblastoma
Infants with SHH tumors frequently har- continues to evolve. More recent publica-
bor germline or somatic mutations in tions have suggested increasing the number
218 Chapter 16

Feature WNT SHH Group 3 Group 4

Age group Children/adults Infants/adults Infants/children Children/adults

Metastases at < 5% 20% 40%–50% 35%–40%


diagnosis
Sex ratio 1:1 1:1 2:1 2:1

Somatic CTNNB1 (90%) PTCH1 (25%) SMARCA4 (10%) KDM6A (10%)


nucleotide SMARCA4 (25%) SUFU (10%, CTDNEP1 (5%) MLL3 (5%)
variants DDX3X (50%) infants) MLL2 (4%)
TP53 (12%) SMO (15%, adults)
TERT (20%, adults)
IDH1 (< 5%, adults)
TP53 (15%,
children)
MLL2 (12%)
Germline variants APC (< 5%) PTCH1 (25%
infants,
10% older)
SUFU (20% infants)
TP53 (8% older)
Focal copy MYCN (8%) MYC (15%) SNCAIP (10%)
number GLI2 (5%) OTX2 (7%), MYCN (6%)
aberrations GFI1/1b activation CDK6 (5%)
(25%) GFI1/1b activation
(5%)
Broad copy Monosomy 6 9q loss (35%) i17q loss (22%) i17q loss (70%)
number (85%) 10q loss (22%) 8 loss (29%) 8p loss (49%)
aberrations 17p loss (18%) 10q loss (45%) 11p loss (28%)
11 loss (30%) X loss (80% of
16q loss (48%) females)
1q gain (23%) 7q gain (40%)
7 gain (25%) 18q gain (20%)
18 gain (20%)
Pattern of relapse Local and Predominantly Metastatic Metastatic
metastatic local
High risk TP53 mutation Infants Metastases
MYCN amp Metastases
Low risk Age < 16 years Infants Whole chr 11 loss

Figure 16.1 Molecular subgroups of medulloblastoma. Amp, amplification; Chr, chromosome; i17q,
isochromosome17q; SHH, sonic hedgehog; WNT, wingless.

of medulloblastoma subclasses to as many as Patients with <1.5 cm3 of residual tumor


12. However, it remains unclear how this will after surgery and no evidence of metastatic
improve our understanding of the disease disease are defined as standard risk. Patients
and, more important, how it will lead to with metastatic disease or a larger postop-
more rational treatment strategies. erative residual tumor volume have been
The initial mode of therapy for all sub- considered high‐risk. Once the diagnosis is
types is surgery, and extent of surgical resec- confirmed, patients should be staged with a
tion directly affects prognosis and treatment. MRI of the spine and LP. As mentioned,
Central Nervous System Tumors 219

additional studies looking for extraneural toma. In 1995, the second Head Start study
metastases are no longer considered neces- added high‐dose methotrexate to the existing
sary unless the patient is symptomatic. chemotherapy; results from this modification
Medulloblastomas are very sensitive to were very encouraging. Patients with stand-
radiation therapy, and radiation has tradi- ard‐risk medulloblastoma had 5‐year event‐
tionally been considered the mainstay of free survival (EFS) rates of 50% and overall
postsurgical therapy. Due to the high likeli- survival rates of 75%. Children who relapsed
hood of metastases throughout the CNS, it following chemotherapy alone were effec-
has been necessary to deliver radiation to the tively salvaged with reoperation and radia-
entire brain and spine (i.e., CSI), with a tion therapy in half of the cases. This strategy
higher (boost) dose delivered to the region of led to over 70% of children with standard risk
their tumor. As the long‐term consequences disease treated on Head Start II being cured
of radiation have become more apparent, without radiation therapy. COG has investi-
especially for young children, strategies to gated a similar strategy for children <3 years
delay or avoid radiation in young children of age with high‐risk medulloblastoma and
have become more widely used, and attempts supratentorial embryonal tumors. The results
have been made to decrease the doses of of this trial have not yet been published.
radiation given to older children. The outlook for children >3 years of age
The value of chemotherapy both in with standard risk medulloblastoma is quite
improving survival among patients with prim- good; 5‐year EFS with current multimodal-
itive neuroectodermal tumors (PNETs) and in ity therapy is >80%. Children with high‐risk
allowing a reduction in the dose of radiation medulloblastoma and pineoblastoma do less
therapy required for cure has become increas- well; the most recently published 5‐year EFS
ingly evident over the past three decades. For rates are 50–60%.
the past several years a combination of cispl-
atin, vincristine, CCNU, and cyclophospha- Gliomas
mide has been used following completion of Gliomas account for just over 50% of all CNS
radiation therapy with good effect. Vincristine tumors in childhood. As the name implies,
is also used during radiation therapy, and the they arise from glial cells, which surround
Children’s Oncology Group (COG) is evaluat- neurons and provide support for and insula-
ing whether the addition of carboplatin dur- tion between them. Types of glial cells
ing radiation therapy will improve outcomes include oligodendrocytes, astrocytes, epend-
further in children with high‐risk disease. ymal cells, Schwann cells, microglia, and sat-
Due to the devastating effects that CSI can ellite cells. Gliomas can occur anywhere in
have in children less than 6 years of age at the the CNS, although certain subtypes have a
time of treatment, an alternative strategy was propensity to occur in specific locations.
developed in the early 1990s that substituted Gliomas are classified histologically into
very intensive chemotherapy followed by Grades 1–4 based on cellular pleomor-
consolidative myeloablative chemotherapy phism, cell density, mitotic index, and
and hematopoietic stem cell rescue without necrosis. Grades 1 and 2 tumors are defined
radiation therapy for these young children. as low‐grade (LGG), while Grades 3 and 4
Known as the Head Start regimen, this strat- gliomas are defined as high‐grade (HGG)
egy proved fairly effective for children with and are clearly malignant. The 2016 revision
standard risk, but results were suboptimal for of the WHO Classification of Tumors of the
patients with high‐risk medulloblastoma or CNS dramatically changed the classification
supratentorial PNET, including pineoblas- of glial tumors through the use of integrated
220 Chapter 16

phenotypic and genotypic parameters for tumors metastasize much less frequently
tumor classification. This has added a level than PNETs, patients do not require CSI but
of objectivity that had been missing from they do require focal radiation doses of
some aspects of the diagnostic process in the 50–60 Gy to their tumors to achieve local
past. This is particularly true for glial control.
tumors; in the event that there is discord- The value of chemotherapy in the man-
ance between the histologic appearance and agement of children with low‐grade gliomas
the genomic profile, the diagnosis is based has become increasingly apparent over the
on the genomic characteristics of the tumor. past 30 years. This is especially true for chil-
A midline glioma may have the histologic dren with neurofibromatosis‐associated
appearance of a juvenile pilocytic astrocy- LGG, as the frequency of secondary malig-
toma (JPA, a Grade 1 tumor), but if profiling nant tumors in these children after treat-
reveals an H3.3K27M mutation, the final ment with radiation therapy is alarmingly
diagnosis is “diffuse midline glioma, high. The goal of therapy in this situation is
H3.3K27M‐mutated, WHO Grade 4.” different than with other tumors.
Surgical resection is a key component of Chemotherapy rarely leads to total disap-
therapy for most gliomas that are amenable pearance of the tumor, but rather produces a
to this approach. This includes cerebellar state of stable disease that is hopefully main-
gliomas and supratentorial gliomas that are tained after therapy discontinuation. In this
not centrally located or within the optic regard, management of incompletely
pathway. Gliomas occurring in the tectal resected LGG is similar to managing a
region of the midbrain are an exception to chronic illness like asthma, with periods of
this rule; these are typically very indolent stability, occasional flares of disease, and a
tumors that often require no intervention. hope that the child will eventually “grow
LGG that are completely resected do not out” of their condition.
require adjuvant therapy and are simply As more of these low‐grade tumors have
observed. This is most frequently seen with undergone molecular profiling, it has
JPAs, even if low‐grade, diffuse gliomas are become apparent that mutations within the
infiltrative in nature and can rarely be com- mitogen‐activated protein kinase (MAPK)
pletely resected. If JPAs recur, re‐resection is pathway are a hallmark of JPAs. The most
generally curative. Surgery should be used commonly mutated gene in low‐grade glio-
judiciously, however. Many LGG can be mas is the BRAF oncogene, one of three
controlled successfully with chemotherapy, RAF kinases which act as downstream effec-
so surgery that leaves the patient with sig- tors of growth factor signaling leading to cell
nificant postoperative morbidity should be cycle progression, proliferation, and sur-
avoided. HGG always require adjuvant ther- vival. Over 70% of JPAs are characterized by
apy, although at this time the ability to cure a fusion between BRAF and KIAA1549 that
patients with these tumors remains poor. leads to constitutive activation of BRAF.
Radiation therapy was a key part of the Another common mutation of BRAF is the
treatment of all gliomas for many years, but point mutation BRAF V600E. This muta-
it is used very rarely now to treat children tion is observed in a variety of cancers
with LGG due to the frequency of long‐term including melanoma, papillary thyroid can-
consequences and the realization that these cer, and colorectal cancer. This mutation is
tumors can be managed successfully with also seen in a variety of CNS tumors, includ-
chemotherapy. It remains a component of ing two‐thirds of pleomorphic xanthoastro-
therapy for HGG, however. Since these cytomas (PXAs), 20% of gangliogliomas,
Central Nervous System Tumors 221

and 10% of extracerebellar JPAs. Tumors ependymomas. Grade 4 ependymoma does


possessing the BRAF V600E mutation can not exist.
be successfully treated with BRAF inhibitors Molecular profiling of ependymomas
including vemurafenib and dabrafenib. has revealed that these tumors also have
Tumors with the KIAA1549:BRAF fusion subgroups with unique molecular charac-
have been shown to actually grow more teristics and prognoses. These are outlined
quickly when treated with such agents, but in Figure 16.2. The initial separation is
they do respond to MEK inhibitors such as based on anatomic location and is divided
trametinib and selumetinib; the latter had into supratentorial, posterior fossa, and spi-
just received FDA approval as of the time of nal locations. Within each location there
publication. have been three subgroups defined based on
Gliomas arising in the brain stem molecular characteristics that align well
deserve special mention. These comprise with outcome data. Each subgroup has a
about 15% of brain tumors in children. specific age group or groups where they
About 70% of these arise in the pons, are occur commonly. Prognosis is also associ-
diffusely infiltrative, and are termed DIPG. ated with the subgroup of the tumor. Grade
These are among the deadliest of all child- 1 tumors tend to require surgery alone for
hood brain tumors; median survival of chil- cure. This is even true in the spine due to the
dren with this diagnosis is 8–9 months, and well‐circumscribed, noninfiltrative nature
90% of affected children are dead within of these tumors that allows gross total resec-
2 years of diagnosis regardless of the treat- tion to be accomplished in the majority of
ment provided. Radiation therapy has been cases. As with medulloblastoma, the identi-
estimated to prolong survival for a median fication of these subgroups has not yet
of 3 months. Traditionally a course of radia- resulted in the development of molecularly
tion has taken 6 weeks to deliver, which has based therapies. In fact, the role of chemo-
limited the benefit of this treatment on therapy in the management of ependymoma
improving quality of life, especially for remains controversial. A recent COG trial
children requiring anesthesia for their radi- that prospectively randomized patients to
ation. More recently, hypofractionated regi- receive or not receive chemotherapy follow-
mens that take only 2–3 weeks to deliver ing surgery and radiation has completed
have made this a more attractive option. patient accrual, but the results have not yet
Many trials have been conducted over the been published.
past 25 years trying to improve the survival The extent of surgical resection is the
of children with this tumor, all with disap- most important prognostic factor in chil-
pointing results. dren with ependymoma. The 5‐year EFS for
Although ependymomas are tradition- patients having a complete resection is 80%,
ally classified with other glial tumors, their versus 40% for those patients having a near‐
behavior and management are quite differ- total or subtotal resection. Unfortunately,
ent. They represent about 5–10% of child- ependymomas are often difficult to com-
hood primary brain tumors. About 60% of pletely resect, especially in the posterior
intracranial ependymomas occur in the fossa due to their predilection to surround
posterior fossa, and 40% are supratentorial. cranial nerves and other vital structures. It
Grade 1 ependymomas are rare and almost appears that while adjuvant chemotherapy
never occur in the brain, although Grade 1 has not been demonstrated to improve sur-
(myxopapillary) ependymomas of the vival in patients with ependymoma, it does
spine comprise about 10% of childhood improve outcome when given to patients
222 Chapter 16

WHO Age
Region Subtype Prognosis
Grade Group
ST-SE I
Subependymoma
Supratentorial Excellent
Balanced
Genome
ST-EPN-YAP1 II / III
(Anaplastic)
Supratentorial Good
Ependymoma
YAP1-Fusion
ST-EPN-RELA II / III
(Anaplastic)
Ependymoma
Supratentorial Chromothripsis; Poor
RELA-fusion

PF-SE I
Posterior Subependymoma
Excellent
Fossa Balanced
Genome
PF-EPN-A II / III
(Anaplastic)
Posterior
Ependymoma Poor
Fossa
Balanced
Genome
PF-EPN-B II / III
(Anaplastic)
Ependymoma
Posterior
Chromosomal Fair
Fossa
Instability

SP-SE I
Spine Subependymoma Excellent
6q deletion

SP-MPE I
Myxopapillary
Spine Ependymoma Good
Chromosomal
Instability
SP-EPN II / III
(Anaplastic)
Spine Good
Ependymoma
NF2 mutation

Figure 16.2 Molecular subgroups of ependymoma.

before second look surgery to improve the therapy for this tumor; the doses required
likelihood that a complete resection can be are similar to those needed to achieve local
achieved for patients unable to be com- control of other glial tumors. Ependymomas
pletely resected at diagnosis. Radiation ther- are more likely to metastasize within the
apy remains an important component of CNS than other gliomas, especially if they
Central Nervous System Tumors 223

originate in the posterior fossa or are Grade have demonstrated that patients can be suc-
3. Assessment of the spinal fluid postopera- cessfully treated with aggressive multiagent
tively is important, although negative spinal chemotherapy even without radical surgery.
fluid cytology does not reliably identify The question of whether patients can be
patients less likely to recur away from their cured without radiation therapy under any
primary tumor site. circumstances remains unanswered.

Atypical teratoid/rhabdoid Germ cell tumors


Tumor (ATRT) CNS GCTs are relatively uncommon. Their
ATRTs are the most common malignant management shares many similarities with
CNS tumors in children ≤1 year of age and GCTs occurring outside the CNS. They are
represent approximately 1–2% of all pediat- discussed in Chapter 21.
ric brain tumors. In the past, they were fre-
quently confused with medulloblastoma In summary, although brain tumors occur
when they occurred in the posterior fossa. relatively commonly in children, the varia-
Once it was appreciated that these tumors tions in histologic appearance, presentation,
were characterized by mutations in genes for treatment, and prognosis make their manage-
components of the chromatin remodeling ment quite challenging. The best outcomes
complex SWItch/sucrose nonfermentable for these children occur when they are treated
(SWI/SNF), most commonly in SMARCB1, at a center with a multidisciplinary neuro‐
it became possible to identify them fairly oncology program, and every effort should be
easily with immunohistochemistry through made to ensure that this happens.
the loss of the intracellular protein INI1.
Rhabdoid tumors can arise from many other
areas within the body, including peripheral Case studies for review
nerve roots, kidneys, head and neck, para-
vertebral muscles, liver, mediastinum, retro- 1. A 9‐year-old right‐handed male presents
peritoneum, bladder, pelvis, heart, scrotum, to the ED with his parents, who report that
and subcutis. They may arise synchronously over the past 2 months the patient has been
in more than one area, especially in patients experiencing worsening weakness on his
harboring a germline SMARCB1 mutation. right side. Initially they noted that he
However, in most of these instances, one seemed to be dragging his right foot when
location is the CNS. walking. However, he subsequently began
Recent work has demonstrated that falling more frequently, and for the past day
rather than being a single entity, ATRT in has been unable to walk. His parents also
fact is comprised of at least three distinct note that he has been having increasing dif-
genomic and epigenomic subtypes, identi- ficulty with writing, and his teachers have
fied as types 1, 2A, and 2B. been complaining that they are having dif-
The survival rate for children with ATRT ficulty reading what he has written. His par-
has traditionally been poor, but is improving ents have also noted that his right eye
due to the development of treatment proto- appears “droopy.”
cols specific to this tumor. However, a stand- a. What elements of the history are con-
ard of care had yet to be defined. Previous cerning, and what additional informa-
reports suggested that complete surgical tion should you seek?
resection greatly increases the likelihood of The patient has a right hemiparesis that has
survival. However, more recent publications been present for several weeks. The onset
224 Chapter 16

was insidious, and the deficit has been is very common in conditions affecting the
becoming progressively worse. The weak- pons but is typically absent with lesions
ness involves both cranial and peripheral higher in the brainstem. The third cranial
nerves on the same side of the body, sug- (oculomotor) nerve nucleus resides in the
gesting a centrally located (brainstem) midbrain. Injury to the oculomotor nerve
lesion. These symptoms could be caused by results in paralysis of the intraocular and
an infectious or inflammatory lesion, or by a extraocular muscles, as well as the levator
tumor. palpebrae muscle, and leads to downward
Additional information to be elicited and lateral deviation of the eye. Facial sen-
includes presence of fever, chills, malaise, or sory loss may be present as well. Midbrain
other signs of infection. Inquire about any lesions lead to contralateral weakness, as the
prior history of similar symptoms, as condi- decussation of long tract fibers occurs at the
tions like multiple sclerosis are character- level of the medulla. In addition to weak-
ized by symptomatic episodes separated in ness, these lesions cause spasticity, hyperre-
time and space. Be sure to inquire about flexia, and typically ataxia as well.
sensory loss, as this is often an early Based on the available information, a
complaint. neoplastic process seems more likely than
If the symptoms are caused by a midline an infectious or inflammatory one. However,
tumor, these can also lead to obstruction of it is still useful to obtain a CBC with differ-
CSF pathways. Inquire about any recent ential, an erythrocyte sedimentation rate
sleepiness, irritability, headache, nausea, or (ESR), and a C‐reactive protein (CRP) to
vomiting. Headache due to increased ICP is look for evidence of inflammation. Multiple
often worse at night or in the early morning, sclerosis is still on the differential diagnosis,
and may be worsened by coughing, bending but the only laboratory test that is likely to
over, or straining in the bathroom. help in making that diagnosis is an analysis
The parents report that the patient has of the CSF. Given the possibility of a tumor
never had any other neurological symptoms. causing these symptoms, a lumbar puncture
They also deny any recent history of fever, should be deferred until after imaging is
chills, or other signs of illness. They deny obtained.
any recent headaches, nausea, vomiting, or The most useful test in this situation is
abnormal sleep patterns or increased fatigue. an MRI scan of the brain with and with-
b. Given what you have learned thus far, out contrast. A properly done scan will
what items do you want to pay particular include T1, T2, and fluid‐attenuated
attention to when doing your physical inversion recovery (FLAIR) sequences,
examination? diffusion‐weighted images, and postcon-
c. Are there any diagnostic tests you trast T1‐weighted images in axial, sagittal,
would like to obtain that could help you and coronal planes.
narrow the differential diagnosis? A CBC, ESR, and CRP are done and are
A careful assessment of the vital signs is normal. MRI is performed and reveals a
important. Although there are no clinical 1.5 × 1 × 1.7 cm sharply defined mass in the L
symptoms suggestive of increased ICP, the cerebral peduncle that is isointense on T1,
presence of hypertension and bradycardia hyperintense on T2 and FLAIR, has facilitated
could suggest this. Careful cranial nerve diffusion on diffusion‐weighted imaging, and
examination can be very useful in localizing has a nodular area of intense enhancement
a brainstem tumor. Sixth nerve involvement within the mass.
Central Nervous System Tumors 225

d. What does the MRI tell you about the The patient should also be carefully
patient’s diagnosis? and prognosis? examined for stigmata of neurofibromato-
e. What would be the next steps in the sis type 1 (NF1). Patients with NF1 are at
patient’s management? increased risk of developing low‐grade glial
Tumors within the cerebral peduncle are tumors. Many of these are in the optic path-
almost always glial tumors. They are con- way, but they can occur in other locations
sidered midline tumors and can be either including the midbrain. Patients with NF1
low‐grade or high‐grade. However, the MRI tend to be short and have relative macro-
characteristics favor a low‐grade neoplasm. cephaly. Careful examination looking for
These include the sharp demarcation, the café‐au‐lait spots, axillary freckling, and
hyperintensity on T2 and FLAIR, and the cutaneous neurofibromas should be done.
facilitated diffusion. Both low‐grade and The presence of attention problems or
high‐grade gliomas can enhance, but low‐ learning disabilities should be identified, as
grade gliomas tend to have solid areas of these occur in ~50% of patients with NF1.
enhancement, while high‐grade gliomas Slit lamp examination can reveal Lisch
often have ring enhancement around nodules on the iris. If there is concern about
necrotic‐appearing regions. the possibility of NF1, it is important to
Given the patient’s recent history of pro- perform germline genomic analysis along
gressive symptoms, serious consideration with tumor genomics. Mutations of the
needs to be given to initiating treatment. NF1 gene are not uncommon in low‐grade
Given the location of the tumor and the glial tumors, and without concomitant
patient’s symptoms, surgical removal is not a germline testing it will not be possible to
realistic option without leaving the patient know whether an intratumoral NF1 muta-
with permanent, significant neurological tion is somatic or germline. This could have
dysfunction. In fact, even a biopsy could major implications for future therapy.
represent significant risk of permanent Treating patients with NF1 with radiation
injury. However, depending on location, it therapy carries a risk of significant mor-
may be possible to obtain sufficient tissue bidity, including secondary malignancy
for diagnosis using a stereotactic approach. and the development of potentially fatal
With the recent advances in genomic analy- Moyamoya disease.
sis and the possibility that molecular profil-
ing could identify mutations for which novel 2. A 12‐month‐old girl is referred for imag-
treatment options exist, an attempt should ing by a neurologist, after presenting with
be made to obtain sufficient tissue to per- bilateral nystagmus for several weeks. Her
form such testing. If this is deemed too risky, past history is significant for FTT, with min-
initiation of therapy with conventional imal weight gain since 9 months of age. She
chemotherapy should definitely be consid- was admitted at 10 months of age due to
ered. While radiation therapy is effective concern for malnutrition and not receiving
therapy for low‐grade gliomas, it should not adequate oral intake. She is breastfeeding
be considered without a biopsy‐proven and taking solid foods, often eating through
diagnosis. Given the patient’s age and the the night.
tumor’s proximity to the brainstem, pitui- At the time of presentation to the neu-
tary gland, and primary cerebral vascula- rologist, she is noted to be alert and attentive
ture, radiation therapy should be considered but cachectic. Her weight to length ratio is
second‐line therapy. <3rd percentile. Her body mass index (BMI)
226 Chapter 16

is 12.2 kg/m2. She is very thin and irritable, begin to gain weight again normally, though
but consolable by her mother. Her anterior it may take many months to see improve-
­fontanelle is flat. She has symmetric tone ment. However, diencephalic syndrome
and strength and no localizing neurologic has been associated with a worse tumor
findings and normal cranial nerves. She outcome.
appears to have good vision, with extraocu- c. How should diagnosis and treatment
lar movements intact, though has easily be approached in this patient?
elicited nystagmus bilaterally. MRI of the brain shows a homogenously
a. What is the most likely diagnosis and enhancing suprasellar mass which is
how is her history relevant? 5.1 × 2.4 × 2.7 cm. The patient subsequently
This patient most likely has a slow growing has a spine MRI which reveals metastatic
low‐grade glioma or astrocytoma located in spinal lesions. The patient undergoes a
the hypothalamic‐optic chiasmatic region. biopsy of the mass which reveals a juvenile
The association of a tumor in this location pilomyxoid astrocytoma (JPA). Given the
in an infant or young child with features of location and size of the tumor, concurrent
FTT is referred to as “diencephalic syn- diencephalic syndrome, and spinal metas-
drome.” This is a rare disorder and children tases, treatment with chemotherapy is
often present severely emaciated despite warranted. The patient is referred for an
a normal to slightly decreased caloric ophthalmology evaluation and ongoing
intake. These children may also appear to be monitoring given the risk of visual deficits
in a hypermetabolic state with hyperactivity, due to optic chiasm involvement. Additio­
hyperalertness, tachycardia, fever, excessive nally, given the risk for development of
sweating, and skin flushing. They are often endocrinopathies secondary to the supra-
thought to have been neglected, poorly fed, sellar location, she has assessments of follicle
or have an underlying malabsorption prob- stimulating hormone (FSH), luteinizing
lem. It is not unusual that these children have hormone (LH), thyroid stimulating hormone
seen multiple specialists including gastroen- (TSH), and growth hormone (GH).
terology and endocrinology to evaluate for
celiac disease, pancreatic insufficiency, or an
endocrine problem. These children are not Suggested reading
developmentally affected. Diencephalic syn-
drome should be included in the differential Millard, N.E. and De Branga, K.C. (2016).
diagnosis of FTT in an infant or very young Medulloblastoma. J. Child Neurol. 31:
child without apparent cause. 1341–1353.
Neill, S.G., Saxe, D.F., Rossi, M.R. et al. (2017).
b. Will treatment of her tumor result in
Genomic analysis in the practice of surgical
reversal of her FTT? neuropathology: the Emory experience. Arch.
It is unclear how a tumor in the hypotha- Pathol. Lab Med. 141: 355–365.
lamic region affects appetite and metabo- Pickles, J.C., Hawkins, C., Pietsch, T., and Jacques,
lism. The therapy is focused on treating the T.S. (2018). CNS embryonal tumors: WHO 2016
underlying lesion as well as optimizing and beyond. Neuropathol. Appl. Neurobiol.
nutrition. Most children do recover and 44: 151–162.
17 Hodgkin and Non‐
Hodgkin Lymphoma
Hodgkin and non‐Hodgkin lymphoma (HL derive from antigen‐selected germinal
and NHL) together account for approxi- center B‐cells. In both forms of the disease,
mately 10–12% of malignancies in children; the tumor cells comprise only 0.1–10% of
they are third in relative frequency after the cells within the tumor.
acute leukemia and brain tumors. NHL com- Epidemiologic studies suggest that two
prises approximately 60% of all lymphomas. different forms of pediatric HL exist: child-
Utilization of a risk‐ and response‐based hood and adolescent/young adult (AYA). The
treatment approach in lymphoma has childhood form is defined as occurring in
resulted in improved outcomes and decreased those ≤14 years of age, and is characterized by
long‐term adverse effects. a male predominance and a histologic sub-
type that is more likely to be mixed cellularity
(30–35%) or nodular lymphocyte‐predomi-
Hodgkin lymphoma nant (10–20%). AYA HL occurs in patients
between 15 and 35 years of age. There appears
HL is rare among children <5 years of age to be a complex interaction between Epstein–
and relatively rare in the adult population, Barr virus (EBV) virus and HL development,
but is the most commonly diagnosed cancer with EBV‐transformed lymphoid and Reed–
among adolescents aged 15–19 years. HL is Sternberg cells effecting apoptotic and repair
classified into two general groups: classical mechanisms. Incorporation of EBV into the
HL (95% of cases) and nodular lymphocyte‐ tumor genome is more commonly seen in a
predominant HL (NLPHL, 5% of cases). younger male population (i.e., below 10 years
Classical HL is further subdivided into of age), in underdeveloped countries and in
nodular sclerosis, mixed cellularity, immune‐deficient patients. It is more com-
lymphocyte‐depleted, and lymphocyte‐rich monly associated with the mixed cellularity
forms. The tumor cells in classical HL are histologic subtype of HL. In the AYA popula-
designated Hodgkin and Reed–Sternberg tion, the incorporation of EBV into the tumor
(HRS) cells, whereas in NLPHL they are genome is unusual, though these patients
designated lymphocyte‐predominant (LP) often have a history of infectious mononu-
cells. HRS cells are thought to derive from cleosis with high EBV titers. Other infectious
germinal center B‐cells that have acquired agents that have been associated with HL
unfavorable immunoglobulin V gene include human herpesvirus 6 and cytomeg-
mutations and normally would have alovirus. There is clearly an association
undergone apoptosis, whereas LP cells between HL and alterations in immune

Handbook of Pediatric Hematology and Oncology – Children’s Hospital & Research Center Oakland,
Third Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
© 2021 John Wiley & Sons Ltd. Published 2021 by John Wiley & Sons Ltd.
228 Chapter 17

f­unction, and HL is associated with congenital the HRS cells within the tumor. Certain
and acquired forms of immune deficiency ­specific symptoms known as “B symptoms”
including HIV disease, post‐hematopoietic have been demonstrated to have a negative
stem cell or solid organ transplant, and auto- prognostic significance. These are unplanned
immune disease. weight loss of >10% of body weight over the
6 months preceding diagnosis, drenching
Clinical presentation night sweats, or unexplained, recurrent fever
The most common presentation of HL is >38 °C. Other common constitutional symp-
painless, progressive enlargement of superfi- toms include generalized pruritus and alco-
cial lymph nodes, most commonly cervical, hol‐induced pain. HL may present with
followed by supraclavicular, axillary, or discrete lesions in the lungs, liver, spleen,
inguinal adenopathy. The nodes are typically bone, bone marrow, or kidneys, and is often
described as feeling rubbery or firm and asymptomatic. A diffusely enlarged liver or
non‐tender, although may be sensitive to spleen is not characteristic of HL.
touch with rapid growth. As the disease pro-
gresses, the nodes enlarge and often aggre- Evaluation
gate into larger masses that may become 1. A careful history and physical
fixed to the underlying tissue. Bulky disease examination are imperative, including
is defined as a node or nodal mass measur- assessment for the presence of B symptoms.
ing >6 cm in the largest diameter. Bulky dis- All superficial nodal groups should be
ease is much more common in the AYA carefully examined, and the size of any
population. As children often have reactive enlarged nodes and their character should
or infectious adenopathy, it is common to be documented.
have received treatment with several courses 2. The laboratory evaluation includes a
of antibiotics before ultimately coming to complete blood count (CBC) with
biopsy. This is more common with cervical differential, complete metabolic panel with
than with supraclavicular adenopathy, as the assessment of liver and kidney function,
latter is less frequently associated with infec- lactate dehydrogenase (LDH), and alkaline
tion or inflammation and leads more quickly phosphatase. Acute phase reactants,
to referral for possible malignancy. At least nonspecific markers of tumor activity, may
two‐thirds of patients with cervical or supra- correlate with prognosis or response
clavicular adenopathy have mediastinal measures and include erythrocyte
involvement as well. This is frequently sedimentation rate (ESR), C‐reactive protein
asymptomatic but may be associated with (CRP), serum copper, and serum ferritin.
cough (usually nonproductive), dyspnea, 3. A posterior–anterior (PA) and lateral
orthopnea, hoarseness, dysphagia, chest chest radiograph is requisite to determine if a
pain, or evidence of superior vena cava syn- mediastinal mass is present. Before initiating
drome (SVCS) (with distended neck veins a diagnostic surgical procedure under anes-
and worsening respiratory or cardiac dys- thesia, it is important to determine if the
function). HL limited to sub‐diaphragmatic patient may be at risk for complications due
sites occurs in <5% of pediatric cases, occur- to an obstructing mediastinal mass. Other
ring most commonly in femoral, superficial important imaging studies include a CT scan
iliac, or inguinal nodes. of the neck (inclusive of Waldeyer’s ring),
Constitutional symptoms occur in chest, abdomen, and pelvis. Oral and intrave-
approximately 25% of patients with HL. nous contrasts are required for accurate iden-
They are caused by cytokines produced by tification of intra‐abdominal adenopathy.
Hodgkin and Non‐Hodgkin Lymphoma 229

Splenic involvement occurs in 30–40% of mediastinum), and the number of involved


children at diagnosis. Positron emission sites then determines the stage. Stage I dis-
tomography (PET) is now used at diagnosis ease involves only a single nodal site, whereas
as a very sensitive tool for identifying sites of Stage II involves two or more sites that lie
involvement as well as to evaluate subsequent together on one side of the diaphragm. Stage
treatment response. III disease spans the diaphragm and Stage IV
4. Bone marrow involvement at initial pres- is disseminated systemic disease. Additional
entation of pediatric HL is uncommon and modifiers include A or B, to describe the
rarely occurs as an isolated site of extranodal absence or presence of specific systemic
disease. However, it is more commonly symptoms, respectively, and E, involvement
associated with advanced stage (Stage III or of a single extranodal site that is contiguous
IV) disease or in patients with B symptoms or proximal to a known nodal site. The pres-
(though likely less in Stage I or II patients). ence of bulky mediastinal disease, defined as
These patients should undergo bone mar- greater than one‐third of the intrathoracic
row aspiration and biopsy from at least two diameter, is also considered. Staging is
sites as part of their diagnostic evaluation. divided into low‐, intermediate‐, and high‐
All patients experiencing a recurrence of risk disease categories (early‐, intermediate‐,
their disease should have their bone marrow and advanced‐stage diseases in Europe)
examined. based on prognostic factors such as the extent
5. Skeletal metastases are also rare at diag- of disease (including extralymphatic and
nosis. FDG‐PET is a sensitive test for identi- splenic involvement) as well as absence or
fying bone metastases and has replaced presence of bulky disease as well as B
technetium bone scan. It is important to symptoms.
include the entire distal extremities in the Disease categorization into these risk
imaging. groups is not uniform with some potential
6. Surgical procedures are limited to a diag- overlap:
nostic biopsy, as resection of mass disease is 1. Low‐risk disease: Stage IA or IIA without
not indicated in the majority of cases. The bulky disease, splenic, or extralymphatic
exception is the Stage IA patients with involvement (potentially IB).
NLPHL who may be observed without fur- 2. Intermediate‐risk disease: IA/IIA with
ther treatment after full resection. Excisional bulky disease, IB, IIB without bulky disease,
biopsy should be performed (not fine‐needle IIIA (potentially also IIB with bulky disease
aspiration) to obtain sufficient tissue for diag- and IVA).
nostic studies including tumor characteriza- 3. High‐risk disease: IIB with bulky disease,
tion for potential targeted therapies. IIIB, IV.
Additionally, excisional biopsy allows for Initial clinical and laboratory risk assess-
preservation of the lymph node architecture ment, stage, and FDG‐PET response to ini-
which aids in diagnosis. Subtypes of HL are tial treatment cycles form the basis of risk
determined based on histology, gross appear- stratification and the development of treat-
ance, and presence of Reed–Sternberg cells. ment algorithms. Additional risk factors
include elevated acute phase reactants, age,
Staging anemia, and male sex.
The most widely used staging system for
pediatric HL is the Ann Arbor staging system Treatment
(Table 17.1). This system divides lymph node The first successful treatment strategy for
regions into nodal sites (i.e., neck, axilla, and HL was based solely on the use of radiation
230 Chapter 17

Table 17.1 Ann Arbor staging system for Hodgkin lymphoma.

Stage Description

I Single lymph node region (I) or extralymphatic organ or site (IE)


II Two of more lymph node regions on the same side of the diaphragm (II) or
single extralymphatic organ with regional or other lymph node involvement
on same side of diaphragm (IIE)
III Involved lymph node regions on both sides of diaphragm (III), potentially inclusive
of contiguous extralymphatic organ (IIIE) or spleen (IIIS) or both (IIIE+S)
IV Disseminated involvement of one or more extra‐lymphatic tissues or organs or
single extra‐lymphatic organ involvement with distant nodal involvement

B symptoms: (i) unexplained and documented weight loss >10%; (ii) unexplained recurrent
documented fever >38.0 °C; and/or (iii) drenching night sweats.
Bulk disease: (i) mediastinal mass ≥ 1/3 thoracic diameter measured on upright PA CXR; and/or
(ii) aggregate nodal tissue >6 cm in longest transverse diameter.

therapy (RT). While this approach was fairly treated without IMRT. A variety of chemo-
effective in curing the disease in low‐risk therapy agents have been shown to be effec-
patients, it resulted in unacceptable muscu- tive in treating HL and many regimens have
loskeletal hypoplasia, cardiovascular and proven successful. Current strategies include
pulmonary dysfunction, and the develop- minimizing radiation especially in females
ment of subsequent secondary cancers. This to decrease risk of breast cancer secondary to
led to the development of combined modal- mediastinal radiation and minimizing
ity therapy with the goal of improving event‐ alkylator therapy especially in males to
free and overall survival, especially in decrease risk of infertility. With current ther-
higher‐risk patients, and decreasing the apy, more than 90% of children diagnosed
long‐term adverse effects of a radiation‐only with classical HL are expected to be long‐
approach. Current research efforts in HL term disease‐free survivors, regardless of
treatment seek to maintain the excellent sur- stage at diagnosis.
vival rates presently achieved while continu-
ing to decrease the frequency of later adverse Classical HL—low‐risk disease
events by utilizing a risk‐based and early‐ A number of studies have attempted to elim-
response approach to decrease chemother- inate the use of RT completely in children
apy and radiation exposure. Today, patients with low‐risk disease based on PET response
with favorable disease presentations receive after two cycles of chemotherapy. Multiple
fewer cycles of multi‐agent chemotherapy therapeutic options exist with similar excel-
than those with advanced and unfavorable lent outcomes utilizing a response‐based
clinical presentations, either alone or com- approach to RT. Current commonly used
bined with low‐dose, involved‐field radia- regimens that are radiation‐sparing include
tion (intensity‐modulated radiation therapy, the German OPPA/OEPA (vincristine
IMRT). The current approach to therapy [Oncovorin], procarbazine or etoposide,
relies on a response‐based assessment and prednisone, Adriamycin [doxorubicin]) reg-
risk stratification utilizing FDG‐PET imag- imen and VAMP (vinblastine, Adriamycin,
ing after two to four cycles of chemotherapy. methotrexate, prednisone). Other regimens
Patients with early response can typically be may also be considered with evaluation of
Hodgkin and Non‐Hodgkin Lymphoma 231

chemotherapeutic agents, their long‐term in females. Many reasonable regimens exist,


side effects, and the potential of the regimen thus there is not a standardized approach. The
to be radiation‐sparing. FDG‐PET response practitioner must weigh the particular side
after two cycles of therapy helps to define effects of each regimen and the potential desire
which patients can avoid IMRT. to avoid RT.

Classical HL—intermediate‐risk Nodular lymphocyte‐


disease predominant HL
Similar to low‐risk disease, the goal for Several studies have demonstrated that
patients with intermediate‐risk disease is to patients with NLPHL who present with Stage
utilize a response‐based approach to mini- I disease and a single involved lymph node
mize need for RT, as well as potentially mini- can be successfully treated with surgery
mizing alkylator therapy in males. As alone. Stage I patients with more than one
mentioned, there is overlap in staging in involved node and Stage II patients, which
regard to which groups are classified as inter- represent more than 80% of patients with
mediate‐risk. Additionally, there are multiple NLPHL, are successfully treated with similar
acceptable treatment regimens. Current ther- protocols used in low‐risk classical HL
apies include ABVE‐PC (Adriamycin, bleo- patients and can avoid RT if with a complete
mycin, vincristine, etoposide, prednisone, response to this therapy. A recent analysis of
and cyclophosphamide) as well as OEPA/ intermediate‐risk NLPHL patients treated
COPDAC for males (cyclophosphamide, with ABVE‐PC showed excellent early
Oncovorin, prednisone, dacarbazine) and response and event‐free response, superior to
OPPA/COPP for females (procarbazine classical HL and with high rates of RT avoid-
rather than dacarbazine). FDG‐PET response ance. Salvage rates are excellent for those that
after two cycles of therapy helps to define are observed after surgery and in those that
which patients can avoid IMRT, depending recur after chemotherapy.
on the treatment regimen utilized.
Novel agents
Classical HL—high‐risk disease Refractory or recurrent HL may require
Given the excellent prognosis and ability to sal- alternative approaches including salvage
vage even high‐risk patients with HL, reduction therapies with conventional chemotherapy,
in radiation remains a priority in this subgroup, IMRT, autologous hematopoietic stem cell
especially in females. There is no standardized transplant, or targeted antibody therapy.
approach, and risk‐grouping may be overlap- Brentuximab vedotin is a promising new
ping for some between intermediate‐ and agent in HL, and is a targeted antibody–drug
high‐risk. Early response may still allow for conjugate that has shown an excellent
elimination of IMRT in some groups, depend- response and toxicity profile in CD30+ lym-
ent on the treatment protocol, such as in the phomas, including HL. A highly toxic agent
use of BEACOPP (bleomycin, etoposide, monomethyl auristatin E is delivered to
Adriamycin, cyclophosphamide, Oncovorin, CD30+ cells after internalization and proteo-
procarbazine, prednisone) followed by COPP/ lytic cleavage. Brentuximab is generally well–
ABV in females. Additional treatment regimens tolerated, though may cause fever, fatigue,
include BEACOPP + RT, BEACOPP/ABVD diarrhea, nausea, neutropenia, peripheral
(Adriamycin, bleomycin, vinblastine, dacar- neuropathy, and pneumonitis. Brentuximab
bazine) + RT in males, as well as OEPA/ has been used successfully in the relapse set-
COPDAC + RT in males and OPPA/COPP + RT ting either as a single agent or in combination
232 Chapter 17

with bendamustine, a bifunctional alkylating lymphoma (BL). During childhood, the


agent with only partial cross‐resistance to disease is more common among non‐
­
other alkylators. Brentuximab has also Hispanic whites and Asians/Pacific Islanders;
proved important in preventing relapse in after age 20 years, it occurs more commonly
high‐risk patients when given as an every 3‐ in African–Americans. The incidence of dif-
week 1‐year maintenance therapy after autol- ferent histologic subtypes is very different in
ogous stem cell transplantation (ASCT). children and adults.
Other new agents are also in develop- Children with congenital or acquired
ment and include targeted PD‐1 blockade immune system dysfunction have a high risk
(PD: programmed cell death), which has of developing lymphoma. Congenital condi-
recently been shown effective given the PD‐1 tions include ataxia‐telangiectasia, autoim-
pathway effect on limiting T‐cell‐mediated mune lymphoproliferative syndrome, as well
tumor killing. Upregulation of PD‐L1 (ligand as Bloom, Wiskott–Aldrich, and Chédiak–
of PD‐1) has been shown in certain tumor Higashi syndromes; acquired conditions
types including HL as a method to evade include HIV and prolonged immunosup-
immunosurveillance. Blocking PD‐1 pressive therapy following bone marrow or
through antibodies such as nivolumab and solid organ transplant or treatment for auto-
pembrolizumab has proven effective, espe- immune conditions. EBV is clearly associ-
cially in tumors with PD‐L1 expression. ated with BL in low‐income countries, and
Studies in heavily pretreated HL patients has been linked to several subtypes of NHL
have shown a response to nivolumab. Early in high‐income countries. Other identified
studies with nivolumab and brentuximab in risk factors include Helicobacter pylori infec-
combination in heavily pretreated patients tion, tobacco exposure, and chemical or
have shown efficacy and are ongoing. PD‐1 other environmental exposures. It is likely
antibodies have been shown tolerable with that there is not a single etiology responsible
rash, diarrhea, and pruritus being common for the development of all cases of NHL.
side effects in addition to less common The histologic classification of NHL has
immune‐mediated effects including pneu- changed frequently over time and has become
monitis, colitis, thyroiditis, hepatitis, and more precise, as our understanding of the
hypophysitis (pituitary inflammation). development of lymphoma has improved and
Tumor pseudoprogression after initiation of better diagnostic tools (immunophenotyp-
a PD‐1 antibody should be a consideration ing, cytogenetics, molecular biology, and
due to the initial inflammatory response. gene profiling) have been developed. The
World Health Organization classification is
now widely used and was most recently
Non-Hodgkin lymphoma revised in 2016. There are four major histo-
logic subtypes of NHL that occur commonly
NHL is about 1.5 times as common as HL in in children (in order of frequency): diffuse,
the pediatric and AYA populations, with a large B‐cell lymphoma (DLBCL), T‐ and
median age of 10 years. The incidence is B‐lymphoblastic lymphoma (LL), BL, and
extremely low in children less than 5 years of anaplastic large cell lymphoma (ALCL).
age, but from that point it increases steadily Other considerations in adolescents in par-
with age throughout life. In all age groups, ticular with mediastinal disease are primary
there is a significant male predominance, mediastinal B‐cell lymphoma (PMBCL) as
particularly among patients with Burkitt well as marginal gray zone lymphoma.
Hodgkin and Non‐Hodgkin Lymphoma 233

DLBCL and ALCL tend to be heterogene- pathologic examination following a “routine”


ous immunologically. Most are B‐cell‐derived, tonsillectomy and adenoidectomy.
although some are T‐cell‐derived or arise
from the macrophage‐histiocyte lineage. Diagnostic evaluation
Burkitt and Burkitt‐like subtypes are virtually It is not unusual to begin with a surgical pro-
all B‐cell tumors, distinguished only by the cedure for excision or biopsy of a node or
amount of cellular heterogeneity. The most laparotomy in the case of an acute abdominal
common site of occurrence of LL is the medi- presentation and incidentally diagnose the
astinum, and these tumors are virtually all of patient with NHL. Many times, the diagnosis
T‐cell origin. Less commonly LL occurs in is not suspected prior to these procedures.
bone or subcutaneously; these tumors are Histologic confirmation is required for diag-
typically of B‐cell origin. All of these subtypes nosis. The diagnosis of NHL is usually made
can invade the bone marrow and undergo using biopsy tissue, but can also be confirmed
leukemic transformation. Pediatric NHL is by cytological examination of bone marrow,
mostly high‐grade, which is distinctly differ- cerebrospinal fluid, or pleural or paracentesis
ent from the adult population in which many fluids. In cases where the patient may be too
cases are indolent. unstable for biopsy (i.e., mediastinal mass
with significant compression of the great ves-
Clinical presentation sels or trachea), sampling from these sources
The clinical presentation of NHL varies and may be sufficient to make a diagnosis and
depends on the primary site of disease, histo- initiate treatment. If possible, sufficient mate-
logic subtype, and extent of disease. More than rial should be obtained for immunologic and
70% of children present with advanced‐stage cytogenetic or molecular biology studies.
disease and up to 25% of patients present with Many therapeutic trials now have require-
a mediastinal mass (usually T‐LL or PMBCL). ments for submission of material for central
NHL can arise anywhere in the body, but pathology review or b ­iology studies; these
occurs most frequently in the lymph nodes, requirements should be taken into considera-
thymus, Waldeyer’s ring, Peyer’s patches, and tion when planning a diagnostic procedure.
bone marrow. In the United States, BL typi- The laboratory studies that should be
cally presents in the intestine at the ileocecal obtained include:
region, resulting in obstruction and may lead ●● A CBC with differential and review of the

to intussusception. These children usually pre- peripheral smear to assess for possible bone
sent with nausea, vomiting, and abdominal marrow involvement or leukemia.
distention. It is not unusual for NHL to be con- ●● Serum chemistries to include liver transam-

fused with a surgical abdomen, such as appen- inases, blood urea nitrogen, creatinine, LDH,
dicitis. BL also presents in Waldeyer’s ring or uric acid, electrolytes, calcium, and phospho-
in the facial bones. T‐LL most commonly rus. Lymphomas, particularly LL and BL, can
arises from the thymus and these children present with overt tumor lysis syndrome
typically present with evidence of mediastinal (TLS) and/or renal dysfunction. The LDH
obstructive symptoms such as respiratory can be useful prognostically, especially in
symptoms (cough, dyspnea, orthopnea), cer- patients with advanced‐stage disease.
vical or supraclavicular adenopathy, or SVCS. ●● Bilateral bone marrow aspiration and

Children may have very limited disease affect- biopsy, as the child may have leukemia
ing the tonsils, nasopharynx, or Waldeyer’s (defined as a marrow with >25% lympho-
ring, and the diagnosis often results from blasts) or lower‐level bone marrow disease.
234 Chapter 17

This assessment may be omitted in certain ❍ PET to include all identified sites of

subsets in which it is a rare site of disease disease. Ideally, this should be performed
involvement (i.e., DLBCL, ALCL, PMBCL). following the diagnostic procedure, as
●● A lumbar puncture to assess the cerebro- these patients frequently have residual
spinal fluid for involvement by NHL. abnormalities in their surgical bed on
●● Consideration of testing for immune‐ plain CT that are discovered on FDG‐
deficiency‐specific infections (HIV), or PET to not represent active disease.
immune dysfunction if appropriate.
●● Radiologic procedures that should be per-

formed based on associated symptoms, but Staging


should include: NHL is staged using the St. Jude (Murphy)
❍ A prompt PA and lateral chest radio- staging system that is presented in Table 17.2.
graph to evaluate for mediastinal
involvement. Treatment
❍ CT scan of the neck, chest, abdomen, Therapy is based on clinical staging, localized
and pelvis if the patient is sufficiently versus disseminated disease, and histologic
stable. subtype. Those patients presenting with an
❍ MRI of the brain for patients with oncologic emergency (i.e., SVCS, respiratory
immunodeficiency‐related NHL, as these compromise, clinical TLS) must have emer-
patients more commonly present with gent therapy to ensure their safety, with
parenchymal brain disease. correction of metabolic derangements and/or
❍ MRI of the brain and spine for patients minimal diagnostic studies prior to therapy
with focal neurologic symptoms. initiation. Patients with LL are treated with

Table 17.2 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
Hodgkin and Non‐Hodgkin Lymphoma 235

acute lymphoblastic leukemia‐type therapy (ALK) is common in ALCL and therefore


with similar excellent outcomes. Patients with ALK inhibitors crizotinib and ceritinib are
B‐cell lymphoma (i.e., DLBCL/BL) or ALCL attractive agents for treating ALCL in addi-
(in addition to other lymphoma subtypes) tion to brentuximab.
require a shorter, more intensive treatment Additional immunotherapies being stud-
regimen with excellent outcomes. In gen- ied in NHL target CD19‐positivity which is
eral, the chemotherapeutic regimens include common in B‐cell lymphoma, similar to
steroids, anthracyclines, vinca alkaloids, pre‐B‐ALL and inclusive of bispecific T‐cell
topoisomerase inhibitors, and alkylating engaging antibodies such as blinatumomab
agents in different combinations and sched- as well as chimeric antigen receptor T‐cell
ules. Patients with low‐stage DLBCL or LL (CAR‐T) cell therapy (see Chapter 15).
(i.e., Stage I resected and abdominal Stage II) Blinatumomab has shown benefit in adult
can be treated effectively with two cycles of patients with DLBCL and has not been stud-
chemotherapy, consisting of cyclophospha- ied systematically in pediatric patients.
mide, Oncovorin (vincristine), prednisone, Similarly, the FDA recently approved axi-
and Adriamycin (doxorubicin) (COPAD). cabtagene ciloleucel (Yescarta) CD19+
Patients with more advanced DLBCL, BL, and CAR‐T cell therapy for adult patients with
ALCL require an initial therapy “prophase” relapsed/refractory DLBCL. Response rates
secondary to the very high risk of TLS due to have been less than CD19+ ALL secondary to
tumor bulk as well as the exquisite sensitivity the solid tumor microenvironment and loss
to chemotherapy. Patients should be moni- of CAR‐T cell persistence. Pediatric studies
tored closely for metabolic abnormalities are ongoing and although have shown excel-
and may require rasburicase prophylaxis lent CAR‐T tolerability, responses have not
especially in the setting of bulky disease or been durable. There is potential to combine
evidence of renal involvement or impairment. these agents with PD‐1 blockade to improve
For B‐cell lymphoma and mature acute B‐cell CAR‐T cell persistence; further study is
leukemia, rituximab is now recommended in required.
addition to combination chemotherapy for
high‐risk patients (i.e., Stage III with LDH > 2×
the upper limit of normal (ULN) and Stage Case study for review
IV) with evidence of benefit.
Radiotherapy is generally reserved for You are a pediatrician seeing a 17‐year‐old
patients with CNS‐positive LL, relapsed female patient for a sick visit who notes
patients, or oncologic emergencies due to weight loss over the last several weeks as
tumor compression. CNS prophylaxis is well as generalized itching. She had
indicated in LL and advanced‐stage B‐cell checked her weight a couple months ago at
lymphoma, as well as in those patients with school and then again yesterday which
parameningeal or overt CNS disease at showed a 5‐pound weight loss. Her mother
diagnosis. was concerned about this and asked her to
see you.
Novel agents a. What are some important follow‐up
Similar to HL, utilization of brentuximab questions?
has shown benefit in CD30+ tumors such as It is first important to ask questions regarding
ALCL as well as PD‐1 blockade in tumors the weight loss—Has the weight loss been
which express PD‐L1 such as PMBCL. Gene intentional? Has she been exercising intensely
expression of anaplastic lymphoma kinase or have concerns regarding body image? Are
236 Chapter 17

there any concerns regarding anorexia or examined for respiratory distress as well as
bulimia? Has she been ill? Are there are any auscultation to rule out a pericardial rub or
food resource issues? Is she skipping meals diminished breath sounds consistent with
especially at school? Any recent travel (to rule pleural effusion.
out infectious diarrhea and tuberculosis) or The exam is concerning for bilateral
unprotected sexual encounters (consideration neck fullness with palpable bilateral cervical
for HIV)? Assuming her answer to all of these nodes, some >1 cm but otherwise normal.
questions are no, you should be concerned c. What are the next steps given the
about unintentional weight loss without evi- concerns?
dence of an infectious cause. It is important The clinical history and exam are concern-
then to ask about other B symptoms including ing for a HL. Given the paucity of physical
recurrent fever and drenching night sweats. symptoms, it appears the patient has a slow
Itching could be due to an infestation such as growing process rather than a rapidly
scabies, but may be a manifestation of under- enlarging NHL, though this is still a possi-
lying HL. It is important to inquire regarding bility. Baseline labs should be done to assess
other systemic manifestations of disease such for anemia, metabolic derangements, as well
as fatigue (anemia can also be seen in HL due as potential tumor markers. The practi-
to chronic disease/inflammation), secondary tioner should order a CBC/diff, complete
amenorrhea, as well as any noted shortness of metabolic panel, phosphorus, LDH, uric
breath, orthopnea, or lumps/bumps. acid, as well as an ESR and ferritin. Copper
She answers no to all of these follow‐up can also be a potential tumor marker in HL,
questions. though is no longer routinely sent. A screen-
b. What are some important considera- ing chest X‐ray (CXR) is a simple way to rule
tions for physical exam? out a mediastinal mass.
Physical exam should be complete but spe- The labs reveal a mild normocytic ane-
cifically focus on potential findings which mia, no metabolic derangements, and ele-
would support the diagnosis of lymphoma, vated LDH, ferritin, and ESR. CXR reveals
specifically the presence of lymphadenopa- mediastinal widening.
thy or hepatosplenomegaly, as well as d. What is your diagnosis? What are the
symptoms consistent with superior vena
­ next steps?
cava/superior mediastinal syndrome (see The patient has mediastinal adenopathy,
Chapter 14; can see plethora, distended neck though will need histologic proof to confirm
veins, jugular venous distension, hepato- lymphoma and specifically HL. The age of
megaly). It is important to review the vital the patient, presentation with B symptoms,
signs to confirm the weight loss as well as labs, and CXR are all consistent with HL,
document potential fever and tachypnea though other etiologies including T‐cell acute
(secondary to pleural effusion or very large lymphoblastic leukemia and other NHL as
mediastinal mass) or tachycardia (second- well as germ cell tumors and sarcomas are
ary to anemia or very large mediastinal still possible. You want to talk with the patient
mass). Cardiac tamponade would be highly and the family regarding your concerns.
unlikely especially with a slow‐growing Given the difficult nature of the conversa-
mass as seen in HL, but must be a considera- tion, you want to ensure that the patient has
tion especially in a more rapidly growing some family support available. It should be
NHL; checking for pulsus paradoxus should understood that limited information will be
be done if other concerning findings for processed at a time of family stress and it will
SVCS are present. The patient should be be important to repeat the information at a
Hodgkin and Non‐Hodgkin Lymphoma 237

later time. It is okay to focus on next steps


Suggested reading
which will include need for a biopsy, likely
from one of the cervical nodes to confirm
Cairo, M.S. and Beishuizen, A. (2019). Childhood,
the diagnosis. If there are concerns about any adolescent and young adult non‐Hodgkin lym-
cardiorespiratory instability or metabolic phoma: current perspectives. Brit. J. Haematol.
abnormalities, the patient should be admit- 185: 1021–1042.
ted and diagnostic steps undertaken more Metzger, M.L. and Mauz‐Körholz, C. (2019).
urgently. Given the reassuring exam and labs Epidemiology, outcome, targeted agents and
and small amount of mediastinal disease immunotherapy in adolescent and young adult
without orthopnea, it is reasonable to refer non‐Hodgkin and Hodgkin lymphoma. Br. J.
the patient for urgent evaluation (within Haematol. 185: 1142–1157.
days) to a pediatric oncologist, as well as Shanbhag, S. and Ambinder, R.F. (2018). Hodgkin
lymphoma: a review and update on recent
pediatric surgeon for an excisional lymph
progress. CA Cancer J. Clin. 68: 116–132.
node biopsy. Following histologic confirma-
Von Keudell, G. and Younes, A. (2019). Novel ther-
tion of disease, the patient will require a stag- apeutic agents for relapsed classical Hodgkin
ing evaluation including a PET/CT prior to lymphoma. Br. J. Haematol. 184: 105–112.
determining the appropriate treatment plan.
18 Wilms Tumor

Wilms tumor, also known as nephroblastoma, (GU) anomalies, such as cryptorchidism,


is the most common renal tumor in children, hypospadias, horseshoe kidney, ureteral
accounting for approximately 6% of child­ duplication, and polycystic kidney. Aniridia
hood malignancies and 10% of all cancers in is present in approximately 1.2% of children
the 1–4‐year age group. It was named after Dr. with Wilms tumor. Most children with Wilms
Max Wilms (1867–1918), a German surgeon tumor have a normal karyotype; however,
who first described the tumor. A tumor of the mutations of the Wilms tumor 1 (WT1) gene
developing kidney, it typically occurs in young on chromosome 11p13 and PAX6 gene are
children between the ages of 1 and 5 years observed in approximately 20% of patients
with a peak incidence at 3–4 years of age and (PAX6 is highly implicated in children with
approximate equal incidence among boys and congenital aniridia). The WT1 gene encodes
girls (though interestingly occurs at earlier a transcription factor important in normal
ages in boys). Bilateral Wilms tumor is more kidney development and deletion of this gene
common in girls (ratio of 0.6 boys to 1.0 girls) imposes a 30–40% risk of developing Wilms
and in younger children, with a median age of tumor. Mutations within the WT1 gene have
diagnosis of 32 months versus 44 months in also been identified in several syndromes
unilateral cases. Most cases of Wilms tumor associated with Wilms tumor: WAGR syn­
are sporadic, with approximately 1% familial drome (Wilms tumor, Aniridia, GU anoma­
and 2–4% associated with rare congenital syn­ lies, and mental retardation), Denys–Drash
dromes. Familial cases are more likely to pre­ syndrome (Wilms tumor, nephropathy, and
sent with bilateral tumors and occur at a GU anomalies including pseudohermaphro­
younger age. Rarer renal tumors in young ditism), and Frasier syndrome (ambiguous
children include clear cell sarcoma of the kid­ genitalia, streak gonads, and focal segmental
ney (CCSK) and rhabdoid tumor of the kid­ glomerulosclerosis). Beckwith–Wiedemann
ney (RTK). The most common renal tumor in syndrome (macroglossia, omphalocele, vis­
adolescents is renal cell carcinoma. ceromegaly, with or without hemihypertro­
phy) is associated with imprinting defects at
several genes at chromosome 11p15.5, a
Genetics locus referred to as the WT2 gene. Patients
with this syndrome have a genetic predis­
Congenital anomalies occur in 12–15% of position to develop Wilms tumor, with an
cases, the most common being hemihyper­ incidence as high as 5–10%. Other genetic
trophy, aniridia, and genitourinary tract syndromes associated with Wilms tumor

Handbook of Pediatric Hematology and Oncology – Children’s Hospital & Research Center Oakland,
Third Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
© 2021 John Wiley & Sons Ltd. Published 2021 by John Wiley & Sons Ltd.
240 Chapter 18

include: Perlman, mosaic ­variegated aneu­


Evaluation of suspected Wilms
ploidy, Fanconi anemia, and Simpson–
tumor
Golabi–Behmel syndrome.
●● History: include family history of malig­

Clinical presentation nancies, congenital anomalies, or syndromes


known to be associated with Wilms tumor.
●● Physical examination: vitals including
Most children with Wilms tumor are gener­
ally in good health at presentation and come temperature and blood pressure, gently
to medical attention due to abdominal assess for abdominal/flank mass (usually
enlargement or mass detected by a family distinct and not crossing the midline), con­
member or primary care physician on a rou­ genital anomalies (hemihypertrophy, GU
tine check‐up. A palpable abdominal mass is malformations, aniridia), abdominal venous
the presenting sign in 60% of children with distention, and liver enlargement.
●● Laboratory studies: urinalysis with micro­
Wilms tumor. Associated signs and symp­
toms include abdominal pain (usually scopic evaluation, complete blood count
related to obstipation or bleeding within the (polycythemia and anemia), serum chem­
tumor), malaise, fever, hypertension (25%, istries (hypercalcemia in rhabdoid tumor
more likely with bilateral disease or com­ or congenital mesoblastic nephroma), and
pression of renal vessels by the mass), and coagulation studies including von
microscopic hematuria (15–20%). Bleeding Willebrand panel.
●● Diagnostic imaging studies to assess uni­
within the tumor may occur spontaneously
or as a result of trauma and result in anemia, lateral/bilateral involvement, metastatic
pallor, and fatigue. Tumor thrombus may disease, or presence of tumor thrombus in
extend into the vena cava, causing partial the renal inferior vena cava (IVC) or renal
obstruction, hypertension, and distention of vessels:
❍ Doppler abdominal ultrasound to evalu­
abdominal veins. Polycythemia may be seen
in Wilms tumor and its association with ate the size and extent of tumor, presence of
erythropoietin levels is not clear. Acquired tumor thrombus, and contralateral kidney.
❍ Abdominal CT or MRI with special
von Willebrand disease with reduced von
Willebrand factor (VWF), factor VIII coag­ attention to evidence of bilateral involve­
ulant, and ristocetin cofactor levels is ment, renal vessel involvement or exten­
reported in approximately 8% of patients sion into the IVC, lymph node involvement,
newly diagnosed with Wilms tumor and and liver metastases.
❍ Chest radiography and chest CT to assess
typically resolves with therapy initiation. Its
occurrence in Wilms tumor is thought to be for lung metastases (most common site).
❍ Bone scan to assess for metastases
secondary to binding of VWF to the tumor
with subsequent degradation as well as hya­ (more common in CCSK) and for all
luronic acid secretion by nephroblastoma patients with Wilms tumor or renal cell
cells leading to decreased efficacy of VWF. carcinoma with bony symptoms.
❍ MRI of the brain for metastases if the
All children with unexplained polycythemia,
the presence of a syndrome highly associ­ tumor is a RTK or CCSK.
❍ Echocardiogram for detecting tumor
ated with Wilms tumor, or congenital
aniridia or GU anomalies, should be evalu­ extension from the IVC to the right atrium
ated for Wilms tumor during the period of and also indicated in the child receiving
greatest risk. anthracycline chemotherapy.
Wilms Tumor 241

association with congenital anomalies and


Pathologic diagnosis
familial cases. Loss of heterozygosity (LOH)
for polymorphic DNA markers at both chro­
Children with suspected Wilms tumor should
mosomes 1p and 16q occurs in about 5% of
have an immediate consultation with a pedi­
patients with FH Wilms and is associated with
atric surgeon. Surgical removal of the involved
poorer outcomes and increased risk of relapse.
kidney and intact tumor is indicated at diag­
Gain of chromosome 1q is frequently seen in
nosis if possible. Most tumors are encapsu­
association with LOH and appears to be an
lated and vascular, and due to the fragile
independent poor prognostic factor.
nature of these tumors, it is imperative that a
meticulous surgical approach be undertaken
by an experienced surgeon. Rupture may
Staging
occur and increase the risk of bleeding or
peritoneal dissemination and upstage the
Staging is based on anatomic findings, surgical
patient. Cases with tumor in the renal pelvis
outcome, histology, and presence or absence of
and IVC require a detailed imaging assess­
regional or distant disease. Patients unable to
ment to guide the surgeon. In some cases,
undergo a radical nephrectomy at diagnosis
complete removal of the primary tumor at
due to excessive risk receive preliminary staging
diagnosis may not be possible. In these cases,
and preoperative chemotherapy with delayed
open biopsy is indicated for pathologic con­
resection. A careful pathologic evaluation at the
firmation and determination of histology.
time of excision should be performed, as the
The classic pathologic features of Wilms
histology may be different than that seen on
tumor show a triphasic pattern of nephroblas­
initial biopsy. This evaluation includes looking
toma tissue elements: blastema, stroma (mes­
for extension of disease outside the kidney,
enchyme), and epithelium. Tumors are also
including penetrance of the tumor through the
classified as either favorable histology (FH)
renal capsule into the renal pelvis and vessels
(well‐differentiated components) or unfa­
and presence of nodal involvement.
vorable (presence of anaplasia with poor dif­
Clinicopathologic staging is as follows:
ferentiation). Anaplastic features include
●● Stage I: tumor limited to the kidney and
nuclear enlargement and atypia, as well as
completed excised with an intact capsule
irregular mitotic figures. Anaplasia is further
without tumor rupture or biopsy prior to
defined as focal or diffuse based on the distri­
resection; negative regional nodes.
bution of the anaplastic elements. Histology
●● Stage II: tumor penetrates the renal cap­
confers a significant prognostic element, with
sule either by biopsy or rupture prior to
the best outcome in cases of FH, followed by
excision, with spillage confined to the flank;
focal anaplasia and then diffuse anaplasia.
tumor may involve the perirenal soft tissue
Nephrogenic rests refer to areas of persistent
or infiltrate vessels outside the kidney but is
embryonal tissue within the kidney, and, if
completely excised; negative regional nodes.
identified, confer increased risk for develop­
●● Stage III: residual nonhematogenous dis­
ment of Wilms (or recurrent disease if present
semination of tumor confined to the abdo­
in residual kidney or contralateral kidney).
men; tumor may extend beyond surgical
Other malignant tumors of the kidney need to
margin at resection or be removed in more
be considered including RTK, CCSK, and
than one piece, may involve local lymph
renal cell carcinoma. Chromosome analysis
nodes, or have tumor spillage with perito­
on the tumor (and potentially germline from
neal implants; patients with initial biopsy in
the peripheral blood) may prove useful in
lieu of resection are considered Stage III.
242 Chapter 18

●● Stage IV: hematogenous dissemination of be done by an experienced surgeon. Chemo­


tumor outside the abdomen to lungs, liver, therapy is then initiated and the tumor
bone, brain, or distant lymph nodes. reassessed by imaging at 6 weeks. If tumor
●● Stage V: bilateral renal involvement. shrinkage has occurred but risk for spillage
Patients with bilateral involvement of or incomplete resection is still thought to
tumor (Stage V) should have each side inde­ be high, another 6 weeks of chemotherapy
pendently staged (abdominal stage) with is indicated; however, definitive surgery
treatment based on the side with the highest should be performed no later than 12 weeks
stage. after therapy initiation.
Wilms tumors are very sensitive to chem­
otherapy and radiation. Based on the exceed­
Treatment ingly good outcomes, contemporary studies
have looked to decrease therapy in the lower
Wilms tumor is highly responsive to treat­ risk stratum. This includes the elimination
ment with approximately 90% of children of chemotherapy in patients younger than
being cured of their disease. Histology is 2 years with Stage I FH disease and tumor
more prognostic for outcome than surgical mass of <550 g without LOH at 1p and 16q.
staging; for example, patients with Stage IV For patients receiving chemotherapy, the
disease but with FH still have a 4‐year sur­ backbone of therapy is based on previous
vival of more than 85%, compared to chil­ studies by the National Wilms Tumor Study
dren with diffuse anaplasia and advanced Group. Treatment consists of an 18‐week
disease who have very poor outcomes. course of vincristine and actinomycin‐D for
Children with Stages I and II FH tumors have Stages I and II FH patients; 24 weeks of treat­
a 5‐year event‐free survival exceeding 95%. ment with vincristine, actinomycin‐D, and
Many tumors are low stage at presenta­ doxorubicin for those with Stages III and IV
tion (43% Stage I and 23% Stage II) and are FH as well as Stages I–IV with focal anaplasia
able to be fully resected. Radical resection or Stage I with diffuse anaplasia. Patients
(nephrectomy) of the involved kidney is with Stages II, III, or IV with diffuse anapla­
typically the primary approach if the tumor sia receive a 24‐week course of chemother­
is confirmed on imaging to be unilateral and apy with vincristine, cyclophosphamide, and
there is marginal “normal” renal paren­ etoposide. Patients who are determined to
chyma at the periphery. High‐quality CT have LOH at 1p and 16q are upstaged after 6
imaging is critical in surgical planning. weeks of therapy with the addition of
Surgical sampling of lymph nodes should ­cyclophosphamide and etoposide. Gain of
include the perihilar, para‐aortic, and para­ chromosome 1q, a common cytogenetic
caval regions in addition to any visibly finding in Wilms tumor, is also indepen­
abnormal appearing nodes. In the case of dently associated with poorer survival and is
bilateral tumors (and those with increased being incorporated into future risk stratifica­
risk to develop bilateral tumors including tion models of treatment.
syndromes, familial cases, known nephro­ High‐risk patients (Stages III and IV FH
genic rests), a renal sparing approach may and abdominal stages I–III anaplastic) gen­
be warranted. Should the tumor be deemed erally receive radiation therapy in addition
inoperable (i.e., complete resection not to a chemotherapeutic backbone. Radiation
possible) due to tumor extension in the is usually begun within 10–14 days of tumor
renal vessels or IVC, or invasion into adjacent resection and the size of the radiation field
organs, biopsy and nodal sampling should is dependent on tumor extension or spill
Wilms Tumor 243

during surgery. The dose to the tumor bed is adverse effects have influenced modern
1080 cGy. Whole abdominal radiation is treatment protocols with the aim of decreas­
given in the situation of peritoneal seeding ing therapy as possible in future to mitigate
or gross tumor spillage as a result of capsular side effects without decrement in outcomes.
rupture prior to or during surgery. Radiation Screening of children at risk for Wilms
at higher doses (1980 cGy) may be given in tumor is recommended for those with known
unresectable tumor or metastatic nodal congenital anomalies (hemihypertrophy,
involvement. In general, patients with pul­ aniridia) or presence of one of the associated
monary metastasis will require lung irradia­ syndromes discussed in the earlier text.
tion (1200 cGy), although newer trial results Screening consists of a careful physical exam­
suggest this can be eliminated in patients ination and an abdominal ultrasound.
with a rapid response to chemotherapy and Children with Beckwith–Wiedemann syn­
no additional risk factors. drome should be screened every 3 months
Relapse is uncommon in Wilms tumor; until 8 years of age in addition to screening
patients may be salvaged with aggressive for hepatoblastoma with a serum alpha‐feto­
multiagent chemotherapy that may include protein (AFP) through 5 years of age. Testing
agents not given initially such as cyclophos­ for the 11p15 mutation and methylation
phamide, ifosfamide, carboplatin, etopo­ analysis (to identify epigenetic changes asso­
side, topotecan, and irinotecan. Radiation ciated with increased risk) should be consid­
may be utilized depending on the site and ered. Children with Denys–Drash or WAGR
extent of recurrence and if previously not syndromes should be screened to age 5 years
given. High‐dose chemotherapy with autol­ and 7 years, respectively. Children with con­
ogous stem cell rescue has been shown to genital aniridia should be screened for the
improve outcomes after relapse in those that presence of the WT1 gene mutation and, if
have a complete response to chemotherapy present, be screened until age 6 years; if not
and radiation therapy. present, the risk of developing Wilms is simi­
As survival is excellent for most children lar to the general population. Children with
with Wilms tumor, much attention has Wilms tumor who have had nephrogenic
focused on late effects related to therapy and rests identified in the tumor are at risk for late
limiting potential toxicities. Patients are at recurrence or development of tumor in the
risk for hypertension and renal insufficiency, contralateral kidney (or both if renal sparing
as most children will have a solitary remain­ surgery is done in the face of potential bilat­
ing kidney. Patients with bilateral disease, eral involvement) and should be screened for
partial resection, and radiation exposure are 10 years post treatment.
particularly at risk for developing proteinu­
ria and renal compromise in addition to
hypertension. Cardiac complications may be Case study for review
seen in those patients who receive anthracy­
clines and lung irradiation. Additionally, You are seeing an 8‐year‐old girl in the
patients with right‐sided tumors who receive emergency department for a history of 2
flank irradiation or those who received weeks of early satiety and abdominal pain.
whole abdomen irradiation have an Her exam is significant for a left‐sided
increased risk of developing a secondary abdominal mass as well as hepatomegaly.
liver cancer. Treatment interventions should Her initial CBC is significant for microcytic
be considered when generating a long‐term anemia, hgb 7.4 g/dl, elevated LDH, hyper­
follow‐up care plan for the patient. These uricemia, and a prolonged APTT.
244 Chapter 18

a. What would your differential diagnosis tumor can be safely removed intact, and, if
be, given this information? not, then a biopsy alone will be performed.
Initially, this case should be concerning for a Histology is critical to staging and determi­
rapidly dividing hematologic malignancy nation of a treatment plan. Further imaging
with organ infiltration such as Burkitt’s with a bone scan (to determine if bone
lymphoma, given the high uric acid and LDH, metastases are present) as well as positron
as well as anemia. However, the left‐sided emission tomography‐computer tomogra­
mass also raises concern for a renal mass. The phy (PET‐CT) can be considered if the
most common primary renal malignancy in patient is symptomatic or has RTK or CCSK,
­children is Wilms tumor. In this case, the but is generally not required for Wilms
hepatomegaly is concerning for hepatic tumor. In this case, the renal tumor was fully
metastases. Other renal tumors should also resected and pathology revealed FH Wilms
be considered including CCSK (which can tumor.
also present with hepatic involvement), renal c. How does this case differ from a typi­
cell carcinoma (though this is more common cal presentation of Wilms tumor?
in adolescents and adults), and RTK (more There are several factors which are atypical
common in infants and young children). It is in this patient’s presentation. This patient is
also important to look closely at the vital significantly older than the average child
signs, as the patient may have hypertension with Wilms tumor, with a mean age of pres­
secondary to involvement or compromise of entation of 44 months. Older age at diagnosis
the renal vasculature. is associated with an adverse prognosis.
b. What are the next steps in the diag­ While lung metastases are common in
nostic evaluation? Wilms tumor (15%), liver involvement and
Imaging with an abdominal ultrasound is peritoneal nodules are far less common.
the next logical step in the assessment, pro­ Given the hematogenous metastases in this
viding relatively quick information on the case, the patient is classified as a Stage IV
location and potential etiology of the mass. Wilms tumor which represents 11% of all
In this case, the abdominal ultrasound patients. Of note, hepatic involvement at
revealed a mass arising from the left kidney diagnosis is not an independent adverse
as well as multiple hepatic nodules. A subse­ prognostic factor. Even for higher‐stage
quent MRI of the abdomen and pelvis con­ disease, the 4‐year overall survival for FH
firmed these findings as well as multiple Stage IV disease with the current standard of
peritoneal lesions. The finding of a mass care chemotherapeutic regimens is 86%.
arising from the kidney is suggestive of a pri­
mary renal tumor. Chest CT imaging with­
out contrast is recommended to determine Suggested reading
if lung metastases are present. Laboratory
assessment should include a ­ urinalysis Dome, J.S., Fernandez, C.V., Mullen, E.A. et al.
(assessing for bleeding), CBC (can see ane­ (2013). Children’s Oncology Group 2013
blueprint for research: renal tumors. Pediatr.
mia, thrombocytosis), and a von Willebrand
Blood Cancer 60: 994–1000.
panel, as 8% of children with Wilms tumor Irtan, S., Ehrlich, P.F., and Pritchard‐Jones, K.
have acquired von Willebrand disease. In the (2016). Wilms tumor: “State of the art” update,
setting of a prolonged APTT, this should be 2016. Semin. Pediatr. Surg. 25: 250–256.
suspected. After careful review by experi­ Nakamura, L. and Ritchey, M. (2010). Current
enced pediatric surgeons and radiology, a management of Wilms tumor. Curr. Urol. Rep.
determination is made whether the renal 11: 58–65.
19 Neuroblastoma

Neuroblastoma is a neoplasm of the impact on prognosis. Similarly, a near diploid


­sympathetic nervous system and the most (low DNA index, DNA index = 1) chromo-
common solid tumor of early childhood, some number in the tumor has also been
with a peak incidence around 2 years of age. found to be a negative prognostic indicator as
Neuroblastoma is a disease of developing compared to patients with a DNA index >1.
neural crest tissue and has an extremely Based on these factors, in addition to age,
­heterogeneous prognosis based on multiple stage, and histology (favorable or unfavorable
factors such as the age of the patient, differ- based on mitosis‐karyorrhexis index [MKI]
entiation and biology of the tumor, and and maturity of the tumor), a complicated
extent of disease spread. Screening in the risk‐stratified treatment regimen has been
Japanese infant population found a high created and was recently modified.
incidence of neuroblastoma, although the
majority of these cases matured or involuted
spontaneously, again implying the heteroge- Clinical presentation
neous prognosis based on patient age.
Survival rates for neuroblastoma have As neuroblastoma originates from primor-
improved over the last 30 years, initially lim- dial neural crest cells that normally give rise
ited to patients with lower‐risk disease. to the adrenal medulla and the sympathetic
Recent breakthroughs utilizing immuno- ganglia, tumors present in the abdomen and
therapy have increased survival in those along the sympathetic neural pathway. The
with high‐risk disease as well. most common presentation is an abdominal
Familial cases of neuroblastoma are quite mass with primary tumor in the adrenal
rare. However, genetic risk factors for neuro- gland, often with metastatic disease via lym-
blastoma have been identified including the phatic and hematogenous spread. Tumors
anaplastic lymphoma kinase 1 and PHOX2B are often seen along the paraspinal ganglion
homeobox genes. Patients with Hirschsprung’s and may be found in the neck, thorax, and
disease, neurofibromatosis type I, and con- pelvis. Infants are more likely to have tho-
genital hypoventilation syndrome may have racic and cervical tumors. Common sites of
genetic alterations in PHOX2B and an metastasis are the lymph nodes, bone mar-
increased risk of developing neuroblastoma, row, bone, liver, and skin.
although clear genetic links have not as yet The signs and symptoms at presentation
been identified. More commonly, amplifica- depend on the site of the tumor, size, and the
tion of the oncogene N‐myc in tumor tissue degree of spread. Patients with locoregional
has been found to have a profound negative disease may be relatively asymptomatic,

Handbook of Pediatric Hematology and Oncology – Children’s Hospital & Research Center Oakland,
Third Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
© 2021 John Wiley & Sons Ltd. Published 2021 by John Wiley & Sons Ltd.
246 Chapter 19

whereas patients with widely metastatic dis- occult neuroblastoma. Children with OMAS
ease are often ill‐appearing with fever, pain, tend to have a low‐stage, highly curable
weight loss, and irritability. Abdominal neuroblastoma. Unfortunately, cure often
­
tumors are usually palpable, hard, fixed has little impact on the OMAS, and these
masses. Compression of the renal vascula- children are frequently left with severe,
ture may lead to hypertension in addition to chronic neurologic deficits. The pathophysi-
potential catecholamine secretion from ology of this syndrome is not well‐defined,
tumor cells. The liver may be enlarged, lead- but is likely due to formation of an antibody
ing to respiratory compromise due to com- directed against the neuroblastoma cells also
pression of the diaphragm, especially in the targeting cerebellar neurons. In addition to
infant. There may be evidence of anemia treatment of the neuroblastoma, these chil-
(pallor, weakness, and fatigue), coagulopa- dren may benefit from immunosuppressive
thy (bruising and bleeding), and bone pain therapy with agents such as dexamethasone,
or limping with bone or bone marrow cyclophosphamide, intravenous immune
involvement. Thoracic masses are usually globulin (IVIG), and rituximab; adrenocor-
picked up in the posterior mediastinum ticotropic hormone (ACTH) may also be
incidentally by imaging studies done for potentially beneficial. Neuroblastoma
other reasons. Cervical masses may initially patients may also present with hypersecre-
be treated as cervical adenopathy related to tory diarrhea secondary to hypersecretion of
infection. The presence of Horner’s syn- vasoactive intestinal peptide (VIP) from the
drome (miosis [contracted pupil], ptosis, neuroblastoma cells. In most cases, treat-
enophthalmos [posterior eye displacement], ment of the underlying disease will eliminate
and anhidrosis), or heterochromia iridis the VIP hypersecretion.
should prompt an evaluation for cervicotho-
racic neuroblastoma. Pelvic masses may
cause bowel or bladder symptoms and Diagnostic evaluation
tumors along the sympathetic ganglia may
cause spinal cord compression. Skin lesions History
tend to be limited to infants and appear as Assess for constitutional symptoms, abdom-
bluish, nontender subcutaneous nodules. inal pain, bowel or bladder control prob-
Sphenoid or retro‐orbital bone involvement lems, bleeding, bone pain, and limping.
may occur and appear clinically as “raccoon
eyes” secondary to periorbital hemorrhage. Physical examination
In addition to Horner’s syndrome, the Assess vital signs (fever and hypertension)
­clinician should be aware of potential para- and evaluate for abdominal mass, spinal
neoplastic syndromes with neuroblastoma. cord compression, unusual signs/symptoms
One unusual presentation is opsoclonus‐ associated with neuroblastoma (heterochro-
myoclonus‐ataxia syndrome (OMAS), also mia, raccoon eyes, Horner’s syndrome),
referred to as “dancing eyes/dancing feet.” subcutaneous nodules in infants, enlarged
These children have cerebellar and truncal liver or lymph nodes, and evidence of ane-
ataxia as well as myoclonus (muscle jerks) mia or coagulopathy.
and opsoclonus (rapid, involuntary, uncoor-
dinated eye movements). Developmental Laboratory studies
delay, language deficits, and behavioral Obtain complete blood count with differen-
abnormalities (e.g., irritability) may also be tial, serum chemistries (liver function
present. Children with OMAS should be ­studies and lactate dehydrogenase), ferritin
carefully evaluated for the presence of an (may be a tumor marker and elevated in
Neuroblastoma 247

neuroblastoma, though also an acute‐phase with ­ potassium iodide (SSKI) drops and
reactant), and urine for catecholamines
­ should have baseline thyroid function (FT4
including homovanillic acid (HVA) and [free thyroxine] and TSH [thyroid stimulat-
vanillylmandelic acid (VMA). These urine ing hormone]) checked. MIBG, a functional
metabolites are elevated in more than 90% analog of norepinephrine, is taken up by
of children with neuroblastoma, especially sympathetic neurons and is a sensitive test
in the higher stages of disease. In patients for neuroblastoma cells. It is also positive in
with clinical suspicion for neuroblastoma pheochromocytoma. Due to the high radia-
and negative urine markers, urine dopamine tion doses with both bone scan and MIBG
can be measured in addition to 24‐hour scanning, patients can be followed solely
urine collections for HVA and VMA. with MIBG, if available and lesions are
Neuron‐specific enolase (NSE), a serum MIBG-avid. In the case of non-MIBG-avid
marker specific to the sympathetic nervous tumors, PET scanning may be required.
system, is relatively specific to neuroblas- Localized tumors should be surgically
toma, although it can be elevated after brain removed if deemed safe and feasible. Many
injury and seizures. tumors are initially unresectable. These
tumors should be surgically biopsied at the
Diagnostic studies most easily accessible site (often a metastatic
Patients with confirmed neuroblastoma lymph node). Diagnosis is generally estab-
should have bilateral bone marrow aspira- lished by pathologic assessment of the resec-
tion and biopsy performed to assess for tion or biopsy and can be confirmed by
bone marrow involvement. Special stains diagnostic features such as bone marrow
(S100, synaptophysin, and NSE) are used to involvement with appropriate immunohis-
differentiate this tumor from other small, tochemical markers, MIBG avidity, and
round blue cell tumors of childhood such as positive urine catecholamines and serum
rhabdomyosarcoma, Ewing sarcoma, lym- NSE. Genetic studies should be conducted
phoma, and primitive neuroectodermal to determine N-myc and loss of heterozygo-
tumor. sity (LOH) as well as ALK (as a potential
Imaging studies should include: therapeutic target) and segmental chromo-
CT or MRI of the primary site (MRI is somal aberration testing (in potential lower
preferred due to the radiation exposure risk patients).
associated with repeated CT imaging).
Calcifications and hemorrhage are com-
monly seen, especially in large abdominal Staging
masses. The tumors tend to be large and dis-
place adjacent organs; however, the tumor The International Neuroblastoma Staging
can wrap around major structures, causing System was replaced by a new risk‐stratified
obstruction and dysfunction. If there is pretreatment classification system by the
suspicion of paraspinal or intraspinal
­ International Neuroblastoma Risk Group
involvement, MRI and plain films of the (INRG) in 2015 and is outlined in Table 19.1.
spine should be done.
Bone scan (technetium‐99 m) or prefer-
rably, MIBG (metaiodobenzylguanidine) Treatment
scan (123I‐MIBG scan preferred over 131I‐
MIBG due to decreased risk to thyroid Risk stratification divides patients into mul-
function). Prior to MIBG scanning, the
­ tiple potential treatment categories that
patient must receive thyroid protection are continually evaluated through clinical
Table 19.1 International Neuroblastoma Risk Group pretreatment staging system.

INRG stage Age (Mos) Histologic category Grade of tumor N-MYC 11q aberration Ploidy Pretreatment risk
differentiation group

L1/L2 GN maturing, GNB A (very low)


intermixed
L1 Any, except GN maturing NA B (very low)
or GNB intermixed Amplified K (high)
L2 <18 Any, except GN maturing NA No D (low)
or GNB intermixed Yes G (intermediate)
L2 ≥18 GNB nodular, Differentiating NA No E (low)
neuroblastoma Yes H (intermediate)
Poorly differentiated or NA H (intermediate)
undifferentiated Amplified N (high)
M <18 NA Hyperdiploid F (low)
<12 NA Diploid I (intermediate)
12 to <18 NA Diploid J (intermediate)
<18 Amplified O (high)
≥18 P (high)
MS <18 NA No C (very low)
Yes Q (high)
Amplified R (high)

GN, ganglioneuroma, GNB, ganglioneuroblastoma; NA, not amplified.


Neuroblastoma 249

s­ tudies. For simplicity, we divide treatment s­ignificantly. Novel therapeutics modalities


strategies into three main groups: (i) obser- including difluoromethylornithine (DFMO),
vation, (ii) chemotherapy, and (iii) high‐ 131
I‐MIBG therapy, and targeted molecular
dose chemotherapy with autologous stem therapies are being studied to determine
cell rescue and adjuvant therapy with immu- if treatment outcomes can be further
nomodulators. Generally, patients which are improved.
in the very‐low‐risk and low‐risk pretreat-
ment groups (INRG A–F) can be observed
(potentially after surgical resection), while Case studies for review
those in the high‐risk groups (INRG K–R)
require multimodal therapy. 1. You are seeing a 2-month-old female
As mentioned, clinical studies are ongo- infant for routine well-child check and first set
ing to refine treatment groups. Specifically, of vaccinations. Birth history is unremarka-
attempts are being made to reduce treat- ble. The parents overall feel that the child is
ment in some of the lower‐risk subgroups doing well, though note that she appears to
that still require therapy but have an excel- tire a little bit with feeding over the past few
lent overall prognosis (>90% survival). weeks and that her abdomen seems more dis-
Survival in patients with high‐risk disease tended. There has been no fever or intercur-
has improved significantly with a combina- rent illness. She has been stooling and
tion of chemotherapy, surgical resection, urinating normally. On exam, the infant is
autologous transplant (i.e., high‐dose well-appearing with normal vital signs except
chemotherapy with stem cell rescue), radia- for mild tachypnea. There is no pallor or lym-
tion, and immunotherapy. The chemother- phadenopathy. The skin and eye exams are
apy backbone includes drugs such as normal. There is no cyanosis. You note mild
carboplatin, cyclophosphamide, doxoru- increased work of breathing with clear lung
bicin, and etoposide. A recent Children’s sounds. The abdomen though is distended
Oncology Group study showed that tandem with a large, nontender palpable liver. There
transplantation with cyclophosphamide/ are no other concerning clinical findings.
thiotepa and carboplatin/etoposide/mel- a. What is your initial differential
phalan (CEM) was superior to single trans- diagnosis?
plant with CEM alone. It is unclear if this Several entities can cause hepatomegaly
tandem regimen is superior to busulfan/ in a young infant including infections,
melphalan, which is the preparative regimen oncologic processes, storage diseases, and
utilized in Europe as a single transplant. The intrinsic liver pathology. Infection seems
single greatest advance in the treatment of unlikely, as the infant is well-appearing
high‐risk neuroblastoma has been the without any concerning history in the peri-
advent of a monoclonal antibody directed natal period. Common neonatal oncologic
against ganglioside (GD2), an antigen on processes include hepatoblastoma and
neuroblastoma cells. Combination therapy neuroblastoma, with more rare entities
with anti‐GD2 antibodies and cytokine including embryonal sarcoma of the liver.
therapy (GM‐CSF, granulocyte‐macrophage Focal nodular hyperplasia, hamartoma,
colony‐stimulating factor), in addition to hepatic adenoma, hemangioendothelioma
therapy with isotretinoin (cis‐retinoic acid; (with associated Kasabach–Merritt), and
cis‐RA), a neuroblastoma cell maturing Kaposiform hemangioendothelioma are
agent, together has augmented survival processes intrinsic to the liver.
250 Chapter 19

b. What are the next best steps? neuroblastoma will mature spontaneously
Baseline laboratory workup should be done over time. In the case of the relatively
to ensure no significant issues with hemo- asymptomatic patient without concerning
globin and platelets. Kasabach–Merritt may findings on tumor pathology, the patient
cause significant thrombocytopenia second- can be monitored with serial laboratories to
ary to platelet trapping (see Chapter 12). follow tumor markers (NSE, urine HVA in
Abdominal ultrasound would be most this case) in addition to imaging. Any dis-
helpful in this case. Alpha-fetoprotein ease progression or symptomatic involve-
(AFP) can be done if there is concern for ment would prompt chemotherapy to
hepatoblastoma. mitigate continued tumor growth or absolve
Abdominal ultrasound shows a small symptoms, especially respiratory distress
right adrenal mass with heterogeneous with massive hepatomegaly.
liver involvement most consistent with
neuroblastoma. 2. You are working in the emergency
c. What should be done next? department and are seeing a 3-year-old male
Diagnosis of neuroblastoma should be who presents with irritability. The parents
confirmed with a combination of imaging, also note that his eyes look “funny” at times.
laboratories, and ultimately, surgical biopsy. These symptoms have been worsening over
Laboratories should include urine HVA and the last few days. It also seems that he has
VMA in addition to serum NSE. Urine been having trouble walking. There has
catecholamines (dopamine, epinephrine,
­ been no intercurrent illness, no past medical
norepinephrine) can be ordered if the diag- history, he is on no medications, and there is
nosis is equivocal. Imaging should include no concern for any toxic ingestion. Vital
MRI of the primary site and metastatic sites signs are normal. Exam shows eyes with
to further characterize the tumor in addi- random movements rather than particularly
tion to 123I-MIBG scan. Although a positive horizontal or vertical nystagmus. The child
123
I-MIBG scan is pathognomonic for is irritable and is difficult to console by the
­neuroblastoma, surgical biopsy is necessary parents. He refuses to walk. The neurologi-
to confirm the diagnosis. Additionally, cal exam is otherwise unremarkable, with
­bilateral bone marrow aspirate and biopsy no noted loss of tone or hyporeflexia/­
should be done concomitant with surgical hyperreflexia. The cranial nerves appear
biopsy to determine potential bone marrow normal and there are no signs of Horner’s
metastatic disease. syndrome. There is no palpable abdominal
Laboratory studies show a positive urine mass.
HVA and NSE. Follow-up imaging studies a. What is the likely diagnosis? What are
show disease limited to the right adrenal the next steps?
with heterogeneous uptake in the liver. Bone The child’s presentation seems most con-
marrow studies are negative. Tumor pathol- sistent with OMAS. As mentioned, timely
ogy is consistent with neuroblastoma with diagnosis and treatment of the underlying
a high DNA index and low MKI. The tumor neuroblastoma can minimize long-term
is not N-myc amplified and otherwise has a sequelae in regard to cerebellar damage. As
favorable genetic profile. with Case 1, the patient should undergo rou-
d. What is the patient’s stage? What are tine laboratory workup in addition to urine
the next steps? HVA, VMA, and serum NSE. Considering
Fortunately, for young children with lim- that many of the patients with OMAS may
ited extent of disease, the large majority of have early stage disease, these laboratory
Neuroblastoma 251

markers may be normal (often NSE is ele- autoantibody. There is no standardized


vated with normal HVA and VMA). Since treatment for OMAS, though many centers
there are no focal findings on clinical exam, use dexamethasone (due to its enhanced
screening abdominal ultrasound would be a ability to cross the blood–brain barrier com-
reasonable next step, though likely the pared with prednisone) in combination
patient will need 123I-MIBG scan for defini- with IVIG and cyclophosphamide. Steroids
tive diagnosis. lead to nonspecific immunosuppression
Workup shows localized uptake along while cyclophosphamide inhibits T-cell
the cervical spine on 123I-MIBG scan with an replication. IVIG helps to overwhelm the
elevated NSE and otherwise unremarkable antibody response (similar to immune
workup. thrombocytopenic purpura, ITP), though
b. What are the next steps? this action is transient. Patients who fail to
In the case of localized disease, surgical improve or relapse after this initial therapy
resection should be undertaken if safe and may benefit from treatment with rituxi-
feasible. Local lymph node resection should mab (CD20 monoclonal antibody). Most
be done to assess appropriate disease stage. patients will need monitoring and therapy
The patient undergoes surgical resection to ensure the best possible speech and
which achieves a gross total resection. Local motor development.
lymph nodes are negative for disease involve-
ment. Concomitant bone marrow studies
Suggested reading
are negative. Tumor pathology shows a low-
stage neuroblastoma without N-myc ampli- Davidoff, A.M. (2012). Neuroblastoma. Semin.
fication. The patient continues to have Pediatr. Surg. 21: 2–14.
concerning symptoms postsurgical includ- Maris, J.M. (2010). Recent advances in neuroblas-
ing the aforementioned irritability, motor toma. N. Engl. J. Med. 362: 2202–2211.
and speech dysfunction, and opsoclonus. Park, J.R., Eggert, A., and Caron, H. (2010).
c. Is this expected? What should be Neuroblastoma: biology, prognosis, and treat-
done? ment. Hematol. Oncol. Clin. N. Am. 24:
OMAS occurs likely secondary to antibody 65–86.
production directed towards neuroblas- Pinto, N.R., Applebaum, M.A., Volchenbourn,
S.L. et al. (2015). Advances in risk classifica-
toma cells which cross-react with cerebellar
tion and treatment strategies for neuroblas-
tissue. This autoantibody therefore contin-
toma. J. Clin. Oncol. 33: 3008–3017.
ues to cause cerebellar destruction even Yu, A.L., Gilman, A.L., Ozkaynak, M.F. et al.
after tumor resection. The goal of therapy (2010). Anti‐GD2 antibody with GM‐CSF,
for OMAS is immunosuppression to hope- interleukin‐2, and isotretinoin for neuroblas-
fully suppress and ultimately e­ liminate this toma. N. Engl. J. Med. 363: 1324–1334.
20 Sarcomas of the Soft
Tissues and Bone
mutations and may benefit from targeted
Soft tissue sarcomas
therapy with tyrosine kinase inhibitors.
●● Fibrosarcoma: infantile and adult types.
Sarcomas comprise a heterogeneous group of
●● Fibromyxoid sarcoma: slow‐growing,
malignant tumors that arise in the soft tissues
affects primarily young adults.
or bone. Soft tissue sarcomas (STS) are
●● Leiomyosarcoma: smooth muscle tumor
derived from primitive mesenchymal cells
often linked to Epstein–Barr virus or HIV;
such as muscle, connective tissue (tendons
also seen as a second cancer in retinoblas­
and synovial tissue), supportive tissue (fat and
toma survivors.
nerves), and vascular tissue (lymph and blood
●● Malignant peripheral nerve sheath tumors:
vessels). Rhabdomyosarcoma (RMS) is the
often linked with neurofibromatosis type 1
most common STS in younger children, com­
(NF‐1), may be low‐grade or high‐grade.
prising more than half of these tumors in chil­
●● Synovial sarcoma: aggressive tumor,
dren up to 9 years of age. RMS is the third
which often forms in tissues around joints.
most common solid tumor in children with
●● Angiosarcoma: aggressive tumor that
approximately 250–400 new cases diagnosed
forms in blood vessels or lymph vessels.
per year in the United States. Other nonrhab­
●● Alveolar soft part sarcoma: more commonly
domyosarcoma soft tissue sarcomas (NRSTS)
seen in teens and young adults and most often
that occur in children and teenagers are:
affects orbit, head, neck, and extremities.
●● Liposarcoma: a rare cancer of the fat cells,
●● Undifferentiated sarcoma: sarcoma that
often low‐grade (i.e., locally grow and
does not fit into the abovementioned cate­
spread slowly) and amenable to surgical
gories (among others) due to lack of clarity
resection.
in malignant cell of origin.
●● Desmoid tumors: also known as aggressive
STS account for approximately 6–7% of
fibromatosis, can be seen in association with
cancer cases in children and adolescents less
adenomatous polyposis coli (APC) gene
than 19 years of age (annual incidence 11 cases
mutations in familial adenomatous polyposis
per million in the United States). The relative
(FAP), low‐grade, but often recurs locally
incidence of various STS varies by age, sex, and
(requires large resection margins). May
race. For instance, NRSTS such as fibrosar­
respond to hormone therapy, nonsteroidal
coma and malignant hemangiopericytoma are
anti‐inflammatory drugs or targeted therapy
more common in infants (less than 1 year of
(i.e., tyrosine kinase inhibitors).
age). NRSTS also account for the majority of
●● Dermatofibrosarcoma protuberans: rare
STS in patients over 10 years of age.
tumor in deep tissues, often have ALK

Handbook of Pediatric Hematology and Oncology – Children’s Hospital & Research Center Oakland,
Third Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
© 2021 John Wiley & Sons Ltd. Published 2021 by John Wiley & Sons Ltd.
254 Chapter 20

Genetics loss of 11p15.5 is associated with multiple


The majority of soft tissue and bone sarco­ oncogenes and potential loss of function
mas develop without a predisposing genetic mutations (including p53). In general,
risk factor, although certain diseases and patients with ERMS have a better prognosis
inherited disorders can increase the risk of than patients with ARMS, in large part due
developing childhood STS. These include to the increased risk of metastatic disease at
NF‐1 (primarily peripheral nerve sheath presentation with ARMS. DNA content or
tumors), Beckwith‐Wiedemann syndrome, ploidy has also been implicated in prognosis
Costello syndrome, Li–Fraumeni familial with hyperdiploid tumors (≤51 chromo­
cancer syndrome, FAP, Werner syndrome, somes) having a better outcome. NRSTS can
SMARCB1 (IN11) gene changes, retinoblas­ occur in sites of prior radiation, and children
toma 1 (RB1) gene changes, and cardio‐ with human immunodeficiency virus may
facial‐cutaneous syndrome. Germline develop leiomyosarcoma (often in associa­
mutations in the p53 suppressor gene, as in tion with Epstein–Barr virus).
Li–Fraumeni syndrome, are associated with
early onset breast cancer, sarcomas, brain
tumors, and adrenocortical tumors in fam­ Clinical presentation
ily members. There may be an association STS may occur anywhere in the body and
with parental use of marijuana and cocaine, are not limited to muscle or connective
as well as maternal first‐trimester exposure tissues. The most common sites for RMS are
to radiation, including diagnostic radio­ head and neck (35%), genitourinary system
graphs. Environmental triggers such as ion­ (26%), and extremities (20%). Younger
izing radiation may lead to STS in genetically children less than 10 years of age tend to
susceptible individuals. develop ERMS in head and neck or
Genetic information may contribute to genitourinary locations, whereas adolescents
the diagnostic and treatment decision‐mak­ tend to develop extremity, truncal, or
ing process, especially in some cases in paratesticular ARMS. A majority of RMS are
which cytochemical subtyping may be localized, and, of the 15–20% of cases with
equivocal. Approximately 90% of alveolar metastatic disease at presentation, most
RMS (ARMS) harbor the characteristic involve the lungs, bone marrow, lymph
translocation of the FOXO1 gene (previously nodes, or bone.
FKHR gene) at 13q14 with PAX3 at 2q35 As RMS can arise anywhere in the body,
(t(2;13)(q35;q14)) or less commonly (25% of the presenting symptoms are highly variable.
cases with the fusion gene) PAX7 at 1p36 Typically, RMS presents as a painless growing
(t(1;13)(p36;q15)), leading to a fusion tran­ mass. Depending on location, adjacent struc­
scription factor that inappropriately activates tures may be compressed and lead to symp­
gene transcription and resultant tumorigenic toms such as airway obstruction or cranial
behavior. This FOXO1–PAX3 fusion prod­ nerve findings with nasopharyngeal tumors,
uct leading to classic ARMS may be a poten­ orbital swelling and proptosis with orbital
tial target in development of future therapies. tumors, cranial nerve dysfunction, sinusitis,
Approximately 25% of patients with ARMS headaches, ear or facial pain with paramenin­
lack a FOXO1–PAX translocation and tend geal tumors, and urinary obstruction with
to behave clinically more like embryonal bladder or prostate tumors. Prevalence of
RMS (ERMS). ERMS display a loss of hete­ head and neck tumors in younger children
rozygosity in the short arm of chromosome often leads to earlier diagnosis given their
11, most commonly at 11p15.5. This allelic more apparent signs and symptoms.
Sarcomas of the Soft Tissues and Bone 255

Approximately 15–20% of patients with ticular disease, and spine MRI in patients
RMS present with metastatic disease at with evidence of medullary compression.
diagnosis, most commonly in the bone Fluorine‐18‐fluorodeoxyglucose positron
marrow or lung. Other potential sites emission tomography (FDG‐PET) imaging
include the lymph nodes and bone. NRSTS may be helpful to determine initial extent of
rarely present with metastases at diagnosis disease and to monitor response to therapy.
with the most common site being the lungs. Patients with lesions in the maxilla or man­
dible should have a Panorex radiograph to
Diagnostic evaluation evaluate extent of local infiltration. Patient
Patients presenting with an enlarging mass outcomes are strongly tied to the extent of
suspected to be malignancy should have an disease, so all attempts should be made to
open surgical biopsy for confirmation of identify completely the sites and extent of
diagnosis. Sufficient tissue should be disease extension.
obtained for pathologic evaluation and stain­ ●● Bilateral bone marrow aspirate and biopsy

ing in addition to cytogenetic and molecular should be performed, especially in patients


biologic studies. Patients may be eligible to with metastatic disease or alveolar histologic
participate in a clinical therapeutic or bio­ subtype of RMS.
logic trial that also mandates submission of ●● Lumbar puncture in patients with para­

tissue. A complete assessment of extent of meningeal head and neck primary lesions to
disease with meticulous attention to sites of evaluate for metastatic disease.
metastatic disease is mandated prior to initi­
ating definitive therapy. Some protocols also Pathologic diagnosis
mandate biopsy of regional nodes or a senti­ RMS is classified pathologically as embryonal
nel node in patients with primary extremity (ERMS) with botryoid and spindle cell
tumors. These evaluations include: variants, alveolar (ARMS) or anaplastic.
­
●● Complete history and physical examina­ Inves­tigation for particular translocations (see
tion with careful assessment of the mass, Genetics section above) should be done to
adjacent structures, and regional lymph assist with definitive tissue diagnosis in com­
nodes. bination with histochemical studies. STS are
●● Laboratory studies including complete one of the small round blue cell tumors of
blood count, complete metabolic panel with childhood, based on the H&E stain appear­
renal and liver function studies, coagulation ance. Tissue diagnosis is important and ample
testing, and urinalysis. material needs to be provided (typically by
●● Radiographic studies including magnetic open biopsy or tumor resection). RMS may
resonance imaging (MRI) or computed appear similar to striated muscle, and immu­
tomography (CT) scan of the primary lesion, nohistochemical staining for muscle‐specific
depending on the location and regions of proteins is done with myosin, muscle‐specific
draining lymph nodes. Chest CT and radio­ actin, desmin, D‐myosin, and myoD1. ERMS
isotope bone scan should also be conducted accounts for approximately 60–70% of cases
to evaluate for potential lung metastases and of RMS and typically evolves from mucosal
bone metastases, respectively. Further site‐ lined organs, primarily in very young children
specific studies may include: brain MRI for <1 year of age. The appearance of early primi­
parameningeal head and neck tumors, ultra­ tive cells is classic for the botryoid variant.
sound and cystourethroscopy in bladder or The spindle variant is rare and resembles
prostate tumors, abdominal MRI or CT to smooth muscle tissues, and is most com­
evaluate lymph node involvement in parates­ monly seen in paratesticular tumors (and is
256 Chapter 20

associated with an excellent prognosis with completely resected (groups I and II)
>95% overall survival at 5 years). The alveolar nonmetastatic unfavorable site ERMS.
variant (ARMS) is seen in approximately Intermediate‐risk group: patients with
20–25% of RMS and is more commonly seen any nonmetastatic ARMS and all ERMS
in teens and young adults. Histologically, the with unfavorable sites (Stage 2 or 3) that
cells appear as densely arranged round cells in have been incompletely resected (group III).
alveolar patterns, resembling lung alveoli. High‐risk group: patients with metastatic
They tend to be in extremity locations and act tumors, both ARMS and ERMS (a subset of
very aggressively. Anaplastic, or pleomorphic, patients age 10 years and older with ARMS
variants have highly anaplastic large cells with and regional node involvement may do bet­
lobate hyperchromatic nuclei and numerous ter with treatment per the high‐risk group).
multipolar mitotic figures, extremely poor NRSTS are generally staged using the
differentiation, and increased heterogeneity. same criteria mentioned in the preceding
Areas of anaplasia can be seen diffusely or text. The major determinants of outcome
localized in tissue samples and confer a worse include tumor grade, size, extent of initial
prognosis. resection, and presence of metastatic
disease. Tumor size and extent of resection
Staging and classification are strongly correlated with treatment
RMS are stratified into clinical groups based stratification and prognosis.
on extent of surgical resection. Staging is also
done and takes into account the size and Treatment
location of the tumor, spread to local or A multidisciplinary approach to the
regional lymph nodes, and distant (meta­ treatment of STS is crucial. Basic principles
static) disease. The principal determinants of of therapy include systemic treatment of
prognosis are primary site, tumor size, histo­ micrometastatic disease with adjuvant
logic subtype, degree of regional spread and chemotherapy and aggressive local control
nodal involvement, distant metastatic dis­ with definitive surgery and addition of
ease, and extent of prechemotherapy tumor radiation as necessary. Some of the less
resection. Risk stratification is essential to common forms of STS may be treated with
tailoring therapy to maximize outcome. A surgery alone.
unique aspect of the RMS staging is the post­ RMS is often in sites not amenable to
operative clinical grouping system that is gross total resection with negative margins
based on extent of surgical resection and without morbidity or loss of function (e.g.,
results of lymph node evaluation. Risk group orbit, genitourinary, and parameningeal
classification is then based on pretreatment sites). Survival is <20% for patients who do
stage, postoperative clinical group, and his­ not have resection with negative margins.
tology. Favorable sites include the orbit, gen­ Fortunately, RMS is a highly chemosensitive
itourinary (nonbladder nonprostate), and cancer and about 80% are expected to
nonparameningeal head and neck locations. respond favorably. Local control is achieved
Unfavorable sites are bladder/prostate, with either complete surgical resection
extremity, cranial parameningeal, trunk, and (with negative margins) or surgery and
retroperitoneum. Risk grouping is as men­ radiation (if microscopic or gross residual
tioned in the following text: disease remains after surgical intervention).
Low‐risk group: patients with nonmeta­ Primary re‐excision may be indicated prior
static favorable site ERMS (Stage I, regard­ to initiation of chemotherapy in certain
less of degree of initial resection) and situations such as: if the diagnosis was not
Sarcomas of the Soft Tissues and Bone 257

suspected and only a biopsy was performed; domly received this maintenance with
gross or microscopic residual disease is pending results). Recent trials have not been
amenable to wide excision without undue able to show improvement in survival in
morbidity; or if there is uncertainty about patients with metastatic rhabdomyosar­
residual disease or margins which would coma. High‐dose chemotherapy with hemat­
negatively impact survival. Sentinel lymph opoietic stem cell rescue has not shown
node biopsy should be attempted in all benefit in patients with high‐risk and meta­
cases. static disease.
STS are moderately sensitive to radiation
therapy (RMS more sensitive than NRSTS), Prognosis
and this treatment modality is critical for Overall, survival without relapse is >70% in
tumors that cannot be fully excised children and adolescents 5 years from
surgically with negative margins. Treatment diagnosis. Low‐risk groups, representing
volume is determined by the extent of the approximately 30% of RMS, can be expected
tumor prior to surgical resection, and the to have an excellent outcome with long‐term
margin is influenced by the surrounding survival >90%. Of the 55% of patients with
normal tissue and vital structures. Intensity‐ intermediate‐risk disease, 5‐year overall
modulated radiation therapy (IMRT) or survival is approximately 55–65%. In
proton beam radiation may offer advantages patients with metastatic alveolar disease
to sparing surrounding critical tissues. (15–20% of the population), survival is
Optimal timing of radiation therapy remains poor, at less than 20%. Several caveats are
unclear, and local control may be given after present that help determine prognosis:
4–20 weeks of systemic chemotherapy. ●● Site of the primary tumor has a major

Earlier definitive therapy may be indicated impact on survival and is associated with
in patients with intracranial primary tumors pathologic subtypes, ease of surgical resect­
or in those with compromise of function ability, timing to presentation, and involve­
that may be debilitating or life‐threatening. ment of regional nodes. Favorable sites
Chemotherapy provides the backbone of include nonparameningeal head and neck
treatment for patients with RMS and is initi­ tumors, nonbladder, nonprostate genitouri­
ated following the initial surgical procedure. nary tumors or those affecting the biliary
After definitive local control, chemotherapy tract.
continues due to the risk of microscopic ●● Extent of disease is the most significant

residual or metastatic disease. The most predictive factor for survival. Children with
active drugs in the treatment of RMS remain localized, completely resected disease do
vincristine, dactinomycin, and cyclophos­ better than those with widespread or dis­
phamide (VAC). Although multiple addi­ seminated disease.
tional treatment regimens have been ●● Patients with smaller tumors (<5 cm) have

attempted to decrease alkylator exposure, improved survival compared to children


VAC remains the standard of therapy, often with tumors >5 cm.
with the addition or substitution of doxoru­ ●● The alveolar subtype of RMS has an

bicin, ifosfamide, etoposide, and irinotecan adverse prognosis and is often associated
depending on the underlying risk group. with an aggressive clinical course and meta­
Recent European trials have shown benefit static disease at diagnosis and relapse.
with a maintenance phase after intensive ●● Patients between 1 and 9 years of age have

chemotherapy for patients with intermedi­ a better prognosis compared to infants and
ate‐risk RMS (high‐risk patients nonran­ older children.
258 Chapter 20

Children with recurrent RMS have a dis­ but there continues to be a paucity of new
mal prognosis with long‐term survival of potential therapeutic agents.
<15%, particularly if the disease recurs in a
metastatic site or area of prior irradiation.
The majority of relapses occur within 3 years Bone sarcomas
of therapy completion. Metastatic recur­
rence is essentially incurable, though treat­ Primary malignant bone tumors in children
ment may offer palliation. Treatment with constitute approximately 6% of all childhood
surgical resection and adjuvant multiagent malignancies. The two most common bone
chemotherapy can be considered with drug tumors are osteosarcoma (OS) and Ewing
combinations such as ifosfamide/carbopl­ sarcoma family of tumors (ESFT), which
atin/etoposide, docetaxel/gemcitabine, and include Ewing sarcoma, extraosseous Ewing
irinotecan in combination with temozolo­ sarcoma, primitive neuroectodermal tumor
mide or vinorelbine. Durable remissions for (PNET), and Askin tumors (i.e., chest wall
several years may be obtained with aggres­ and pulmonary area). Other rare malignant
sive local retreatment and systemic therapy. bone tumors include chondrosarcoma and
High‐dose chemotherapy followed by non‐Hodgkin lymphoma of bone. Bone
hematopoietic stem cell rescue has not been lesions may also represent Langerhans cell
shown to be advantageous in this group. histiocytosis, benign tumors, or metastatic
disease. Osteosarcoma arises from primitive
Nonrhabdomyomatous soft cells of mesenchymal origin (thus a sarcoma)
tissue sarcomas and exhibits osteoblastic differentiation
Few clinical trials and prospective studies producing malignant osteoid. OS is an
have been conducted in this population of aggressive cancerous tumor of bone with a
children. Surgery is the mainstay of effective bimodal age distribution peaking in early
therapy and every effort should be made to adolescence and in adults >60 years of age.
completely excise the tumor with clear tumor ESFT are malignant tumors of bone and soft
margins. Patients with completely excised tissue thought to derive from neural crest
low‐grade tumors, or high‐grade tumors cells and harboring the Ewing sarcoma
<5 cm, have a favorable outcome with sur­ translocation. Eighty percent of ESFT arise
vival exceeding 85%. Patients with high‐ in children <20 years of age and have a racial
grade NRSTS larger than 5 cm and those predilection with rare presentation in
with unresectable, localized disease have an African‐American and Asian ethnicities.
intermediate prognosis with approximately
50% survival. Patients with high‐grade Genetics
tumors and positive tumor resection mar­ Osteosarcoma is characterized by complex
gins typically receive adjuvant radiation unbalanced karyotypes with alterations in
therapy. It is not clear if adjuvant chemother­ the Rb and p53 tumor suppressor pathways,
apy confers a survival advantage in this linking this tumor to retinoblastoma and Li–
group of patients. Radiation is considered in Fraumeni syndrome, respectively. Other
unresectable tumors or if tumor shrinkage genetic associations include fibrous dyspla­
may lead to complete resection. Patients with sia, enchondromatosis, hereditary multiple
metastatic NRSTS have a dismal long‐term exostosis, and Rothmund–Thomson syn­
prognosis and fewer than 20% survive 5 drome (AR‐associated congenital bone
years from diagnosis. Neoadjuvant chemo‐ defects; teeth, hair, and skin dysplasia; hypo­
radiotherapy is being explored in this group, gonadism and cataracts). Osteosarcoma has
Sarcomas of the Soft Tissues and Bone 259

long been thought to be associated with ado­ originating around the knee. Approximately
lescent growth spurts and the higher inci­ 10% of patients have primary tumors in the
dence in large dog breeds and taller people axial skeleton including the pelvis, and
supports this correlation. Girls have a peak 15–20% present with metastatic disease
incidence younger than boys (12 vs. 16 years) (lung, bone, lymph nodes, and rarely brain).
correlating with the different average age for Most ESFT occur in bones and their loca­
pubertal onset. Well‐documented risk fac­ tions tend to differ from that of OS. Flat
tors include radiation exposure, Paget’s dis­ bones of the axial skeleton are more com­
ease, and other disorders associated with monly affected; in long bones, the diaphyseal
increased bone turnover. Soft tissue and portion is usually involved. The most com­
bone sarcomas also occur as secondary mon primary locations include the pelvic
malignancies. bones, the long bones of the lower extremi­
Unlike OS, ESFT are not associated with ties, and the bones of the chest wall. Metastatic
familial cancer syndromes and do not appear disease is present in 25% of patients at diag­
to increase in risk following radiation expo­ nosis and is primarily located in the lungs,
sure. The translocation t(11;22)(q24;q12) is bones, or bone marrow. Site of primary dis­
present in >95% of ESFT and some consider ease influences the relative incidence of
this finding to be pathognomonic and suffi­ metastases at diagnosis; central primaries are
cient to confirm the diagnosis. This translo­ associated more frequently with distant dis­
cation results in the EWS–FLI1 fusion ease (40%), whereas distal primary lesions
transcript that acts as an aberrant transcrip­ have the lowest incidence (15%).
tion factor altering tumor suppressor gene
pathways. Diagnostic evaluation
The diagnostic evaluation should determine
Clinical presentation site and extent of disease, including presence
Patients typically present with a growing and location of metastatic disease. The
mass or swelling in the involved area and requisite elements of evaluation include:
pain, with symptoms preceding diagnosis ●● A complete history and physical
often by several months. Patients frequently examination.
attribute the pain to a minor trauma and ●● Routine laboratory studies include a com­

indeed may present with a pathologic frac­ plete blood count, complete metabolic panel
ture in the affected weakened bone. A palpa­ with renal and liver function tests, serum
ble mass or swelling of the involved site creatinine, creatinine clearance, and urinal­
typically arises after the onset of pain. ysis. A measured or calculated glomerular
Systemic symptoms such as weight loss or filtration rate should be obtained prior to
shortness of breath may be late sequelae and initiation of nephrotoxic chemotherapy. The
secondary to metastatic disease. Fever is a alkaline phosphatase may be elevated and
common symptom of ESFT (20%) and may has been associated with an inferior out­
lead to confusion with osteomyelitis result­ come in OS. The serum lactate dehydroge­
ing in delayed diagnosis. nase may also be elevated and may correlate
Osteosarcoma may occur in any bone with tumor burden.
but primarily occurs in the metaphyses of ●● Imaging studies should begin with plain

the most rapidly growing bones. The most radiographs to visualize osseous changes and
common primary sites of origin are the confirm suspicion of a malignant tumor.
distal femur, proximal tibia, and proximal Osteosarcoma usually produces dense scle­
humerus with approximately 50% of tumors rosis in the metaphyses of long bones with
260 Chapter 20

soft tissue extension seen in 75% of tumors, perform the biopsy, so that the biopsy tract
radiating calcifications (sunburst pattern) in can be excised en bloc with the planned
60% of tumors, osteosclerotic lesions in 45% surgical resection. Sufficient tissue should
of cases, lytic lesions in 30% of cases, and be obtained for diagnostic histopathology as
mixed lesions in 25% of cases. A triangular well as for any submissions required for
area of periosteal calcification in the border participation in a clinical trial. Biopsy may
region of the tumor and healthy tissue is be taken from an extraosseous component,
known as a Codman triangle. ESFT are if present, to prevent pathologic fracture.
described as lytic with an “onion peel” peri­ A characteristic feature of OS is
osteal reaction and typically occur in the dia­ malignant osteoid (bone formation) in the
physis or flat bones. Patients with soft tissue tumor. This may appear pleomorphic
ESFT only may have normal radiographs. (anaplastic), and giant atypical mitotic
Additional assessment of the primary tumor features are often present. Tumors are sub‐
site with MRI should be undertaken to view classified by resemblance to bone, cartilage,
the soft tissue component, surrounding or fibroblast cells. The three major subtypes
structures, vessels and nerves, and the are: osteoblastic, chondroblastic, and
intramedullary extension to assist with sur­ fibroblastic, reflecting the predominant type
gical planning. MRI should include the of matrix in the tumor. Histologic variants
entire involved bone and neighboring joints include telangiectatic, small cell, periosteal,
to evaluate for skip lesions. Metastatic dis­ and parosteal.
ease is typically in the lungs or bone and To differentiate ESFT from other small
therefore imaging with chest CT and techne­ round blue cell tumors of childhood, an
tium‐99m bone scan is essential in the initial expanded immunohistochemical panel is
workup for staging. FDG‐PET imaging is a done to include detection of CD99 (a cell sur­
sensitive screening tool for the detection of face glycoprotein with strong expression in
bone metastases in ESFT, but its role in eval­ ESFT) with a characteristic honeycomb
uation of OS has yet to be determined. staining pattern. Though CD99 may be pre­
●● Bilateral bone marrow aspirate and biopsy sent in other tumors (including Wilms tumor,
should be performed in patients with ESFT. clear cell sarcoma of the kidney, T‐cell lym­
●● Baseline audiogram and echocardiogram phoma and leukemia, and rhabdomyosar­
should be obtained prior to initiation of oto­ coma), the pattern of staining in ESFT is
toxic and cardiotoxic chemotherapies, membranous, distinct from the cytoplasmic
respectively. pattern present in these other tumors.
●● Fertility preservation should be discussed Molecular genetic studies using fluorescent
with patients prior to chemotherapy. Sperm in situ hybridization or reverse transcriptase‐
banking should be offered as well as oocyte polymerase chain reaction are valuable in
cryopreservation. Age, pubertal status, and diagnosis, specifically for the detection of
time may determine feasibility. characteristic translocations that allow for
definitive diagnosis of ESFT, RMS, and syno­
Pathologic diagnosis vial cell sarcoma.
Patients presenting with a bone mass
suspicious for malignancy should undergo a Treatment
diagnostic incisional or core biopsy. It is Osteosarcoma
imperative that an experienced oncologic Systemic multiagent chemotherapy prior to
orthopedic surgeon who will also be and following definitive radical surgery is the
performing the definitive procedure standard of care for treatment of OS. Complete
Sarcomas of the Soft Tissues and Bone 261

en bloc resection of the primary site, as well as agent chemotherapy. The degree of tumor
all sites of metastatic disease, is critical to necrosis, as determined post definitive surgi­
long‐term survival. The ability to perform cal resection, is a significant independent
such procedures is affected by tumor location, predictor of survival, both event(relapse)‐
neurovascular involvement, ability for wide free and overall. Those good responders
resection with a normal margin of muscle/tis­ whose tumors show ≥90% necrosis have
sue, and the ability to perform functional superior outcomes compared to those with
reconstruction. Preoperative (neoadjuvant) poor (<90% necrosis) response.
chemotherapy following initial biopsy offers Radiation therapy has limited utility in
several advantages which include a decrease patients with OS, but may play a role in
in tumor‐related edema, shrinkage of the pri­ those unable to undergo complete resection.
mary tumor, treatment of presumed micro­ Patients presenting with metastatic dis­
metastases, and assessment of sensitivity ease undergo the same therapeutic approach
(histologic response). The orthopedic onco­ with systemic chemotherapy and definitive
logic surgeon determines the type of surgical surgery of the primary site in addition to
procedure performed, and factors used in this surgery of metastatic sites, as feasible.
determination include the location and size of Approximately one‐third of OS patients with
the tumor, patient age and skeletal maturity, pulmonary metastases will have additional
presence of metastatic disease, and patient microscopic pulmonary nodules not visible
lifestyle choices. Limb‐sparing procedures are on the current, highest resolution CT imag­
feasible in the great majority of patients and ing. It is recommended that pulmonary nod­
the development of expandable endoprosthe­ ules identified on CT imaging be surgically
ses has allowed the use of these techniques in resected.
younger children. Tumors of the pelvis or
axial skeleton present a difficult situation, as Ewing sarcoma family of tumors
resection with adequate margins may not be As with OS, cure in ESFT can be achieved
possible. only with a multimodal approach, using sur­
Chemotherapy is the backbone of treat­ gery and/or radiation therapy for local con­
ment for OS, with the exception of the com­ trol of the primary lesion and chemotherapy
pletely resected paraosteal subtype (often for eradication of subclinical micrometasta­
cured with radical surgery alone). Without ses. Definitive surgery follows a period of
adjuvant chemotherapy, more than 80% of induction chemotherapy and many patients
patients will recur, often with metastatic dis­ are candidates for complete surgical resec­
ease. Standard components of multiagent tion with limb‐salvage procedures. Induction
chemotherapy include high‐dose metho­ chemotherapy may also render initially
trexate, doxorubicin (Adriamycin), and cis­ unresectable tumors resectable. ESFT are
platin, referred to as MAP. The addition of radiation‐responsive, although typically this
dexrazoxane, a cardioprotectant, is now therapeutic modality is reserved for surgi­
commonplace in patients with OS receiving cally unresectable tumors or in tumors with
MAP therapy. Ifosfamide and etoposide are positive margins after resection. Patients
additional agents shown to have activity but who receive radiation therapy as the only
not beneficial in the upfront management of local control modality have an inferior out­
OS. Treatment cycles are given for approxi­ come. Frequently, these patients have other
mately 30–40 weeks with a definitive surgical adverse features including large tumor size,
procedure performed at approximately week unfavorable locations (e.g., vertebral
11–12, followed by continuation of multi­ tumors), or both. Radiation is also utilized in
262 Chapter 20

the treatment of metastatic disease. metastasis at diagnosis and poor response to


Debulking procedures have not been shown neoadjuvant chemotherapy, other factors
to improve outcome and should not be part associated with a poor prognosis include:
of local control therapy. incomplete resection, axial skeletal prima­
Systemic chemotherapy has significantly ries, larger tumor size, and proximal location
improved outcomes in ESFT with rapid on the limb. To date, intensified treatment
reduction in tumor volume facilitating suc­ for those with less‐favorable tumor necrosis
cessful complete surgical resections or allow­ has not improved outcome.
ing for limb‐sparing procedures. Active The 5‐year overall survival rate in
chemotherapy agents include vincristine, patients with ESFT treated upfront with
cyclophosphamide, and doxorubicin (VDC) chemotherapy, surgery, and radiation is
alternating with ifosfamide and etoposide approximately 60–70%. As with OS, those
(IE). As with OS, definitive local control presenting with evidence of metastatic
occurs at week 12 followed by additional disease have a dismal expected survival of
chemotherapy. Compressed interval, every 2 10–30%, though patients with lung
weeks with the use of filgrastim, rather than metastasis alone may have a survival benefit.
3‐week cycles, has led to superior outcomes The most significant adverse prognostic
and is now standard of care in ESFT, though factors are: older age, axial skeletal tumor
this increased time–dose intensity schedule location, larger tumor size, and elevated
has shown benefit in pediatric patients only. serum LDH at diagnosis.
Outcomes of patients with metastatic The prognosis for patients with recur­
ESFT are dismal, related to site of metastasis, rence is quite poor. Most local recurrences
and with tumors that may have more chemo­ are associated with concomitant distant dis­
therapy resistance. High‐dose chemotherapy ease. Time to relapse and tumor burden cor­
with autologous stem cell rescue is being relate with post‐relapse outcome. Late
investigated, though has not shown clear recurrence (i.e., >5–10 years) has been
benefit in those with metastatic disease. reported in bone sarcoma. Surgical resection
remains the mainstay of therapy for curative
Prognosis second‐line therapy. Patients with OS who
Most patients with bone sarcomas have develop pulmonary lesions after completion
micrometastatic disease at diagnosis as evi­ of therapy can be cured with surgery alone.
denced by a high percentage of patients who The role of adjuvant chemotherapy in relapse
develop lung metastases following local con­ is controversial, though patients are often
trol therapy (i.e., surgery and/or radiation) offered such treatments including cyclo­
alone. Prior to the use of systemic chemo­ phosphamide/topotecan, irinotecan/temo­
therapy, survival rates at 2 years were zolomide, and gemcitabine/taxotere.
15–20%. Currently, patients with localized Radiation has a role in the treatment of
disease can expect a long‐term survival of patients with ESFT who develop new meta­
>70%. Survival in patients with metastatic static sites and for palliation and pain control
disease remains dismal, estimated at 2 years in either OS or ESFT.
to be 10–30%. The degree of tumor necrosis Targeted and biologically based therapies
following neoadjuvant (pre‐surgery) chemo­ are being investigated in patients with poor
therapy is an independent predictor of response to initial chemotherapy, those with
event‐free and overall survival, presumably metastatic disease, and those with recurrent
reflecting superior tumor sensitivity to disease. A range of mutations associated
chemotherapy. In addition to the presence of with OS do not provide obvious therapeutic
Sarcomas of the Soft Tissues and Bone 263

targets, as they primarily reflect loss of For OS patients, contemporary orthope­


tumor suppressor genes (i.e., p53, Rb1, dic surgery techniques are aimed at preser­
PTEN) rather than activation of targetable vation of function and improved limb
oncogenes. Recent novel therapies for OS salvage without compromising survival.
including receptor activator of nuclear fac­ Amputation may result in superior limb
tor kappa beta ligand (RANKL), antibody function in some cases. In a skeletally imma­
(denosumab), and anti‐GD2 antibody ture child, expandable prostheses may
(dinutuximab with sargramostim) have not require subsequent revisions, though some
shown benefit. Transcription inhibition of may benefit from newer self‐expanding
the EWSR1–FLI1 translocation is an area of prostheses. Late prosthetic failures or infec­
interest in ESFT in addition to utilization of tion can occur, requiring surgical revision or
an antibody to insulin growth factor recep­ delayed amputation. Radiation therapy can
tor‐1 (IGFR‐1; ganitumab). lead to local effects including growth
impairment, muscle fibrosis, and increased
risk of secondary malignancy, in a dose‐
Late effects in soft tissue dependent manner.
and bone sarcomas

Site and extent of disease, as well as therapeu­ Case study for review
tic interventions with chemotherapy, radia­
tion, and surgery all contribute to late sequelae A 9‐year‐old boy presents with a nontender
in sarcoma patients. A majority of first‐line 3 × 4 cm2 mass in the right inguinal region.
protocols utilize high cumulative doses of He denies fever, weight loss, or pain
anthracyclines (i.e., doxorubicin), resulting in elsewhere. The mass has been present and
a lifelong risk of developing cardiomyopathy. growing for several weeks.
Addition of a cardioprotectant, dexrazoxane, a. What key elements should you obtain
is now commonly used to decrease the risk of in the history?
cardiac late effects. For all patients, lifelong Key elements in the history include length
cardiac monitoring with serial echocardio­ of symptoms, possible relationship to infec­
grams and periodic functional assessments is tion (could this be a lymph node?), animal
recommended. Alkylator therapy (ifosfamide exposures and bites, travel, cuts or injuries
and cyclophosphamide) can lead to gonadal to the affected leg, systemic symptoms such
toxicity (primarily in males) and increased as malaise, fevers, unexplained weight loss,
risk of secondary malignancy. Second malig­ or discomfort that limits activities or neces­
nant neoplasms occur in approximately 3% of sitates pain medication.
patients within 10 years of therapy comple­ b. What are you looking for on the phys­
tion, likely related to both a genetic predispo­ ical examination?
sition and treatment with mutagenic The physical examination should be
therapies. Platinum therapy (cisplatin) can ­complete, head to toe. The mass should be
lead to long‐term ototoxicity and nephrotox­ measured and characterized for mobility,
icity. High‐frequency hearing loss is common ­tenderness, warmth and erythema. The mass
and some patients will require hearing aids. may be hard and nontender if malignant
Renal dysfunction is rare but could result in disease is present. If the mass is suspected to
chronic electrolyte wasting due to proximal be a node, it may be enlarged as a result of
tubular damage (see Chapter 30 for more disease spread and therefore the distal
details). extremity must be carefully examined. All
264 Chapter 20

other lymph node chains as well as the liver f. Assuming you find out this is a local­
and spleen should also be carefully assessed. ized undifferentiated sarcoma, what is
c. What is your differential diagnosis the child’s prognosis and what is the gen­
for this boy? eral treatment plan?
The differential diagnosis of a mass in the In this case, the child had a nonpalpable
inguinal area consists of lymphadenopathy mass in the fifth distal toe found on MRI.
secondary to an infectious or malignant eti­ The pathologic diagnosis of undifferenti­
ology. Infections may be bacterial, protozoal, ated sarcoma was made on excisional
fungal, or viral, and may be the result of biopsy of the inguinal mass. No other evi­
introduction by a cut (which may have since dence of distant tumor was found on fur­
healed), animal bite, or scratch. A mass that ther imaging. Prognosis for undifferentiated
is hard, nontender, and nonmobile, without sarcoma is generally similar to ARMS, and
signs and symptoms of infection, is con­ patients with localized disease who receive
cerning for malignancy. Malignancies that systemic chemotherapy and local control
can lead to regional spread in an extremity (complete surgical excision with negative
include sarcomas (bone and soft tissue). margins) can expect to have approximately
d. How do you approach diagnostic a 70% event‐free survival. This is in stark
evaluation and staging? contrast to patients with metastatic disease
Assuming you could find no lump or tender in which case survival is less than 20%
area on the extremity exam, it is prudent to despite aggressive therapy. Those patients
begin a detailed investigation with imaging with regional spread, similar to this case,
and biopsy. Imaging should consist of MRI have an intermediate prognosis. Such cases
of the inguinal area and leg (to the tips of should be treated with multiagent chemo­
the toes). An excisional biopsy should be therapy in addition to local control. In this
performed as well given the concern for child, local control will involve treatment of
malignancy (and to prevent potential tumor the toe and the inguinal mass in addition to
tracking which can occur with an incisional other potential draining nodes.
biopsy). Once the mass is a biopsy‐proven
malignancy, further staging should evaluate
for distant disease and include CT of the
chest in addition to bone scan for OS and Suggested reading
FDG‐PET scan for Ewing sarcoma.
Borenstein, S.C., Steppan, D., Hayasi, A. et al.
e. What do you tell the child and his
(2018). Consensus and controversies regard­
family? ing the treatment of rhabdomyosarcoma.
Prior to initiation of the biopsy and imag­ Pediatr. Blood Cancer 65: e26809.
ing, it is best to be straightforward with the Reed, D.R., Hayasi, M., Wagner, L. et al. (2017).
family and let them know your concern that Treatment pathway of bone sarcoma in chil­
this mass likely represents a malignancy. As dren, adolescents, and young adults. Cancer
the mass is in the inguinal area, it could rep­ 123: 2206–2218.
resent regional spread of a distant tumor.
21 Germ Cell Tumors

Germ cell tumors (GCTs) arise from pri- more frequent in males. Testicular GCTs
mordial cells involved in gametogenesis and have several known risk factors including
occur primarily in the testes and ovaries. infertility/testicular atrophy as well as cryp-
These tumors represent approximately 4% torchidism. 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 sacrococcy- spontaneous descent. However, early cor-
geal region, midline of the brain, mediasti- rection may partially ameliorate this risk.
num, and retroperitoneum. Due to the The incidence of testicular GCTs in the
pluripotentiality of PGCs, GCTs are an array Caucasian population is increasing at a rate
of different histologic subtypes, adding to of 3–6% per year. Perinatal and environ-
the complexity of the disease. In addition, mental risk factors are thought to play a role,
GCTs can occur due to tumorigenesis or as although these are yet to be clearly eluci-
part of normal embryonal development, in dated. Young parental age at birth, low birth
part explaining the pediatric bimodal age of order, low birth weight, and breech birth
distribution (2 and 20 years). In addition, have all been noted to be statistically signifi-
age of presentation and histologic subtype cant risk factors in multiple studies.
varies significantly between males and Maternal exposure to pesticides or hor-
females. This is thought to be due in part to mones, parental smoking, and alcohol con-
the differential timing of male and female sumption may also be factors. Genetic risk
development; female germ cells enter meio- factors also play a role, specifically in tes-
sis at 11–12 weeks of gestation, while male ticular GCT, as a family history of cancer,
germ cells begin meiosis with the onset of especially at a young age, has been associ-
puberty. ated with increased risk in pediatric patients.
Additionally, there is a higher incidence in
monozygotic twins and some familial
Epidemiology ­clusters have been reported. A number of
congenital genitourinary anomalies such as
Due to the differential timing of germ cell retrocaval ureter, bladder diverticulum,
maturation, GCTs are more common in and inguinal hernia have been associated
females until adolescence and then become with increased risk, emphasizing that

Handbook of Pediatric Hematology and Oncology – Children’s Hospital & Research Center Oakland,
Third Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
© 2021 John Wiley & Sons Ltd. Published 2021 by John Wiley & Sons Ltd.
266 Chapter 21

a­ bnormalities in development likely play a and therefore are treated with complete sur-
role. Similarly, disorders of sexual develop- gical resection. Immature teratomas are
ment, especially in cases with excess Y chro- tumors of intermediate malignant potential
mosome material (e.g., gonadal dysgenesis), with less differentiated tissues.
pose an increased risk in both males and Germinomas may be of pure histology
females. Children with Down syndrome, or mixed. Syncytiotrophoblastic cells
who generally have a decreased risk of solid occur in about 5% of germinomas and
tumors, are at a predisposition to develop secrete the beta subunit of human chori-
testicular GCT, whereas patients with onic gonadotropin (β‐HCG). Embryonal
Klinefelter syndrome have an increased risk carcinoma is most often found as a compo-
of mediastinal GCT. Gain of chromosome nent of testicular mixed GCT and is
12p is also seen in the majority of testicular thought to be the driver for the develop-
and malignant ovarian GCT. Approximately ment of other histologic subtypes includ-
half of all pediatric GCT are extragonadal in ing teratoma, yolk sac tumor, and
origin, with about 15% of these being choriocarcinoma. Yolk sac tumor is gener-
malignant. ally found as a part of testicular mixed
GCT but can also be seen as a pure histo-
logic variant in young children. Yolk sac
Pathology and serum tumor tumor secretes α‐fetoprotein (AFP).
markers Choriocarcinoma contains syncytio-
trophoblastic cells and therefore also
The main histologic variants of GCTs secretes β‐HCG. Ovarian cases should be
include germinomas (dysgerminoma in the distinguished from metastatic gestational
ovary and seminoma in the testis), embryo- choriocarcinoma.
nal carcinoma, yolk sac tumor (endodermal In the ovary and testis, GCT should be
sinus tumor), choriocarcinoma, and tera- distinguished from sex cord stromal tumors,
toma (mature and immature). Mixed GCTs which arise from the nongerminative com-
are composed of two or more histologies. ponents of gonadal tissue and include
Teratomas contain tissues from all three tumors involving the Sertoli–Leydig cells of
germ layers (ectoderm, endoderm, and the testis and granulosa cells of the ovary.
mesoderm). Mature teratomas contain no Sarcoma as well as gonadal infiltration by
malignant germ cell elements and are most leukemia and non‐Hodgkin lymphoma
commonly located in the ovary or sacrococ- should also be considered in the differential
cygeal region. The vast majority of female diagnosis of gonadal tumors.
GCTs are teratomas. Teratomas in the pre- Due to the specific histologies that
pubertal male are benign, whereas those secrete AFP and β‐HCG, these tumor mark-
occurring during or after the onset of ers 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 GCT (specifi- Mature teratomas secrete neither marker;
cally embryonal carcinoma). Therefore, ter- therefore, if a serum marker is present in the
atomatous elements in the testis largely patient diagnosed with a mature teratoma,
occur as a component of a mixed GCT. In one must consider that the tumor contains
the female or prepubertal male, these one or more foci of yolk sac tumor if the
tumors are thought to derive from a benign AFP is positive, choriocarcinoma or germi-
germ cell but may rarely develop malignant noma if the β‐HCG is positive, and that the
components (i.e., squamous cell carcinoma) patient in fact has a mixed malignant GCT.
Germ Cell Tumors 267

Due to physiologic changes in the secretion The differential diagnosis of a testicular


of fetal AFP, this marker can be difficult to mass includes hydrocele, hematocele, vari-
utilize in young infants. AFP is made in the cocele, inguinal hernia, and torsion of a
fetal liver and does not decline to adult levels normal testis. Girls most frequently present
until approximately 6–8 months of age. Age‐ with abdominal pain, distention, and
appropriate levels can be obtained from weight gain. Torsion of an involved ovary,
published tables. AFP can also be secreted hemorrhage, or tumor rupture may occur
by regenerating liver postinjury or by cer- and present as an acute surgical abdomen.
tain liver tumors such as hepatoblastoma. Secretion of β‐HCG may lead to isosexual
Degree of AFP elevation may be related to precocity. Occasionally, the increasing
tumor volume. β‐HCG is elevated in preg- abdominal girth and elevated urine and
nancy and can also be seen in association serum β‐HCG have been confused with
with childhood hepatic tumors and other pregnancy.
cancers involving the breast, stomach, and The most common site of extragonadal
pancreas in adults. Nonspecific markers of tumors is the sacrococcygeal region.
GCT include lactate dehydrogenase (LDH) Although usually benign, patients can have
isoenzyme 1, neuron‐specific enolase, and malignant tumors of higher stage. Older age
placental alkaline phosphatase. at diagnosis is associated with a greater risk
of malignancy. Sacrococcygeal tumors are
classified as four anatomic variants: type I
Clinical presentation (predominantly outside the body), type II
(equally inside and outside the body), type
GCT should be considered in children who III (largely intra‐abdominal), and type IV
present with a pelvic, ovarian, or testicular (completely within the body). More than
mass, or a midline mass in the sacrococcy- 90% of tumors present with some compo-
geal region, retroperitoneum, mediastinum, nent of an externally visible mass. These
or pineal or suprasellar regions of the brain. tumors are often detected in utero with pre-
GCTs present with signs and symptoms natal ultrasonography. They may also pre-
related to the site and size of the primary sent later in children with constipation,
tumor. Compression of internal structures urinary obstruction, urgency or frequency,
can lead to pain, constipation, and urinary lower back or pelvic pain, lower extremity
obstruction. Large ovarian tumors can addi- weakness, or sensory changes, all signs and
tionally lead to decreased lung volumes and symptoms due to compression of normal
respiratory distress. GCTs can metastasize, structures by tumor.
most commonly to the lungs, but patients Approximately 20% of children with
rarely present with clinical symptoms refer- GCTs will present with metastatic disease,
able to these metastases. Occasionally, most commonly to the lung, liver, or
patients may present with hormonal mani- lymph nodes, and rarely to the central
festations secondary to secretion of β‐HCG. nervous system, bone, or bone marrow.
Specifically, peripheral conversion of andro- Regional spread resulting in surgically
gen to estrogen can lead to gynecomastia or unresectable tumors occurs in approxi-
uterine bleeding. mately 25% of patients at presentation.
Boys with gonadal GCT typically pre- Tumor dissemination occurs by local
sent with a slowly enlarging, painless tes- extension, intracavitary seeding, or hema-
ticular mass, occasionally complicated by togenous spread. Intracavitary seeding
acute severe pain due to testicular torsion. may involve the omentum, bowel, spleen,
268 Chapter 21

diaphragm, or pelvic organs. Rarely, bone the patient and family prior to radical
involvement can occur by direct extension. 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
●● Physical examination. peritoneal washings (in ovarian tumors),
●● Baseline laboratory studies consisting of a and presence of residual or metastatic dis-
complete blood count, transaminases, blood ease. Factors known to affect prognosis
urea nitrogen, creatinine, and electrolytes. include extreme tumor marker elevation
●● Baseline tumor markers including AFP, and failure of these markers to appropriately
β‐HCG, and LDH. decline following tumor resection, based on
●● Imaging studies to evaluate the extent of the appropriate half‐life for each marker.
the mass and potential metastatic sites: Site of tumor is prognostic, with mediastinal
ultrasound, CT, or MRI of the primary mass tumors fairing more poorly. Histology is
(testicular ultrasound in males, abdominal also important, as pure germinomas are
and pelvic ultrasound, CT, or MRI in generally very chemotherapy‐sensitive and
females); abdominal and pelvic MRI or CT have a high response rate. Patients with
as well as baseline chest radiography with or higher‐stage, metastatic disease also fair
without CT of the chest to evaluate for met- more poorly.
astatic disease (all CTs should be done with In the patient with suspected GCT, every
contrast). effort should be made to perform a com-
plete resection with negative margins. In
Determination of the extent of disease at situations where the patient presents emer-
diagnosis allows proper decision‐making gently with torsion, there may not be time to
regarding the safest and most reasonable wait for a definitive diagnosis prior to sur-
place to biopsy. Localized tumors of the gery; every effort should be made in these
ovary or testis should be completely cases to perform a gross total resection
removed. Resection of ovarian tumors (GTR), assuming there is low operative risk,
should include abdominal exploration and after sending the appropriate tumor mark-
peritoneal washings for cytologic assess- ers. In the case of a large, invasive tumor,
ment. Risks and benefits of lymph node biopsy should be done followed by adminis-
sampling and biopsy of the contralateral tration of chemotherapy and delayed surgi-
testis or ovary should be discussed with cal resection. Due to the location of some
the surgeon, patient, and family. extragonadal tumors, especially those in the
GCTs are known to grow rapidly and mediastinum and retroperitoneum, neoad-
metastasize frequently. Diagnostic studies juvant chemotherapy is often required prior
should be done expeditiously to allow ini- to surgery in order to help facilitate the
tiation of therapy. Due to the potential chance of a safe GTR.
underlying risk of contralateral testicular Depending on tumor location and his-
atrophy, infertility, or carcinoma in situ, tology, some GCTs can be observed after
sperm banking should be discussed with GTR without additional therapy. Young
Germ Cell Tumors 269

patients with extragonadal tumors and a retroperitoneal lymph node dissection may
GTR can generally be observed after sur- also be recommended in patients with
gery, as these tumors rarely have malignant extensive disease at presentation. Patients
elements. Sacrococcygeal tumors must be with a poor response to therapy or recurrent
removed with the coccyx in order to disease may benefit from second‐line thera-
decrease the risk of local recurrence. Young pies such as paclitaxel (Taxol), ifosfamide,
patients are more likely to present with pure cisplatin (TIP); vinblastine, ifosfamide, cis-
yolk sac tumors, and these patients can also platin (VeIP); or gemcitabine/oxaliplatin.
be observed after GTR. Gonadal tumors GCTs are also quite radiosensitive, although
that are histologically consistent with the high cure rates seen with chemotherapy
mature teratoma similarly require only alone have obviated the need for radiation in
observation after GTR. Localized testicular most cases. Radiation may be considered in
seminoma can be observed, although refractory or recurrent disease, or for pallia-
patients with continued elevation of tumor tion. Additionally, high‐dose chemotherapy
markers should undergo adjuvant chemo- with autologous stem cell rescue may be
therapy. Patients with this pure histology are beneficial in a subset of refractory patients.
very chemotherapy‐sensitive and easily sal- Patients treated with high‐dose platinum
vaged in the event of relapse. In general, agents may develop high‐frequency hearing
patients with localized testicular‐mixed loss and require hearing assistive devices.
GCT can also be observed assuming that Additionally, these drugs may lead to tran-
tumor markers normalize after surgery, sient proximal tubule renal dysfunction and
again due to the high rate of salvage with resultant electrolyte wasting. Patients who
potential relapse. Due to this risk of relapse, receive alkylator therapy (e.g., ifosfamide) are
which is directly related to the amount of at risk for secondary malignancy and infertil-
vascular tumor invasion, size of the primary ity (especially in males). Topoisomerase II
tumor, and percent embryonal carcinoma, inhibitors (etoposide) also increase risk for
patients should be presented the options of secondary neoplasms. Bleomycin may lead to
observation versus chemotherapy. Due to pulmonary fibrosis, although the risk is
the rarity of malignant ovarian GCT, recom- poorly quantified.
mendations for chemotherapy versus obser-
vation generally follow those presented for
testicular tumors. Germ cell tumors of the central
Higher‐stage, nonlocalized GCTs are nervous system
treated with chemotherapy and are gener-
ally very chemotherapy‐sensitive. Patients Intracranial GCTs comprise approximately
are treated with bleomycin, etoposide, and 3% of primary childhood brain tumors with
cisplatin (BEP) regimens for three to four a peak incidence between the second and
cycles. These tumors exhibit a steep dose– third decades of life. The majority of these
response curve to platinum compounds, tumors are germinomas, which account for
though the risk of toxicity (ototoxicity and 50–70% of cases. Nongerminomatous germ
nephrotoxicity) must be considered. Due to cell tumors (NGGCT) account for the
the potential risk of long‐term pulmonary remaining third and consist of multiple his-
fibrosis from bleomycin, etoposide/cisplatin tologies including endodermal sinus (yolk
(EP) and etoposide (VP‐16), ifosfamide, and sac) tumors, choriocarcinoma, mature and
cisplatin (VIP) regimens have been devel- immature teratoma, and embryonal carci-
oped as alternative therapies to BEP. Bilateral noma. Most NGGCT are of mixed histology
270 Chapter 21

and may contain germinomatous elements, surgical GTR, as safely feasible. In other
but will not have this as a pure histology. tumor types, the value of a surgical resec-
Mature and immature teratomas predomi- tion has not been clearly delineated and is
nate in the neonatal period. Patients typi- recommended only in specific situations
cally present with symptoms depending on (i.e., persistence of tumor following induc-
the location and size of the tumor, further tion chemotherapy). Pure germinomatous
influenced by the extent of pituitary dys- tumors are highly sensitive to both radio-
function and the presence or absence of therapy and chemotherapy. In the past,
hydrocephalus. Midline pineal tumors are radiotherapy was the modality of choice for
often associated with symptoms related to these tumors due to the high cure rate (5‐
increased intracranial pressure due to year survival >80%). Due to the recognized
obstruction of the cerebral aqueduct and long‐term risks from this therapy (neu-
Parinaud’s syndrome (paralysis of upward ropsychological and endocrine), adjuvant
gaze) due to involvement of the tectal plate. chemotherapy is now being used to allow a
Tumors in the suprasellar area often lead to reduction in the dose and extent of radio-
endocrinopathies such as diabetes insipidus therapy without increasing relapse rate.
(DI) and visual field defects. Occasionally, Chemotherapeutic agents are similar to
DI is related to occult involvement of the those used for systemic GCTs and include
infundibulum. In children with clinical platinum agents (cisplatin and carbopl-
symptoms of DI and consistent MRI find- atin), alkylators (cyclophosphamide and
ings (i.e., absence of a posterior pituitary ifosfamide), and topoisomerase II inhibi-
bright spot, possibly in association with tors (etoposide). Of note, patients with DI
thickening of the pituitary stalk), the differ- should be carefully monitored in an ICU
ential diagnosis should include GCT and setting while receiving alkylators or plati-
Langerhans cell histiocytosis. The presence num therapy due to the increased risk of
or absence of CSF tumor markers may assist nephrotoxicity. Management during this
with this diagnostic dilemma. therapy can be simplified by the use of IV
Children with midline brain tumors vasopressin administered as a continuous
should be assessed for a GCT. CSF should infusion.
be obtained for routine studies (glucose, Patients with NGGCT have had a poorer
protein, chamber count, and cytology) in outcome in the past, as these tumors are
addition to assessment of tumor markers generally less radiosensitive than pure ger-
(AFP and β‐HCG). Patients with classic minoma. Outcome is related to tumor his-
findings on MRI and elevated AFP with or tology, as those patients with pure embryonal
without a significantly elevated β‐HCG do carcinoma, endodermal sinus tumor, and
not require a biopsy for diagnosis, as they choriocarcinoma fair much more poorly
have chemical evidence of NGGCT. Patients than those with mixed histology, especially
with negative markers or a modest elevation those with a high proportion of teratoma or
in β‐HCG only should have a biopsy per- germinoma. Neoadjuvant platinum‐based
formed. Central nervous system GCTs often chemotherapeutic regimens have shown
spread along the CSF pathways; thus, MRI benefit in patients with NGGCT prior to
of the spine should be obtained in addition radiation therapy. Second‐look surgery is
to head imaging for diagnostic and staging beneficial in those patients with an incom-
purposes. plete radiologic response or persistently
Patients with benign tumors such as elevated tumor markers following induction
mature teratoma should have a curative chemotherapy, prior to radiation therapy.
Germ Cell Tumors 271

High‐dose chemotherapy with autologous acute myelogenous leukemia (AML) or


stem cell rescue has been shown to be of myelodysplasia (MDS). This often occurs
benefit in patients with residual malignant secondary to the presence of isochromo-
disease after chemotherapy and surgery. some 12p in both malignancies. Patients
Patients with recurrent disease may be may also present in rare cases with a
successfully treated, depending on initial mixed tumor with both germ cell as well
therapy. Patients with pure germinoma as primitive neuroectodermal tumor
treated solely with chemotherapy may ben- (PNET) elements. Finally, patients may
efit from additional chemotherapy fol- develop a growing mature teratoma or
lowed by radiation therapy. Those that have other nongerm cell malignancy after
previously received radiation may benefit chemotherapy.
from high‐dose chemotherapy with autolo-
gous stem cell rescue. Patients with recur-
rent NGGCT have a much more dismal Case study for review
outcome, though may benefit from addi-
tional chemotherapy, surgery, radiation, You are seeing a 17‐year‐old male who was
and/or high‐dose chemotherapy with stem admitted from the emergency department
cell rescue. (ED) after presenting with acute onset of
shortness of breath and a near syncopal
episode. Follow‐up questioning in the ED
Paraneoplastic syndromes reveals no prior illness, no fever, no loss of
appetite or weight loss, no recent travel,
Neurological manifestations can occur at and no night sweats. The ED resident
presentation as a clue to the underlying astutely asks about orthopnea and the
diagnosis of a GCT. The most common patient does note that it has been more dif-
paraneoplastic syndrome is the develop- ficult to lie flat, and he has been using
ment of NMDA (anti‐N‐methyl‐d‐ about two to three pillows now to feel com-
a­spartate) receptor antibody encephalitis fortable sleeping. He has been having
seen most commonly in mature ovarian increasing coughing which he attributes to
­teratoma and less frequently in immature a “stitch” on his right side. The patient’s
ovarian teratoma. Patients present with vitals are abnormal with an elevated heart
psychiatric symptoms and progressive neu- rate (120), elevated respiratory rate (16),
rologic dysfunction including seizures, and decreased O2 sat (93%). Exam reveals
dyskinesia, and autonomic dysfunction. decreased breath sounds in the bilateral
Diagnosis can be determined by the pres- bases with egophony. He is unable to take a
ence of autoantibodies against the gluta- big breath without coughing. There is no
mate receptor. Female patients with these increased jugular venous pressure (JVP),
findings must be screened for ovarian plethora, or hepatomegaly. There are no
pathology and, if found, proceed to emer- palpable cervical, supraclavicular, axillary,
gent tumor resection. or inguinal nodes.
You are initially concerned about an
anterior mediastinal mass causing ortho-
Concomitant malignancies pnea and order a chest X‐ray (CXR). CXR
reveals a large mediastinal mass with associ-
Patients with GCTs may in rare cases pre- ated bilateral pleural effusions, R > L.
sent with concomitant or secondary a. What is the differential diagnosis?
272 Chapter 21

b. What are the appropriate next steps? ­vascular and airway compromise—this will
The most common etiologies in pediatric assist the anesthesiologist’s assessment of
patients with an anterior mediastinal mass the airway and risk of decompensation with
are acute lymphoblastic leukemia (ALL) and different levels of sedation. The patient will
non‐Hodgkin lymphoma (NHL). These need a mediastinal biopsy and thoracentesis
typically will present with clinical findings with cytology of the pleural effusion may
and laboratory abnormalities suggestive of assist with diagnosis as well as alleviate
these underlying etiologies. ALL will likely symptoms. Depending on the extent of
present with cytopenias, possibly peripheral orthopnea as well as compromise noted on
blasts, as well as potential elevation in mark- the CT imaging, the patient may benefit
ers of increased cellular turnover (i.e., LDH, from observation in the critical care unit.
uric acid, most commonly). NHL often will CT chest returns with the finding of a
­present with increased LDH and uric acid as large mediastinal tumor and a large right‐
well as markers of inflammation including sided pleural effusion compromising the
ESR and ferritin. Hodgkin lymphoma is also lower half of the right lung. There is minimal
­common in an adolescent patient and may compromise of the superior vena cava and
present with B symptoms (objective fever, bilateral subclavian veins. In the interim,
night sweats, objective weight loss), as well the AFP returns elevated at 30 000 ng/ml
as elevation in inflammatory markers. (normal <10 ng/ml), while the β‐HCG is
Lymphoma and ALL may have clinical signs normal.
of tumor bulk including lymphadenopathy d. How does this aid in diagnosis?
and hepatosplenomegaly. Other possible e. What are the next steps?
causes of an anterior mediastinal mass Given the very elevated AFP, the patient has
include GCTs, Ewing s­ arcoma, undifferenti- a GCT, likely yolk sac histology with possi-
ated sarcoma, and rarely thymoma or thy- ble embryonal carcinoma pathology (i.e.,
roid carcinoma. non‐seminomatous). Male patients with
Given the likely etiologies, you order the elevated β‐HCG can present with gyneco-
following labs: CBC/diff, CMP, phosphorus, mastia secondary to β‐HCG stimulating
uric acid, LDH, ESR, ferritin, AFP, and androgen production (secondary to com-
β‐HCG. The initial labs are all normal with monality with luteinizing hormone [LH])
β‐HCG and AFP pending. Given the lack of subsequently converted to estrogen). β‐
any signs of tumor bulk you wonder if this will HCG is produced by syncytiotrophoblastic
be something other than ALL or NHL. elements within the tumor. Male patients
c. What are the next steps? with underlying Klinefelter’s syndrome are
Given the orthopnea, there is risk for at increased risk of GCTs. Remember that
cardiovascular decompensation. As there
­ infants will have a physiologic elevation in
are no markers that this is ALL or NHL, it is AFP with standard available normal refer-
unclear that steroids will be of benefit in an ence ranges for age. Though laboratory
emergent situation. Fortunately, the other markers are important in assisting with
potential ­etiologies in the differential diag- diagnosis as well as following for response
nosis have a slower doubling time meaning and tumor recurrence, it is still vital to prove
that the risk of acute decompensation is less. pathology based on a biopsy specimen. The
Either way, it is important to obtain a CT patient should still proceed with mediastinal
chest with/without contrast to evaluate the biopsy and thoracentesis of the pleural
great vessels and determine the extent of effusion.
Germ Cell Tumors 273

Biopsy shows a pure yolk sac tumor and


Suggested reading
the patient is started on standard therapy with
BEP. You perform baseline pulmonary func-
Fetcko, K. and Dey, M. (2018). Primary central
tion testing and audiology screening given the nervous system germ cell tumors: a review
risk of pulmonary fibrosis from bleomycin and update. Med. Res. Arch. 6: 1719.
and ototoxicity from cisplatin. Additionally, a Frazier, A.L., Hale, J.P., Rodriguez‐Galindo, C.
PET/CT is performed as a staging workup and et al. (2015). Revised risk classification for
does not show any sites of disease. Although pediatric extracranial germ cell tumors based
the overall prognosis for GCTs is quite good, on 25 years of clinical trial data from the
mediastinal GCTs have a poorer prognosis. United Kingdom and United States. J. Clin.
AFP response is important and the AFP Oncol. 33: 195–201.
should be followed closely to review whether Olson, T.A., Murray, M.J., Rodriguez‐Galindo, C.
et al. (2015). Pediatric and adolescent extrac-
it is decreasing as appropriate (based on a
ranial germ cell tumors: the road to collabora-
half‐life of 5 days). In this case, with an initial
tion. J. Clin. Oncol. 33 205: 3018–3028.
AFP of 30 000 ng/ml, the AFP should drop to Plant, A.S., Chi, S.N., and Frazier, L. (2016).
around 500 ng/ml after 1 month (i.e., six half‐ Pediatric malignant germ cell tumors: a com-
lives). Patients with mediastinal GCTs may parison of the neuro‐oncology and solid
require definitive resection of the mass and tumor experience. Pediatr. Blood Cancer 63:
additional ­chemotherapy cycles as compared 2086–2095.
to the standard three to four cycles of BEP for
most GCTs.
22 Rare Tumors of
Childhood
The rarer tumors of childhood collectively affected children. Genetic counseling is of
comprise fewer than 20% of all pediatric utmost importance in affected families due
cancers. The more frequently seen of these to the risk of tumors in siblings as well as
include retinoblastoma (2–3%), liver tumors offspring of survivors, especially in those
(1%), and epithelial tumors of the adrenal or with bilateral retinoblastoma.
thyroid gland (2–3%), which are discussed The RB1 gene is a tumor suppressor gene
herein. Many other rare pediatric malignan- and individuals with loss of function of both
cies also exist that are beyond the scope of allelic copies are predisposed to malignancy.
this chapter. Multiple tumors developing within the eye
may be synchronous or metachronous.
Germline de novo mutations of the RB1
Retinoblastoma gene largely occur during spermatogenesis.
Secondary mutations may then be germline
Retinoblastoma is the most common malig- or somatic depending on the timing of
nant ocular tumor in childhood, affecting events. Those mutations that are germline
200–300 children per year in the United are heritable for future generations. In spo-
States. Retinoblastoma is rarely diagnosed radic cases, two somatic mutations occur
after 5 years of age, and the median age of postconception in the RB1 gene, resulting in
diagnosis is 2. This unique neoplasm has a a tumor that is typically unifocal or unilat-
strong genetic component related to loss of eral. Prenatal diagnosis can identify ger-
function mutations of the RB1 gene located mline mutations.
on chromosome 13q14. It was the model for Retinoblastoma arises from the photo-
the development of Knudson’s two‐hit receptor cells of the innermost layer of the
hypothesis after observation that children retina. The tumor frequently extends into
with bilateral retinoblastoma developed dis- the vitreous cavity, presenting with a
ease at a much earlier age compared to those fleshy nodular mass visible on ophthal-
with unilateral disease, typically in the first mologic examination. Large tumors may
year of life. The genetic form of the disease is occupy most of the posterior chamber.
inherited in an autosomal dominant man- Less frequently, tumors may extend exter-
ner with high penetrance, yet only approxi- nally resulting in retinal detachment. The
mately 15% of children have a family history, tumor tends to outgrow its blood supply
suggesting the acquisition of a new germline and may develop areas of necrosis and
or somatic mutation in the majority of calcification.

Handbook of Pediatric Hematology and Oncology – Children’s Hospital & Research Center Oakland,
Third Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
© 2021 John Wiley & Sons Ltd. Published 2021 by John Wiley & Sons Ltd.
276 Chapter 22

Clinical presentation patients tend to have a poor prognosis. The


Due to the large mass or retinal detachment, intracranial tumor may present at the same
the normal papillary red reflex is replaced by time as the retinoblastomas, or after the iden-
leukocoria (white papillary discoloration) tification and treatment of the original
and is often first noticed by the family (or in tumors, emphasizing the need for close
a photo with a white reflex). Strabismus is ­follow‐up after initial treatment.
the second most common presenting sign
and is due to visual impairment from the Diagnosis and staging
tumor. Other less common presenting signs Retinal imaging, magnetic resonance imag-
include heterochromia, inflammatory ing (MRI), and computed tomography (CT)
changes, hyphema, and glaucoma. Familial are typically used in addition to ophthalmo-
cases are often detected early when the fam- logic examination to further characterize
ily is appropriately advised on the need for the tumor. CT is the most useful test, as it
frequent and careful screening with ophthal- effectively demonstrates intraocular calcifi-
mologic examinations under anesthesia. The cations that confirm the diagnosis, but it
differential diagnosis of leukocoria includes creates risk due to radiation that increases
a number of rare nonmalignant conditions the likelihood of secondary malignancy.
such as Coats disease, congenital cataracts, MRI of the orbits and brain is most useful
toxocariasis, retinopathy of prematurity, and for identification of extraocular extension
persistent hyperplastic primary vitreous. and trilateral retinoblastoma. MRI of the
Careful retinal examination by an ocular spine and lumbar puncture for cerebrospi-
oncologist or retinal specialist can rule out nal fluid cytology should be done in patients
these other diseases. The ophthalmologic with intracranial spread, involvement of the
exam is done typically under anesthesia with optic nerve, or evidence of tumor invasion
careful assessment of the extent of intraocu- beyond the lamina cribrosa on pathology
lar tumor and evaluation of both eyes with after eye enucleation. In patients with delay
maximal dilation. Patients with advanced in diagnosis and concern for metastatic dis-
disease may present with proptosis and ease, bone marrow studies and bone scan
swelling depending on the degree of extraoc- should be performed.
ular invasion. Metastatic spread can occur
through infiltration of the optic nerve, dis- Treatment
semination into the subarachnoid space, Patients with known or suspected retino-
invasion into the choroid plexus, and anteri- blastoma should be referred to a pediatric
orly into the conjunctiva. Invasion through oncologist in specialized centers with both
the optic nerve and subarachnoid can lead to oncologists and ophthalmologists with
involvement of the brain and spinal cord, expertise in the management of these chil-
invasion of the vascular choroid plexus can dren. Goals for treatment of children with
lead to vascular spread, and anterior invasion retinoblastoma include preservation of life,
can lead to regional lymph node involve- as much vision as possible, and avoidance of
ment. Given the concern for rupture of the radiation in patients with RB gene muta-
tumor, surgical biopsy is not recommended tions given the high risk of second malig-
for tissue confirmation. nancies. Treatment for retinoblastoma is
Patients with bilateral retinoblastoma may individualized and dependent on multiple
present with a concurrent intracranial neuro- factors including unilaterality or bilaterality
blastic tumor (typically of the pineal gland) of disease, potential for preserving vision,
referred to as trilateral retinoblastoma. These and extent of intraocular and extraocular
Rare Tumors of Childhood 277

disease. Enucleation performed by an expe- to non‐target tissues. Lower doses of radia-


rienced ophthalmologist is indicated for tion are also being investigated.
large tumors filling the vitreous and in those Patients who have undergone enuclea-
with little or no likelihood of vision preser- tion and determined to have disease at the
vation. The eye must be removed intact to cut edge of the optic nerve or extension into
avoid seeding and care must be taken to the sclera or extrascleral tissues should con-
resect enough optic nerve to ensure negative tinue with either adjuvant chemotherapy or
margins. After enucleation, patients are fit- local radiation.
ted with a prosthetic implant. Patients with regional extraocular dis-
Treatments aimed at local tumor control ease and metastatic disease have historically
include cryotherapy, infrared laser (ther- faired very poorly. Newer studies have
motherapy), laser photoablation, or a shown the potential benefit of chemother-
­radioactive plaque (I‐125 brachytherapy). apy (or high‐dose chemotherapy with autol-
Chemotherapy may be utilized for tumor ogous stem cell rescue in those with
reduction to facilitate more effective focal metastatic disease) with external beam radi-
treatment and prevent the need for enuclea- ation therapy. Impacts on outcomes utiliz-
tion. Systemic chemotherapy may also ing this treatment plan are still to be
be given in combination with enucleation determined.
in unilateral or bilateral disease (unifocal
enucleation) or following enucleation in
­ Follow‐up
patients with high‐risk pathologic features Children treated for retinoblastoma are at
predisposing to recurrence or metastatic high risk for cosmetic and functional
disease (i.e., massive choroidal involve- impairment related to either enucleation or
ment, postlaminar optic nerve involvement radiation. Patients with a heritable muta-
[ONI], or any uveal involvement in the tion in the RB1 gene have an especially high
presence of ONI). A number of different rate of secondary malignancy after radia-
chemotherapeutic regimens have been tion therapy. Reported second malignancies
studied and most typically include carbopl- include head and neck cancers, osteosar-
atin, etoposide, and vincristine (CEV) for coma, soft tissue sarcoma, and melanoma.
six cycles. Intra‐arterial chemotherapeutic Adult tumors also have an increased inci-
(IAC) delivery under fluoroscopic guidance dence in those with germline RB1 muta-
into the ophthalmic artery is a more recent tions, especially epithelial tumors such as
intervention and may aid in preserving the lung cancer. Radiation therapy may also
eye and preventing systemic effects of lead to decreased visual acuity, eye irrita-
chemotherapy in a young child. tion, and dry eye, as well as growth abnor-
External beam radiation therapy is gen- malities in the orbital bones. After therapy,
erally reserved for patients with refractory patients should be followed closely due
or recurrent disease after these other treat- to the risk of recurrent disease. Patients
ment modalities have been exhausted due to with bilateral retinoblastoma and those
the high incidence of side effects including diagnosed with unilateral retinoblastoma at
increased risk of secondary malignancy a young age (i.e., <1 year of age) should
both in and out of the radiation field. be observed for the development of an
Current protocols with conformal, stereo- intracranial mass and, in the case of
­
tactic, and proton‐beam radiotherapy, as unilateral disease, a contralateral retinal
­
well as intensity‐modulated radiotherapy, mass. Recurrence or new disease rarely
have been developed to minimize exposure occurs after 5 years of age, although an
278 Chapter 22

­phthalmologist should examine these


o Patients with Beckwith–Wiedemann
patients annually under anesthesia. ­syndrome or hemihypertrophy should be
screened every 3 months with serum α‐
fetoprotein (AFP) and abdominal ultra-
Liver tumors sound until 5 years of age given the
significant association with HBL (with
Primary hepatic tumors in childhood continued ultrasound screening until age 8
and adolescence account for just >1% of to rule out Wilms tumor). The strongest
malignancies in these age groups. association has been reported in infants
Hepatoblastoma (HBL) is the most com- born at <1500 g. Additional associations
mon, representing more than 60% of may include maternal smoking (independ-
hepatic tumors, and occurs almost exclu- ent of birth weight), fetal alcohol syn-
sively during infancy and young child- drome, use of oral contraceptives during
hood. The incidence of hepatoblastoma is pregnancy, and occupational exposure to
increasing, possibly due to increased sur- metals, petroleum, paints, and pigments.
vival in very low birth weight infants. HCC is seen in association with hereditary
Hepatocellular carcinoma (HCC), approx- tyrosinemia, biliary cirrhosis, glycogen
imately 20–25% of childhood primary liver storage diseases, hemochromatosis, ataxia
tumors, is seen most often in children telangiectasia, Wilms tumor with liver
above 10 years of age. Although chronic metastasis treated with hepatic radiation,
hepatitis B infection is the leading cause of and α1‐antitrypsin deficiency. Infection
HCC in Asia, few American children with with hepatitis B or C has been identified as
HCC have an apparent etiology. Benign a risk factor for HCC as have exposure to
hepatic tumors, such as hemangioendothe- alcohol, anabolic steroids, aflatoxin, and
lioma, hemangioma, hamartoma, mature certain carcinogens such as pesticides and
teratoma, angiolipoma, biliary cyst, and vinyl chloride.
adenoma, also occur in childhood and Factors influencing survival include
account for up to 20% of liver tumors in staging, histology, resectability, and treat-
this age group. Other extremely rare malig- ment response. Overall survival for children
nant hepatic tumors include cholangiocar- with HBL is 70% and only 25% for HCC.
cinoma, rhabdoid tumor, yolk sac tumor, Patients with stage I HCC have a good out-
rhabdomyosarcoma, undifferentiated sar- come while those with stage IV disease have
coma, angiosarcoma, leiomyosarcoma, and a high mortality.
lymphoma.
Malignant hepatic tumors of childhood Clinical presentation
have been associated with a number of The most common presenting signs of a
genetic syndromes and environmental risk malignant hepatic tumor in childhood are
factors. HBL has been reported in children generalized abdominal enlargement or an
with Beckwith–Wiedemann syndrome, asymptomatic abdominal mass palpated by
familial adenomatous polyposis (which the family or physician. Patients with HBL
includes Gardner syndrome), Li–Fraumeni or HCC may present with constitutional
syndrome, trisomy 18, glycogen storage symptoms including weight loss, anorexia,
disease type I, Prader–Willi syndrome,
­ emesis, and abdominal pain. Rarely, chil-
Meckel’s diverticulum, congenital absence dren may present with jaundice including
of an adrenal gland or kidney, umbili- cases with biliary tree involvement.
cal hernia, and isolated hemihypertrophy. Occasionally, males with HBL present with
Rare Tumors of Childhood 279

signs of isosexual precocious puberty due to specifically liver ultrasound with Doppler
excessive production of the beta subunit of flow to characterize the tumor as well as the
human chorionic gonadotropin (β‐HCG) relationship between the tumor and hepatic
that is converted to testosterone. Metastatic vessels. Abdominal CT or MRI should also
disease (typically asymptomatic) may occur be obtained prior to surgical intervention.
at presentation in up to 20% of children with MRI angiography and cholangiogram may
HBL or HCC. Extrahepatic extension may be useful to assess degree of intrahepatic
include peritoneal implants as well as spread disease and aid in determining a surgical
to regional and distant lymph nodes, bone, plan, but is rarely needed now due to the
bone marrow, and rarely the central nervous excellent resolution of current generation
system. Liver tumors may also invade into CT and MRI. Evaluation for metastatic dis-
adjacent intra‐abdominal structures. ease should include chest radiographs and
CT, as up to 10–20% of patients with liver
Diagnosis and staging tumors will have pulmonary metastases.
Work‐up of a suspected hepatic mass Dependent on extent of disease and symp-
should include a complete history and toms, consideration can be given to obtain-
physical including assessment of co‐mor- ing a bone scan and bone marrow aspirate/
bid or predisposing genetic syndromes, biopsy. The right lobe of the liver is most
laboratory measurement of liver function commonly affected in both HBL and HCC,
with coagulation studies, urinalysis, testing but can involve both lobes in up to 30% of
for hepatitis B (surface antigen, core anti- cases of HBL.
gen, core antibody) in addition to transam- Staging of liver tumors is complex and
inases, total bilirubin, and alkaline both presurgical (PRETEXT, stages 1–4)
phosphatase. Patients should have a base- and postsurgical (stages I–IV) staging sys-
line complete blood count (CBC) in addi- tems are utilized. The PRETEXT staging for
tion to metabolic studies, as patients may HBL is utilized internationally, though some
present with thrombocytosis (>500 × 109/l) differences in treatment are employed in
and/or moderate leukocytosis. Patients Europe and the United States with similar
with suspected HBL typically present with outcomes and current study of a more uni-
an elevation of AFP and, less commonly, of fied approach.
β‐HCG. In certain cases with an extremely
high AFP, the value reported may be falsely Treatment
low due to overwhelming of the assay In cases where clinical presentation, imag-
(hook effect). When the index of suspicion ing, and tumor markers are highly suggestive
is high, serial dilution of serum can allow of HBL, patients should undergo a primary
for more accurate reporting. Those with complete resection if it is deemed safe and
HCC may also have elevation in AFP feasible. Only patients who achieve a com-
but not to the same magnitude as hepato- plete surgical resection have a reasonable
blastoma. β‐HCG may be elevated in chance of cure. Decisions regarding an initial
patients with carcinoma of the biliary tract. surgical approach (as opposed to presurgery
Carcinoembryonic antigen (CEA) and vita- chemotherapy) are dependent on expected
min‐B12‐binding capacity may be elevated degree of resectability with suitable candi-
in some cases of HCC (with elevated B12‐ dates including those with tumor confined to
binding capacity associated with the one lobe of the liver or originating in the right
fibrolamellar variant of HCC). Imaging lobe and not extending beyond the medial
studies should begin with abdominal and segment of the left lobe. Patients should be
280 Chapter 22

referred to a pediatric liver transplant center resection rate and improved survival com-
with an experienced liver transplant surgeon pared to other forms of HCC. Novel tar-
for initial decisions regarding attempted geted agents have shown a modest benefit at
resection upfront or delayed resection post best, but may have promise when used as
chemotherapy. Orthotopic liver transplant part of multimodal therapy.
may also be warranted in patients. In cases Radiation therapy has not been proven
where a complete resection is not feasible or to be effective in the treatment of HCC, but
the diagnosis is uncertain, the patient should it may have a role in palliation of lung
first undergo biopsy. metastases. Similarly, high‐dose chemother-
Patients with low‐stage (I and II) disease apy with autologous stem cell rescue has not
and pure fetal histology can be safely as yet been found beneficial. Local therapies
observed without adjuvant chemotherapy. including hepatic artery chemoemboliza-
Patients who cannot obtain a complete tion, percutaneous radiofrequency ablation,
resection (stages III and IV; metastatic dis- and percutaneous ethanol injection may be
ease) or have a less‐favorable histology beneficial in patients with localized yet
should receive chemotherapy followed by unresectable HCC.
resection (including orthotopic liver trans-
plant if not resectable), with resection of Follow‐up
residual pulmonary metastasis in stage IV AFP is the most sensitive marker for dis-
patients if unresolved with chemotherapy ease, especially in HBL. Approximately 90%
alone. Various combinations of chemother- of children with HBL and 50% of those with
apy have been utilized and most commonly HCC present with an elevated AFP. Patients
employ cisplatin, 5‐FU (fluorouracil) vin- with small‐cell undifferentiated histology
cristine, and doxorubicin. Chemotherapy is have a poorer prognosis and tend to have
utilized preoperatively to shrink tumor and low serum AFP, which is a poor prognostic
facilitate complete surgical resection (or pro- marker. Decline in AFP levels with courses
vide time for identification of an orthotopic of treatment is prognostic and AFP should
liver) in addition to after resection for treat- be followed for normalization after surgical
ment of potential micrometastatic disease. resection and as a marker for residual dis-
In general, HCC has a poor prognosis ease or recurrence. Of note, serum AFP lev-
and often presents with multifocal liver and els vary with age in infancy and are normally
metastatic disease. Previous studies treated elevated in the newborn period, declining
pediatric patients with a similar chemother- to adult levels by around 6–8 months of age.
apeutic regimen as HBL with disappointing In addition, AFP has a long serum half‐life
results. Cisplatin and doxorubicin may be (5–6 days) and therefore may take several
given presurgically, but clinical trials have weeks to normalize in cases with an
not shown a survival benefit. Gross total extremely high level at presentation.
surgical resection remains the mainstay of
cure for HCC. In cases where resection is
not safe or feasible at presentation, neoadju- Adrenocortical carcinoma
vant chemotherapy may be of benefit and
allow for a later total resection or for ortho- Adrenocortical carcinoma (ACC) is a rare
topic liver transplantation. malignancy in childhood and adolescence
A distinct pathologic variant, fibrolamel- (0.2% of childhood malignancies) with a
lar HCC, has been associated with a higher median presentation of 3–4 years of age and
Rare Tumors of Childhood 281

female predominance (two to three times in the serum or urine. Baseline studies
the number of affected boys). ACC is also should include a 24‐hour free cortisol,
seen in adults. The incidence varies world- ­dexamethasone suppression test, basal cor-
wide with a ten times higher incidence in tisol, and adrenocorticotropic hormone
southern Brazil associated with a high pop- (ACTH) to measure glucocorticoid
ulation prevalence of the R337H germline ­secretion; ­testosterone, estradiol, andros-
mutation in the TP53 tumor suppressor tenedione, dehydroepiandrosterone sulfate
gene. ACC has been reported in association (DHEA‐S) (most commonly elevated), and
with specific genetic syndromes such as 17‐OH‐progesterone to measure sex ster-
Beckwith–Wiedemann syndrome, isolated oids and their precursors; and aldosterone
hemihypertrophy, Li–Fraumeni syndrome, and renin in patients with hypertension or
and in association with congenital adrenal hypokalemia to rule out mineralocorticoid
hyperplasia and multiple endocrine neo- excess. Pheochromocytoma can be ruled
plasia type 1 (MEN‐1 syndrome). Germline out by checking catecholamines and
mutations in the TP53 tumor suppressor metanephrines. Imaging with CT or MRI
gene have been implicated in the majority of of the abdomen confirms the presence of a
pediatric cases in the United States and suprarenal mass. Radiologic features of a
Brazil. In addition to the association with malignant adrenal tumor include the pres-
constitutional genetic abnormalities, ACC ence of a large inhomogeneous enhancing
can also be seen with somatic genetic muta- mass in the adrenal gland (or appearing to
tions, including overexpression of IGF‐2 replace it) with areas of hemorrhage, calci-
and constitutive activation of the Wnt/β‐ fication, and necrosis. FDG‐PET may be
catenin pathway. utilized in equivocal cases or if CT and
MRI are negative with positive hormone
Clinical presentation markers. FDG‐PET is negative in benign
ACC may be functionally inactive or secrete adrenocortical tumors. The differential
hormones such as glucocorticoids, sex ster- diagnosis of an adrenal mass noted on
oids and their precursors, and mineralocor- imaging includes other malignant tumors
ticoids. Benign adrenocortical tumors are such as neuroblastoma, ganglioneuroblas-
more common and must be carefully differ- toma, and teratoma. Therefore, the compi-
entiated from ACC. Patients frequently lation of hormonal lab results and imaging
present with clinical signs and symptoms of findings should be considered in a diag-
excessive cortisol production (Cushing’s nostic approach. Many patients present
syndrome) and/or excessive androgen pro- with large primary tumors and evidence of
duction (virilization). The diagnosis is often metastatic disease involving the lungs,
delayed. Patients may present with an liver, or bones. CT of the chest, abdomen,
abdominal mass, pain, weight loss, or hyper- and ­pelvis in addition to bone scan should
tensive urgency/emergency, but typically be done for staging purposes prior to surgi-
come to medical attention due to the high cal ­resection. Several staging systems are
rate of endocrinopathy. utilized for ACC. Prognosis is linked to
stage, tumor size, invasion of major blood
Diagnosis and staging vessels, degree of resection (i.e., complete
The diagnosis is made based on the pres- or not), tumor spillage during surgery, his-
ence of elevated concentrations of adreno- tologic characteristics, and presence of
cortical hormones and their intermediates metastatic disease.
282 Chapter 22

Treatment ­ eurologic side effects. As mitotane is an


n
ACCs are locally aggressive tumors with thin adrenocorticolytic agent, replacement with
pseudocapsules; an open adrenalectomy hydrocortisone and fludrocortisone is nec-
should be performed to prevent ­extensive essary to prevent adrenal insufficiency and
hemorrhage, tumor rupture, or seeding supraphysiologic dosing is often necessary
along the needle tract. Surgical resection is due to mitotane’s effect on steroid metabo-
the only known curative therapy for ACC, lism. Multiple chemotherapeutic regimens
both for primary and metastatic sites. and targeted therapies have been utilized
Surgery is also indicated in recurrent disease in the treatment of ACC, but treatment of
to prolong survival. Patients with evidence refractory or recurrent disease remains
of hypercortisolism should be assumed to be challenging. The combination of mitotane,
adrenal‐insufficient and receive appropriate gemcitabine, and capecitabine has been
stress dosing of steroids. used with some success as second‐line or
Due to the poor response to chemother- third‐line therapies. ACC is considered
apy, patients with advanced disease have a radioresistant, but some studies indicate a
very poor prognosis. A low tumor possible role of radiation therapy as an
weight <200 g (<5 cm in diameter in adults) adjuvant post resection. Radiation therapy
is a good prognostic factor. Patients with in children has not been established and
small tumors and localized disease should given the tumor’s association with TP53
undergo aggressive gross total surgical mutations, it is not considered a therapeutic
resection. Patients with advanced disease option.
(invasive or metastatic) should be treated
with aggressive en bloc surgical resection of
Follow‐up
the primary tumor and metastatic lesions. If
The initial hormonal evaluation assists with
resection of metastases is not feasible,
diagnosis, but also provides useful tumor
­resection of the primary tumor may not be
markers that may be utilized to detect persis-
of benefit.
tent or recurrent disease. Long‐term follow‐
Systemic chemotherapy is recom-
up includes assessment of tumor marker(s)
mended for patients with unresectable or
and periodic imaging. Patients who have
metastatic disease. Mitotane, a synthetic
received mitotane can take many months to
derivative of the insecticide dichlorodiphe-
recover from the adrenal suppressive effects
nyltrichloroethane (DDT), has a specific
and may require replacement or stress dos-
cytotoxic effect on adrenocortical cells. It
ing. Patients with identified genetic syn-
has shown benefit in a subset of patients
dromes or TP53 mutations need lifelong
with invasive, metastatic, or recurrent dis-
surveillance for other malignancies.
ease. Mitotane may have benefit in patients
with localized, fully resected disease,
although this question has not been clearly
answered. Mitotane is used as monotherapy Thyroid tumors
or in combination with typical antineoplas-
tic drugs including doxorubicin, etoposide, The incidence of malignant thyroid carci-
and cisplatin (EDP). Monthly monitoring noma in children is very low, comprising
of mitotane levels is important to achieve a less than 2% of pediatric cancers. The peak
therapeutic level of 14–20 mg/l. Toxicities incidence is in the older adolescent age
that may limit dosing or treatment with group, 15–19 years of age, with a prepon-
mitotane include gastrointestinal and derance in females (this gender difference is
Rare Tumors of Childhood 283

not present in children <10 years of age). much more likely than adults to present with
Childhood ­thyroid carcinomas are clinically advanced or metastatic disease (primarily
and bio­logically distinct from those seen in lungs) and metastatic disease is commonly
adults. The majority of malignant thyroid seen with the papillary variant.
cancers arise from the follicular epithelium
and include papillary and follicular carcino-
mas. Fewer than 10% of malignant child- Diagnosis and staging
hood thyroid tumors are medullary The initial evaluation of the child with a
carcinomas, arising from neural crest cells. thyroid nodule should include thyroid
Exposure to head and neck radiation is asso- ultrasound as well as measurement of free
ciated with an increased risk of thyroid car- thyroxine (FT4) and thyroid‐stimulating
cinoma (more commonly the papillary hormone (TSH) (though thyroid function
variant), particularly in children <5 years of studies are typically normal). Up to 20% of
age. An increased incidence has been seen thyroid nodules in children will be malig-
in children receiving therapeutic radiation nant, and younger age, past history of radia-
with a median latency of 13 years and has tion, genetic cancer syndromes, and family
also been observed in survivors following history are all highly predictive of malig-
the Chernobyl nuclear accident. Incidence nant disease. Ultrasound characteristics may
of medullary thyroid carcinoma is highest in guide the clinician in the diagnosis, as
the 0–4‐year age group. Genetic predisposi- malignant tumors are more likely to have
tion plays a major role in the development of indistinct margins, enhanced vascularity, an
these tumors which are thought to arise absence of an echogenic halo around the
from inherited or de novo activating muta- nodule, and presence of calcifications.
tions of the RET proto‐oncogene. Hereditary Nuclear medicine thyroid scintigraphy (usu-
medullary carcinoma is highly associated ally with 123I) can be utilized for surgical
with MEN types 2A and 2B or as part of planning, screening for postoperative persis-
familial medullary carcinoma. Patients with tent or metastatic disease, and to determine
MEN type 2A develop medullary thyroid if a patient may benefit from treatment with
carcinoma, pheochromocytoma, and para- 131
I. The majority of thyroid nodules are cold
thyroid tumors. Patients with MEN type 2B on thyroid scan and most of these are benign
may develop medullary carcinoma or pheo- follicular adenomas. Image‐guided fine
chromocytoma and are associated with a needle aspiration is recommended to obtain
marfanoid body habitus, mucosal neuro- tissue diagnosis. Surgical removal may be
mas, and ganglioneuromatosis. A high inci- considered in higher‐risk patients with sus-
dence of papillary carcinoma of the thyroid picious features on imaging, positive family
is seen in familial adenomatous polyposis history, or past history of radiation. When
coli and Cowden disease. a thyroid carcinoma is diagnosed, staging is
completed with a neck ultrasound, CT, or
MRI to evaluate possible lymph node
Clinical presentation involvement. Chest CT is done to evaluate
Patients typically present with a painless for metastasis (often not visualized on plain
solitary thyroid nodule (approximately radiographs), and neck MRI can assess for
70–75%) and up to 35–40% present with degree of local invasion. Staging in children
palpable cervical adenopathy. If the tumor is based on the presence or absence of metas-
has invaded locally, patients may present tases. Children diagnosed before age 10 years
with dysphagia or dysphonia. Children are may have an increased risk of recurrence.
284 Chapter 22

Presence of metastatic disease, age, size of recurrence as well as late effects of treat-
tumor, and degree of extrathyroid invasion ment including development of second
are predictive of outcome. cancers.
Patients with medullary carcinomas are
treated with surgical excision, but should
Treatment undergo biochemical evaluation for pheo-
Surgical resection performed by an experi- chromocytoma preoperatively. These
enced surgeon is key to curative therapy tumors may also express serum calcitonin
in thyroid carcinoma. Patients should or CEA which can serve as useful tumor
undergo a total thyroidectomy, particularly markers. Genetic testing for RET should be
for tumors >1 cm in size. This approach has done and the patient and family referred
been associated with lower rates of recur- for genetic counseling. Medullary thyroid
rence and better survival than with lobec- carcinoma does not trap iodine and there-
tomy. Lymph node dissection should be fore is not treated with 131I. Molecular ther-
compartment‐focused and comprehensive. apies targeting tyrosine kinase inhibitors
When lymph node involvement is con- that inhibit RET and other receptor tyros-
firmed by imaging or biopsy, a modified ine kinases known to inhibit angiogenesis
lateral neck dissection is recommended. are currently under investigation. These
Following surgery, a diagnostic whole‐body tumors are not sensitive to chemotherapy.
scan with 123I (preferred) or 131I is recom- Post-operatively, patients are treated with
mended to define areas of residual disease. thyroid hormone to normalize and not sup-
Radioiodine ablation is then recommended press TSH levels.
and has been shown to reduce the incidence
of locoregional recurrence. Recombinant
TSH can also be utilized in low‐risk patients Suggested reading
in lieu of 131I. Patients with progressive dis-
ease not amenable to surgery and no longer Aerts, I., Sastre‐Garau, Y., Savignomi, A. et al.
responsive to 131I may benefit from treat- (2013). Results of a multicenter prospective
study on the postoperative treatment of uni-
ment with chemotherapy, typically doxoru-
lateral retinoblastoma after primary enuclea-
bicin alone or in combination with cisplatin tion. J. Clin. Oncol. 31: 1458–1463.
or α‐interferon. Molecular therapies tar- Aronson, D.C. and Meyers, R.L. (2016).
geted against tyrosine kinase inhibitors are Malignant tumors of the liver in children.
showing promise and sorafenib is currently Semin. Pediatr. Surg. 25: 265–275.
FDA approved in adults. External beam Chen, Q.L., Su, Z., Li, Y.H. et al. (2011). Clinical
radiation to treat microscopic residual dis- characteristics of adrenocortical tumors in
ease in the neck is not recommended, but children. J. Pediatr. Endocrinol. Metab. 24:
may have a role in palliation of sympto- 535–541.
matic distant metastases. Lifetime thyroid Dermody, S., Walls, A., and Harley, E.H. Jr.
(2016). Pediatric thyroid cancer: an update
replacement is required after surgery and/
from the SEER database 2007‐2012. Int. J.
or radioablation. Most children with follic-
Pediatr. Otorhinolaryngol. 89: 121–126.
ular or papillary carcinomas have an excel- Ribeiro, R.C., Pinto, E.M., and Zambetti, G.P.
lent prognosis with 10‐year survival of (2010). Familial predisposition to adrenocor-
almost 100%, even in the presence of meta- tical tumors: clinical and biologic features and
static disease. Lifelong surveillance is war- management strategies. Best Pract. Res. Clin.
ranted in these patients to monitor for Endocrinol. Metab. 24: 477–490.
Rare Tumors of Childhood 285

Schmid, I. and von Schweinitz, D. (2017). hepatoblastoma: current perspectives. Hepat.


Pediatric hepatocellular carcinoma: chal- Med. 9: 1–6.
lenges and solutions. J. Hepatocel. Carcinoma Yikilmaz, A., George, M., and Lee, E.Y. (2017).
4: 15–21. Pediatric hepatobiliary neoplasms: an over-
Trobaugh‐Lotrario, A.D., Myers, R.L., O’Neill, view and update. Radiol. Clin. North Am. 55:
A.F., and Feusner, J.H. (2017). Unresectable 741–766.
23 Histiocytic Disorders

Histiocytes (composed of dendritic cells and Langerhans cell histiocytosis


monocytes/macrophages; also called tissue
macrophages) are cells of the mononuclear LCH is now the accepted umbrella term
phagocytic system that fight infection and for a wide variety of other conditions pre­
clear debris. Histiocytic disorders occur due viously described, including histiocytosis
to abnormal proliferation or activity of X, ­eosinophilic granuloma, Hand–Schüller–
these cells and have a wide variety of clinical Christian syndrome, Letterer–Siwe disease,
presentations, from localized and mild to Hashimoto–Pritzker syndrome, self‐heal­
generalized and severe. ing histiocytosis, pure cutaneous histiocy­
The classification of these conditions tosis, Langerhans cell granulomatosis,
can be as confusing as the conditions them­ Langerhans cell (eosinophilic) granuloma­
selves. The most recent World Health tosis, type II histiocytosis, and nonlipid
Organization (WHO) classification was reticuloendotheliosis. The pathogenesis of
revised in 2016 and is presented in LCH is poorly understood. Immune dys­
Table 23.1. While most of the histiocytic regulation likely plays a role and may be
disorders are considered nonmalignant reactive, occurring secondary to trauma
conditions, many are treated similarly to and infection, e­ specially in those patients
malignant disease, and the mortality of sev­ with remitting disease. A neoplastic pro­
eral conditions is quite high. The etiology of cess may also be possible in cases that are
most types is unknown but appears second­ systemic and widely disseminated. LCH is
ary to poorly understood pathophysiologi­ a misnomer, as the Langerhans cell, an epi­
cal mechanisms. This chapter focuses on dermal dendritic cell, is not the sole cul­
the more common presentations of disease, prit; rather, like other histiocytic disorders,
specifically Langerhans cell histiocytosis LCH is due to abnormalities in the mono­
(LCH) and hemophagocytic lymphohistio­ nuclear phagocytic system. Diagnosis is
cytosis (HLH). Less common disorders based on histologic features of these
include juvenile xanthogranuloma (includ­ Langerhans cells (characterized by Birbeck
ing Erdheim–Chester disease) and sinus granules on electron microscopy) in addi­
histiocytosis with massive lymphadenopa­ tion to specific immunophenotypic mark­
thy (Rosai–Dorfman disease). ers (CD1a and CD207).

Handbook of Pediatric Hematology and Oncology – Children’s Hospital & Research Center Oakland,
Third Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
© 2021 John Wiley & Sons Ltd. Published 2021 by John Wiley & Sons Ltd.
288 Chapter 23

Table 23.1 Classification of histiocytic disorders.

Group Disease Condition

L Langerhans‐related Langerhans cell histiocytosis (LCH)


Indeterminate cell histiocytosis
Erdheim–Chester disease (ECD)
• Juvenile xanthogranuloma
Mixed LCH/ECD
C Cutaneous and mucocutaneous Cutaneous non‐LCH
• Xanthogranuloma family
• Non‐xanthogranuloma family
Cutaneous non‐LCH with a major
systemic component
R Rosai–Dorfman disease Familial Rosai–Dorfman Disease (RDD)
Sporadic RDD
• Classical RDD
• Extra‐nodal RDD
• RDD with neoplasia or immune disease
• Unclassified
M Malignant histiocytosis Primary malignant histiocytosis
Secondary malignant histiocytosis
• Histiocytic
• Interdigitating
• Langerhans
• Indeterminate cell
H Hemophagocytic lymphohistiocytosis Primary HLH
(HLH) and macrophage activation Secondary HLH
syndrome HLH of unknown/uncertain origin

LCH most often presents in the bone as a s­ignificant soft tissue swelling exists, there­
solitary or as multifocal lesions and may also fore radiation therapy should be considered.
be localized to the skin or lymph nodes. 2. Large or symptomatic solitary bone
Bone lesions may be painful or painless. lesions may benefit from surgical curettage
On plain film, lesions appear lytic, often or resection, intralesional steroid injection,
with a “punched out,” beveled appearance or radiation therapy.
and may have an associated periosteal 3. For solitary bone lesions, the recommen­
reaction with soft tissue swelling. Skin
­ dation for observation excludes sites that put
lesions are scaly, erythematous papules the patient at increased risk for the develop­
often involving the scalp. Localized disease ment of diabetes insipidus (DI) (facial bones
in these areas ­portends a good prognosis and anterior/middle cranial fossa). For these
with chance for spontaneous remission due “special sites,” and for multifocal bone, skin,
to “burning out” of the underlying immune or lymph node involvement, chemotherapy
reaction. Observation is generally favored in with 12 months of prednisone and vinblas­
this situation, with certain caveats: tine is recommended.
1. Vertebral lesions have an inherent risk of
causing vertebral compression (vertebra Systemic disease, which involves multiple
plana) or spinal cord compression if organs including the bone marrow, liver,
Histiocytic Disorders 289

spleen, or lungs, has a much worse prog­ Hemophagocytic


nosis, especially in those patients without lymphohistiocytosis
an early response to therapy. Poor respond­
ers likely will require more intensive ther­ Like other histiocytic disorders, immune
apy including, potentially, hematopoietic dysregulation is the key feature in HLH.
stem cell transplantation. Initial staging This condition presents with prolonged and
should include a skeletal survey and poten­ excessive activation of macrophages, histio­
tial PET/CT scan to rule out additional cytes, and cytotoxic T‐lymphocytes (CTL).
sites of disease. Brain MRI imaging should Natural killer (NK) cells, vital to antigen
be done in patients with symptoms con­ ­recognition and killing as well as contrac­
cerning for DI and can be considered in all tion of the immune response from T‐cells
patients at presentation. MRI may demon­ and histiocytes, often have decreased activ­
strate a thickened pituitary stalk or change ity, in part allowing for this hyperprolifera­
in the brightness of the posterior lobe tive immune response. Viral infection by
of the pituitary gland. Patients, especially Epstein–Barr virus is the most common
those with recurrent disease, should be known acquired cause, although multiple
screened for the BRAF‐V600E mutation, other viruses have been implicated, and
for which there is potentially ­beneficial tar­ often the pathogen is not identified. Familial
geted therapy. A neurodegenerative central HLH can occur secondary to multiple
nervous system disease has been rarely known genetic causes that lead to NK and
reported as a late finding after LCH diag­ CTL dysfunction. The familial causes are
nosis and treatment. outlined in Table 23.2. An underlying

Table 23.2 Genetic mutations associated with hemophagocytic lymphohistiocytosis.

Gene Screening result Pathophysiology

Perforin 1 (PRF1) Perforin decreased Synthesized in natural killer and cytotoxic


T‐cells, permeabilizes target membrane
allowing granzyme B to initiate
apoptotic pathways
MUNC13–4 (UNC13D) CD107a decreased Important for cytolytic granule fusion to
target membrane
RAB27A (Griscelli syndrome) CD107a decreased Docking of secretory granules
Syntaxin (STX11/STXBP2) CD107a decreased Failure of degranulation when
encountering susceptible targets
LYST (Chédiak–Higashi Involved in maturation of cytolytic
syndrome) enzyme granules
AP3B1/BLOC1S6 (Heřmanský– Polarization/intracellular movement of
Pudlák syndrome type II) cytolytic granules
SH2D1A (X‐linked SAP decreased Abnormal signaling for T‐ and NK‐cell
lymphoproliferative activation, impaired cytokine
syndrome type I) production
BIRC4 (X‐linked XIAP decreased Immune dysregulation, increased
lymphoproliferative T‐cell apoptosis, impaired cytokine
syndrome type II) production

Additional mutations include: ITK deficiency, CD27 deficiency, lysinuric protein intolerance
(SLC7A7), GATA2 deficiency.
290 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)

genetic predisposition may still require a underlying pathogenesis is due to an auto­


“second hit,” such as a viral infection, to immune condition, infection, or underlying
develop HLH. malignancy. Systemic onset juvenile
Clinical findings are secondary to the ­idiopathic arthritis (adult Still’s disease) is
chronic inflammatory state and most nota­ the most common autoimmune culprit.
bly include fever, splenomegaly, cytopenias, Multiple other autoimmune diseases have
and hepatitis. Diagnostic criteria for HLH been reported to cause MAS. NK cells are
are summarized in Table 23.3. For the vital in the prevention of autoimmunity;
patient with high clinical suspicion for decreased NK activity could result in both
HLH, treatment should be initiated the development of an autoimmune disease
promptly even while pending pertinent and secondary HLH. Often it is difficult to
laboratory evaluation. Current therapy separate the symptoms of MAS from the
includes dexamethasone and etoposide. underlying cause or primary HLH. Patients
Cyclosporine was previously utilized but may have leukocytosis, thrombocytosis, and
has been eliminated from the treatment hyperfibrinogenemia secondary to the
regimen, as it has been shown to provide no underlying inflammatory state, but these
additional benefit in HLH. For patients that levels will trend down with the development
have a complete response after 8 weeks, of MAS. Fevers may become non‐remitting,
treatment can be stopped, unless they are and there may be a paradoxical improve­
shown to have an underlying familial muta­ ment in inflammatory markers such as the
tion which predisposes them to HLH. For erythrocyte sedimentation rate (ESR) and
those that have not responded completely fibrinogen which can help differentiate
or relapse after stopping therapy, as well as MAS from HLH. Treatment for MAS
patients with a genetic mutation, hemat­ includes pulse steroids and cyclosporine. In
opoietic stem cell transplantation is the the very sick patient, a monoclonal antibody
treatment of choice. again IL‐1 should be considered though has
Excessive activation of macrophages and not been studied systematically. Duration of
T‐lymphocytes may also lead to a secondary therapy is dependent on clinical response;
or reactive HLH known as macrophage patients that do not respond or relapse
­activation syndrome (MAS). Clinical crite­ may benefit from primary HLH therapy,
ria for MAS are similar to HLH, and the although outcomes are poor.
Histiocytic Disorders 291

HLH less likely. In addition, the high CRP


Case studies for review
makes HLH less likely.
You review the lab trends:
1. You are seeing a 6‐year‐old child that
presents to the emergency department with
3 weeks of an erythematous and painful dif­ Hemoglobin: 12.5 → 10.3 → 8.9
Platelets: 150 → 98 → 67
fuse and spreading rash. Fever began soon
White blood cell count: 18.4 → 28.7 → 39.7
after the rash with associated weakness,
(continued left shift)
malaise, and anorexia. Initial laboratory ESR: 50 → 47
tests include the following: CRP 6.3 → 12.9 → 18.1
Complete blood count: Fibrinogen: 441 → 387 mg/dl

12.5
19.8 150
A ferritin is sent and is very elevated at 68 300
mcg/l. Fasting triglycerides are 253 mg/dl.
 Differential: 35% segs, 44% bands, 6% lymphs, b. How do these lab values help with the
and 12% monos differential?
The patient continues to have signs of an
ESR: 47 mm/h CRP: 6.3 mg/dl underlying inflammatory picture and hyper­
Complete metabolic panel: cytokinemia. Sepsis should still be consid­
ered on the differential with the continued
131 103 12
3.7 28 0.7
222 elevated ESR, CRP, and fibrinogen. The
worsening bicytopenia though should make
Total protein 5.2 g/dl
Albumin 2.1 g/dl
HLH and MAS higher on the differential.
AST 146 U/l The extremely elevated ferritin is relatively
ALT 44 U/l specific for HLH and MAS, and a level this
Total bili 0.3 mg/dl high is concerning for MAS. The decreasing
ESR and fibrinogen also raise the concern
that the patient is moving from symptoms of
On examination, the patient is alert and an underlying autoimmune disorder to
interactive, but seems somewhat uncom­ frank MAS. Finally, the elevated white blood
fortable from the rash. There is mild spleno­ cell count with neutrophilia makes MAS
megaly and no hepatomegaly. The patient is much more likely than HLH.
started on antibiotics for presumed sepsis/
toxic shock with initial resolution of fever. 2. You are seeing a 3‐year‐old female in
Cultures are all normal. Due to worsening your clinic. Mom notes that she has devel­
labs and rash, as well as no source of infec­ oped a non‐tender scalp swelling over the
tion, the differential diagnosis is revisited. last couple of months. There was a previous
The patient again becomes febrile. fall that may have precipitated the swelling,
a. What findings in the initial labora­ but the mom is surprised the area is still
tory tests suggest sepsis as well as HLH swollen. The child has otherwise been well
and MAS? without complaint: no fever, no weight
The patient has elevated inflammatory loss, no skin rash, good appetite, no gastro­
markers (CRP and ESR). Hyponatremia intestinal symptoms. On examination, the
and hypoalbuminemia are often seen with child is well‐appearing with a 2 × 3 cm2 area
an underlying inflammatory condition. of swelling in the right parietal region of
There are no significant cytopenias, ­making the scalp. There are no skin findings and
292 Chapter 23

there is no hepatosplenomegaly. There are


Multiple choice questions
no other areas of swelling or tenderness
noted.
1. You are seeing a 5-year-old in the pedi­
a. What potential complication of LCH
atric intensive care unit who was admitted
is it important to inquire about?
for hypotension and thought to have
Diabetes insipidus due to pituitary involve­
hypovolemic shock secondary to sepsis.
ment may occur at presentation, so it is
The sepsis rule out has been unremarka­
important to ask questions to help deter­
ble although the patient continues with
mine if this may be a consideration. You ask
features worrisome for DIC. All of the
the mother if the child has been drinking
following support a diagnosis of HLH
­
more and urinating more and she has not
EXCEPT:
noted any change in this regard. She is
a. Splenomegaly
potty‐trained and does not use the bath­
b. Transaminitis
room at night while sleeping.
c. Cytopenias
b. What is the best first diagnostic step?
d. Fever
In a young child, LCH should always be a
e. Elevated fibrinogen
consideration with localized swelling, espe­
Explanation: HLH should be considered in
cially in the scalp which is a common loca­
an ill‐appearing child who does not have
tion and may occur after trauma. A plain
an underlying etiology. Generally these
film of the scalp is often sufficient to make
patients will continue to worsen, though in
the likely diagnosis of LCH. Plain film shows
certain cases may have an indolent course.
the classic “punched‐out” lytic lesion that is
Their picture will often mimic the patient
most common with LCH.
with sepsis but will not improve with nor­
c. What are the next best steps?
mal supportive care measures. The previ­
The patient will need to be staged to
ous HLH guidelines suggested a diagnosis
determine if this is truly LCH and also
­
based on 5 of 8 criteria including: 1) daily
whether this is a solitary lesion or is present
fevers; 2) splenomegaly; 3) bicytopenia;
with additional lesions. Baseline laboratory
4) hypertriglyceridemia or hypofibrino­
markers should include complete blood
genemia; 5) hemophagocytosis; 6) low or
count (to rule out associated systemic
absent NK function; 7) elevated ferritin; 8)
involvement), complete metabolic panel (to
elevated soluble CD25 (IL2). Newer guide­
rule out p ­otential pituitary involvement),
lines also suggest additional helpful criteria
ESR (as a potential tumor marker), and a uri­
including hepatitis and hyponatremia.
nalysis for specific gravity to rule out a dilute
The answer is e.
urine with DI. Additional imaging with a full
skeletal survey and bone scan can be done to
2. True or False; Hemophagocytosis is
determine if additional lesions are present
required for the diagnosis of HLH.
which may be more amenable to biopsy.
Explanation: Although hemophagocytosis is
More institutions are doing PET/CT rou­
suggestive of the diagnosis of HLH it is not
tinely to determine if additional lesions are
required for diagnosis. As above, 5 of 8 crite­
present which may not be seen on X‐rays.
ria are useful and may or may not include
Additionally, many are doing MRI brain on
hemophagocytosis. Additionally patients
all patients rather than only in patients with
may be considered to have HLH even if they
symptoms of DI or in those with “special
do not meet the 5 criteria but have sugges­
sites” (i.e., craniofacial bone, ear, eye, central
tive symptoms. The answer is false.
nervous system involvement).
Histiocytic Disorders 293

3. All of the following lab values are con­ d. Asymptomatic solitary bone lesion of
sistent with the diagnosis of HLH EXCEPT: the femur
a. Elevated IL2 e. Asymptomatic solitary bone lesion
b. Elevated ferritin involving the mastoid
c. Low fibrinogen Explanation: Unifocal bone lesions can
d. Low trigylcerides often be monitored or could potentially
e. Low NK activity receive surgical curettage if symptomatic
Explanation: As above, patients may have unless there is involvement of “special sites,”
high triglycerides as a marker of HLH. The specifically bones in the face and anterior/
answer is d. medial cranial fossa which may lead to pitu­
itary involvement and the development of
4. You are seeing a 14-year-old female who diabetes insipidus. Multifocal bone lesions
presents with altered mental status after a have an increased risk of recurrence or pro­
seizure like event. She has fever and a rash. gression without chemotherapy. A solitary
Her inflammatory markers are elevated and bone lesion involving the spine can be
her ferritin is significantly elevated. What is asymptomatic and be monitored but also
the most likely underlying diagnosis? may lead to soft tissue swelling and spinal
a. Systemic onset JIA compression necessitating therapy. Systemic
b. Systemic lupus erythematosus LCH has a much worse prognosis and
c. Sepsis involves multiple organs including the skin,
d. Macrophage activation syndrome liver, spleen or lungs. Young children may
e. Hodgkin lymphoma manifest the rash of LCH as a severe
Explanation: The patient appears to have ­sebhorrheic dermatitis (i.e. cradle cap) that
enough symptoms consistent with second­ does not respond to normal therapy. The
ary HLH or macrophage activation syn­ answer is d.
drome (MAS). MAS can be secondary to
many of the other listed conditions, espe­
cially autoimmune conditions. Sepsis or Suggested reading
viral infections may also trigger HLH or
­secondary HLH (i.e., MAS). Additionally, Emile, J.F., Abla, O., Fraitag, S. et al. (2016).
neoplastic conditions may present with Revised classification of histiocytosis and
HLH or lead to secondary development of neoplasms of the macrophage‐dendritic cell
HLH (i.e., anaplastic large cell lymphoma, lineages. Blood 127: 2672–2681.
other non‐Hodgkin lymphomas, Hodgkin Filipovich, A., McClain, K., and Grom, A. (2010).
lymphoma). The answer is d. Histiocytic disorders: recent insights into
pathophysiology and practical guidelines.
5. All of the following potential presenta­ Biol. Blood Marrow Transplant. 16: S82–S89.
Haupt, R., Minkov, M., Astigarraga, I. et al.
tions of Langerhans cell histiocytosis (LCH)
(2013). Langerhans cell histiocytosis (LCH):
require chemotherapy or radiation therapy guidelines for diagnosis, clinical work‐up, and
EXCEPT: treatment for patients till the age of 18 years.
a. Multifocal bone LCH Pediatr. Blood Cancer 60: 175–184.
b. Systemic LCH Jordan, M.B., Allen, C.E., Weitzman, S. et al.
c. Spinal cord compression with verte­ (2011). How I treat hemophagocytic lympho­
bra plana histiocytosis. Blood 118: 4041–4052.
24 Hematopoietic Stem
Cell Transplantation
Hematopoietic stem cell transplantation is generally recommended for hematologic
(HSCT) has become increasingly accepted as a malignancies due to the underlying bone
therapeutic modality for a variety of malignant marrow involvement (Table 24.1). In addi-
and nonmalignant conditions. HSCT involves tion to the effectiveness of high‐dose chemo-
the full or partial ablation of the recipient’s bone therapy in tumor killing, there is b ­ enefit of
marrow with high doses of chemotherapy as a “graft‐versus‐leukemia” (or lymphoma)
well as, in some cases, radiation ­therapy in effect in allogeneic transplant in which the
order to allow engraftment of the donor’s stem immunosensitized donor T‐cells can theoreti-
cells. Collected stem cells can either be allo- cally kill residual malignant cells. By contrast,
geneic (from a separate donor) or autologous autologous transplant is generally reserved
(from the patient). The rationale for HSCT is for patients with solid tumors and no evi-
based on the logarithmic dose–response curve dence of bone marrow involvement as part of
for many chemotherapeutic agents: a much a standard regimen or after failure of stand-
higher dose effectively increases tumor killing ard chemotherapeutic regimens (Table 24.2).
at the cost of p ­ rofound myelosuppression. Chimeric antigen receptor T‐cell (CAR‐T)
Secondarily, multidrug therapy is required to therapy is a novel methodology to re-engineer
overcome resistance and heterogeneity within the patient’s T‐cells to recognize a particular
the malignant cell population. And finally, for target, such as CD19+ acute lymphoblastic
those undergoing allogeneic transplantation, leukemia (ALL) cells. CAR‐T cells can be
the addition of a new immune system allows reinfused to the patient in an autologous
for potential improved tumor surveillance fashion after a lymphodepleting preparative
after donor cell engraftment. chemotherapeutic regimen.
Recommendations for allogeneic HSCT
in nonmalignant conditions are summarized
Transplantable conditions in Table 24.3. Genetic engineering (i.e., gene
therapy) is being used in an increasing
Diseases treated using HSCT can be divided ­number of nonmalignant conditions, whereby
into malignant and nonmalignant conditions. altered nucleated cells are reintroduced to
Recommendations for malignant conditions the patient after a myeloablative preparative
are continually being updated due to changes regimen, often single agent busulfan. Studies
in the effectiveness of standard chemother- are ongoing in the utilization of this technology
apy and the risks and benefits of HSCT. In in sickle cell disease, transfusion‐dependent
pediatric patients, allogeneic transplantation thalassemia as well as hemophilia.

Handbook of Pediatric Hematology and Oncology – Children’s Hospital & Research Center Oakland,
Third Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
© 2021 John Wiley & Sons Ltd. Published 2021 by John Wiley & Sons Ltd.
296 Chapter 24

v­ olume of bone marrow required to ensure


Table 24.1 Malignant conditions potentially
benefiting from allogeneic transplant.
successful engraftment of donor cells is
10–20 ml/kg of recipient body weight.
Acute lymphoblastic leukemia (ALL) in first Younger donors have a higher proportion
remission and high risk for relapse of marrow repopulating cells. Peripheral
Relapsed ALL in second or subsequent blood stem cells are ­collected via apheresis
remission and identified by the presence of a cell sur-
Acute myelogenous leukemia (AML) with
face marker, CD34 (CD, cluster of differen-
high‐risk features in first remission
tiation). In general, 5 × 106 CD34+ cells/kg
Acute myelogenous leukemia (AML) in
second or subsequent remission
are required to ensure engraftment. For
Juvenile myelomonocytic leukemia (JMML) allogeneic transplant, the physician must
Hodgkin or non‐Hodgkin lymphoma in balance the risks, benefits, and availability
second or subsequent partial or complete of cells derived from bone marrow, periph-
remission eral blood, and UCB. Peripheral blood has
Myelodysplastic syndromes (MDS) the highest yield of CD34+ cells (with G‐CSF
Relapsed, refractory, or familial mobilization), whereas UCB has the lowest
hemophagocytic lymphohistiocytosis (due to the ­volume of UCB). The risk of
graft‐versus‐host disease (GVHD) is lowest
after UCB transplantation and highest
after peripheral blood stem cell transplant
Table 24.2 Malignant conditions potentially (PBSCT). Engraftment of donor cells occurs
benefiting from autologous transplant. earliest after PBSCT. Availability of alloge-
neic stem cells is often limited by the lack of
High‐risk neuroblastoma
an eligible donor.
High‐risk brain tumors (medulloblastoma/
PNET)
Metastatic retinoblastoma
Recurrent high‐risk germ cell tumors Donor matching in allogeneic
Relapsed Hodgkin or non‐Hodgkin transplantation
lymphoma
Relapsed Wilms tumor The major histocompatibility complex
(MHC) is a large genomic region on chro-
Abbreviation: PNET, primitive neuroecto­
mosome 6 that encodes the human leukocyte
dermal tumor.
antigen (HLA) system. MHC is vital for the
immune system to recognize self versus non‐
self and varies greatly between individuals.
Types of transplantation MHC is divided into two major classes, class
I and class II. MHC class I molecules are
Stem cells for allogeneic transplantation are found on nucleated cells and present the
collected from bone marrow, peripheral MHC to cytotoxic T‐cells and natural killer
blood, or umbilical cord blood (UCB), cells. MHC class I molecules include HLA‐A,
whereas stem cells for autologous trans- HLA‐B, and HLA‐C. MHC class II mole-
plantation are collected most commonly cules are located on antigen‐presenting cells
from peripheral blood. Collection of (B‐cells and macrophages) and present the
stem cells from peripheral blood occurs MHC to T helper cells. MHC class II includes
after mobilization with granulocyte col- HLA‐DP, HLA‐DQ, and HLA‐DR. Many
ony‐stimulating factor (G‐CSF). The usual other minor histocompatibility complexes
Hematopoietic Stem Cell Transplantation 297

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, familial HLH
Hematologic disorders Sickle cell disease, β‐thalassemia major, Fanconi
anemia, Shwachman–Diamond syndrome,
Diamond–Blackfan anemia, dyskeratosis congenita,
chronic granulomatous disease, Chédiak–Higashi
syndrome, leukocyte adhesion deficiency
Metabolic disorders Storage diseases, lysosomal diseases, mucolipidosis,
mucopolysaccharidoses
Acquired syndromes
Severe aplastic anemia
Paroxysmal nocturnal hemoglobinuria

Abbreviations: HLH, hemophagocytic lymphohistiocytosis; SCID, severe combined immunodefi-


ciency syndrome.

are important in the immune response; how- but novel techniques such as the addition of
ever, current donor matching for HSCT is post‐transplant cyclophosphamide as well as
limited to class I and II molecules. depletion of donor αβT‐cells have both been
Identification of a suitable donor requires successful modalities to mitigate GVHD
matching the recipient’s MHC class I and II from cytotoxic T‐cells and T helper cells.
antigens with those of the donor. Greater dis- Currently, HLA matching is limited to
parity between donor and recipient leads to HLA‐A, HLA‐B, HLA‐C, HLA‐DR, and
increasing risk of rejection of the donor cells HLA‐DQ. National Marrow Donor Program
and, if engraftment occurs, GVHD. Matched retrospective reviews on HLA matching in
family member donor grafts have been found unrelated donor bone marrow transplanta-
to be the least immunogenic. Unfortunately, tion have shown that mismatches at MHC
60–70% of patients will not have a matched class I (HLA‐A, HLA ‐B, and HLA ‐C) and
family donor. In these cases, unrelated volun- MHC class II DR (specifically HLA‐DRB1)
teer donors can be found through bone mar- each had a separate, significant effect on sur-
row donor registries, such as the National vival and risk of GVHD. Additionally, an
Marrow Donor Program in the United States increasing number of mismatches led to
and Bone Marrow Donors Worldwide that decreasing survival. HLA‐DQB1 was also
coordinates multiple worldwide registries. found to have an additive negative effect in
Due to the unavailability of a matched donor patients with other mismatches. UCB trans-
in many cases, haploidentical transplantation plantation has been shown to not require the
is becoming a more widely studied and uti- same level of HLA matching, and matching
lized approach due to ready availability of a is limited to HLA‐A, HLA‐B, and HLA‐DR.
parent or sibling with this level of matching. UCB transplantation is thought to be less
In the past, ­ haploidentical transplantation immunogenic secondary to the decreased
would have invariably led to severe GVHD, alloreactivity of these immature cells.
298 Chapter 24

Ultimately, the physician must balance the percentage of donor and recipient cells and
availability of a matched donor with other thus also help determine marrow engraft-
factors such as the patient’s disease stage, ment or primary graft failure. Factors in
remission status, and general condition. In graft failure include: a nonmyeloablative
most cases, disease stage has a greater impact preparative regimen, insufficient volume of
on survival than the level of HLA mismatch. stem cells (i.e., UCB transplant), increased
immunogenicity due to mismatched HSCT,
and the use of myelosuppressive agents (i.e.,
Pretransplant preparative medications for GVHD). Secondary graft
regimens failure (graft rejection) occurs after the ini-
tial wave of engraftment secondary to the
High‐dose chemotherapy, with or without continued presence of recipient cytotoxic
radiation, is administered in order to maxi- T‐cells. Chimerism studies must again be
mize tumor killing and, in patients undergo- utilized to show that the majority of cells are
ing allogeneic transplant, to affect a of host origin. Other causes of graft failure
sufficient amount of immunosuppression to including infection and recurrence of an
overcome recipient rejection of the HSCT. underlying hematological malignancy
Commonly used conditioning agents, their should be ruled out.
mode of action, and potential side effects are
summarized in Table 24.4.
Complications of hematopoietic
stem cell transplantation
Engraftment and graft failure
HSCT recipients have a unique set of poten-
Evidence of donor replacement of the recipi- tial complications that must be well under-
ent’s bone marrow begins with increasing stood when caring for these patients. The
white blood cell counts and decreasing trans- pretransplant preparative regimen can lead
fusion dependency. Engraftment is defined to to a number of post‐transplant complica-
occur when the transplant recipient achieves tions as outlined in Table 24.4. A com-
three consecutive days of an absolute neutro- mon complication is veno‐occlusive disease
phil count (ANC) > 0.5 × 109/l. Due to the (VOD) of the liver, now known as sinusoi-
­volume of CD34+ stem cells, engraftment of dal obstructive syndrome (SOS). The pro-
donor cells occurs earliest after PBSCT and found immunosuppression that occurs
latest after UCB transplantation. both before and after engraftment puts
Engraftment syndrome, characterized by HSCT patients at high risk for infection
fever, rash, as well as pulmonary symptoms with a variety of organisms. Finally, immu-
and weight gain (secondary to capillary nogenic donor T‐cells can lead to GVHD
leak), may take place during the initial rapid due to the recognition of host alloantigens
rise in the white blood cell count. After as foreign. Multiple late sequelae of HSCT
infection has been ruled out, a short course must also be considered as summarized in
of intravenous steroid therapy can alleviate Table 24.11.
symptoms.
Primary graft failure is defined as a Infections
lack of engraftment within 6 weeks after Practitioners must be cognizant of the
­transplant. Chimerism studies, either of the many potential infectious complications
peripheral blood or bone marrow, measure in the HSCT patient. The duration of
Hematopoietic Stem Cell Transplantation 299

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,


elimination of T‐cells anaphylaxis, serum sickness
BCNU, Alkylation leading to DNA Nausea and vomiting, pulmonary
bischloroethylnitrosurea damage infiltrates and fibrosis,
(Carmustine) transaminitis, nephrotoxicity
Busulfan Alkylating agent Nausea and vomiting, electrolyte
abnormalities, seizures,
mucositis, alopecia,
hyperpigmentation, sterility
Carboplatin Inhibits DNA synthesis by Nausea and vomiting, type I
forming hypersensitivity, renal
DNA cross‐links impairment and electrolyte
wasting, ototoxicity
Cyclophosphamide Alkylating agent, inhibition Fluid retention (SIADH),
of T‐cell replication hemorrhagic cystitis, nausea,
vomiting, and anorexia,
cardiomyopathy, sterility
Etoposide Inhibits topoisomerase II Hypotension, nausea, skin blisters
causing DNA strand or erythema, nephropathy,
breakage hemorrhagic cystitis, alopecia,
stomatitis, 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.

severe neutropenia (ANC < 0.5 × 109/l) though the white blood cell count recovers
and time to engraftment are significant in weeks after transplant, functional
risk factors, as the large majority of recovery of humoral and cellular immu-
patients will have a documented infection nity takes months to years depending on
after 4–5 weeks of neutropenia. Even the type of transplant (autologous vs.
300 Chapter 24

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 Candida 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.

a­ llogeneic) as well as the continued use of hepatic vessels. SOS typically occurs in the
immunosuppressive agents to prevent or first 21 days after allogeneic transplant and
treat GVHD. The presence of a central presents with weight gain, jaundice, and
venous catheter is a secondary risk factor. hepatomegaly. Reversal of portal flow on
Infections tend to occur at different times Doppler ultrasound of the liver is pathogno-
after transplant as outlined in Table 24.5. monic for SOS, though not required for
Workup of fever in the post‐transplant diagnosis. In the past, the treatment of SOS
period is outlined in Table 24.6. was supportive, although now early initia-
Engraftment syndrome, GVHD, and med- tion of defibrotide is considered standard of
ications such as G‐CSF can all be causes of care; general guidelines are outlined in
fever in the post‐transplant period but are Table 24.7.
diagnoses of exclusion. Clostridium diffi-
cile should be considered in the patient Graft‐versus‐host disease
with associated symptoms such as fever, GVHD occurs due to the proliferation of
abdominal pain, and diarrhea. Infection donor T‐cells that recognize host antigen as
prophylaxis is an important aspect of sup- foreign. Direct effects of the T‐lymphocytes
portive care and is outlined in more detail and cytokine response lead to the signs and
in the following text (and Table 24.12). symptoms of GVHD. Acute GVHD can
begin within weeks of transplantation;
Veno‐occlusive disease/sinusoidal chronic GVHD is defined as GVHD lasting
obstructive syndrome beyond 100 days after transplant. Signs and
VOD, now referred to as SOS, results from symptoms of acute and chronic GVHD are
hepatic injury caused by high‐dose chemo- summarized in Table 24.8, grading of
therapy (especially alkylators) and total GVHD is outlined in Table 24.9, and proph-
body irradiation leading to fibrosis of small ylaxis and treatment are described in
Hematopoietic Stem Cell Transplantation 301

Table 24.6 Evaluation and empiric treatment of fever in the post‐transplant period.

Rule out bacterial infection Daily blood cultures from central catheter (aerobic and
anaerobic) while febrile
Consider non‐catheterized 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., cefepime)
If with prolonged fevers (i.e., ≥5–7 days), consider switching
empiric cephalosporin to carbapenem for broader coverage
Addition of an echinocandin (i.e., micafungin) or broad‐spectrum
azole (i.e., voriconazole) for broader fungal coverage with
prolonged fevers (i.e., ≥3–7 days)
Consider vancomycin for staphylococcal and streptococcal
coverage if patient worsening clinically
Directed antimicrobial coverage or further studies based on
positive workup

Abbreviations: CT, computed tomography; CMV, cytomegalovirus; HHV‐6, human herpesvirus 6;


EBV, Epstein–Barr virus; CXR, chest radiography.

Table 24.7 Treatments for sinusoidal obstructive syndrome (SOS).

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 [twice daily] PO
[orally])
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 Defibrotide has fibrinolytic and antithrombotic properties, and has
shown therapeutic benefit in SOS. Patients must not receive other
thrombolytic therapies. Defibrotide is given at 25 mg/kg/day div Q6 h
IV for at least 21 days
302 Chapter 24

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 or
corneal inflammation caused by dryness)
Pulmonary Interstitial pneumonitis, bronchiolitis obliterans
Other Arthritis, immunologic abnormalities

Abbreviations: GVHD, graft‐versus‐host disease; GI, gastrointestinal.

Table 24.9 Grading of graft‐versus‐host disease.

Clinical Skin Liver (TB, Diarrhea (children; Diarrhea (adults; ≥70 kg)
grade mg/dl) <70 kg) (ml/kg/day) (ml/day)

I <25% involvement, 1.5–3.0 10–15 500–1000, nausea/vomiting


maculopapular
II 25–50% involvement, 3.0–6.0 16–20 1000–1500, nausea/vomiting
maculopapular
III >50% involvement, 6.0–15.0 21–25 >1500, nausea/vomiting
maculopapular or
generalized
erythroderma
IV Desquamation/bullae >15 >25, pain/ileus >2500, pain/ileus

Abbreviation: TB, total bilirubin; adapted from Lanzkowsky (2011).


Source: Based on Lanzkowsky P. Hematopoietic stem cell transplantation. In: Manual of Pediatric
Hematology and Oncology, 5th ed. New York: Elsevier, 2011.

Table 24.10. Patients should be checked acute and chronic GVHD through nonspe-
daily for signs and symptoms of acute cific T‐cell suppression. Multiple newer
GVHD while hospitalized following HSCT. agents have been trialed recently for patients
Medications including cyclophosphamide, with GVHD refractory to steroids or to
methotrexate, cyclosporine, tacrolimus, and spare steroids given the multitude of side
sirolimus are used for prevention of GVHD effects. Ibrutinib and ruxolitinib were both
through inhibition of T‐cell replication. recently approved for steroid‐refractory
Steroids remain the mainstay of treatment of GVHD and both lead to profound T‐cell
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
Cyclophosphamide Selective depletion of Fluid retention (SIADH),
alloreactive donor and host hemorrhagic cystitis, nausea,
T‐cells vomiting, and anorexia,
cardiomyopathy, sterility
Methotrexate Cell cycle specific; inhibits Transaminitis, mucositis, renal
T‐cells division insufficiency, 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
mechanism not well sugars, increased appetite,
understood insomnia, mood swings, acne,
truncal obesity
Mycophenolate mofetil Inhibits T‐cell proliferative Nausea, vomiting, anorexia, bone
(MMF) response marrow suppression, multiple
others
Anti‐thymocyte Alteration of function and Fever and chills, pruritus,
globulin (ATG) elimination of T‐cells anaphylaxis, serum sickness
Sirolimus Blockage of mTOR leading to Peripheral edema,
inhibition of T‐ and B‐cells hypertriglyceridemia,
hypertension,
hypercholesterolemia,
increased creatinine
Etanercept/infliximab TNFα inhibitors; consideration Severe immunosuppression
in refractory GVHD
Mesenchymal stem cells Immunomodulation with Fever, infection, organ
suppression of T‐cell dysfunction
proliferation
Subcutaneous IL2 Immunomodulation through Immune suppression, infection,
expansion of T‐regulatory cells local reaction
Extracorporeal Leukocyte apoptosis; cytokine Immune suppression, infection,
photopheresis modulation, tolerogenic anemia
response
Ruxolitinib JAK1/2 inhibitor; T‐cell Immune suppression, infection,
suppression neutropenia,
thrombocytopenia
Ibrutinib Bruton’s tyrosine kinase and Infection, bleeding, diarrhea,
IL‐2 inducible T‐cell kinase cough
inhibitor; B‐cell and CD4
T‐cell suppression

Abbreviations: SIADH, syndrome of inappropriate antidiuretic hormone;TNF, tumor necrosis


factor; GVHD, graft‐versus‐host disease.
304 Chapter 24

suppression. Immunomodulation through Transplant‐associated


stimulation of the T‐regulatory cell popula- thrombotic microangiopathy
tion has more recently been found to be an (TA‐TMA)
important way to mitigate GVHD through TA‐TMA occurs due to small vessel
inhibition of the T‐effector cell populations. endothelial injury and typically occurs sec-
Methods to do this include subcutaneous ondary to calcineurin inhibitors (i.e., cyclo-
IL2 and extracorporeal photopheresis. sporine, tacrolimus) for immunosuppression
The most common acute symptoms of after allogeneic transplantation, though may
GVHD are related to the integument and also occur after autologous transplantation
gastrointestinal systems. Skin findings are for unclear reasons. Typical symptoms of
often nonspecific with the development of TA‐TMA include unremitting hyperten-
an erythematous, pruritic, maculopapular sion, kidney injury leading to proteinuria, as
rash. This finding in conjunction with white well as signs and symptoms of microangio-
blood cell count recovery can be a harbinger pathic hemolytic anemia with elevation in
of acute GVHD, though generally skin LDH, presence of schistocytes on peripheral
biopsy should be performed for confirma- blood smear, as well as activation of termi-
tion. Additional diagnostic considerations nal complement (sC5b‐C9). Early diagnosis
including viral infection and drug reaction is vital for appropriate management of
should be ruled out. Gastrointestinal symp- TA‐TMA.
toms are often lower‐tract‐related including
abdominal pain and diarrhea. Bloody stool Late sequelae
is possible, though C. difficile and other Multiple late effects must be considered and
stool pathogens should be sought. Upper are summarized in Table 24.11.
tract symptoms including nausea and ano-
rexia may also be present. Additionally, eti-
ologies including infection, mucositis, and
post‐transplant anorexia should be Supportive care in transplant
considered. patients

Idiopathic pneumonia syndrome Routine care for the HSCT patient is a


(IPS) unique and vital aspect of preventing infec-
IPS is severe, non‐infectious lung injury tions and a multitude of other potential
that occurs post‐transplant. Immune acti- complications including bleeding, transfu-
vation leading to progressive inflammation sion‐associated GVHD, and cytomegalovi-
is the underlying driver for IPS and rus (CMV) reactivation, as well as ABO
explains the benefit of blockade of tumor incompatibility between recipient and
necrosis factor (TNFα) with etanercept. donor.
Evidence of widespread alveolar injury
without an infectious etiology is the hall- Infection prophylaxis
mark of IPS. A myeloablative preparative Due to profound immunosuppression,
regimen with total body irradiation (TBI) HSCT patients are at risk for reactivation of
and acute GVHD are risk factors for the latent viral infections in addition to fungal
development of IPS. Rapid progression of and bacterial infection. Guidelines for infec-
symptoms is common with IPS with a high tion prophylaxis and surveillance are out-
mortality rate. lined in Table 24.12.
Hematopoietic Stem Cell Transplantation 305

Table 24.11 Late effects of hematopoietic stem cell transplantation.

Late effect Underlying cause

Endocrine disorders (gonadal failure, delayed TBI


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
Post‐transplant lymphoproliferative disorder Immunosuppressive therapy
Poor dentinogenesis (in young children) TBI
Decreased bone mineral density Multiple factors

Abbreviations: GVHD, graft‐versus‐host disease; TBI, total body irradiation; ATG, anti‐thymocyte
globulin; IT, intrathecal; adapted from Lanzkowsky (2011).
Source: Based on Lanzkowsky P. Hematopoietic stem cell transplantation. In: Manual of Pediatric
Hematology and Oncology, 5th ed. New York: Elsevier, 2011.

Diet post‐transplant anorexia, many centers have


Due to the pretransplant preparative regi- liberalized this practice.
men, patients will have an extended period
of anorexia requiring total parenteral nutri- Transfusion guidelines
tion (TPN). Triglycerides should be fol- Due to immunosuppression, all HSCT
lowed weekly while on TPN in addition to patients should receive irradiated blood
routine monitoring of electrolytes. Trace products in order to eliminate the risk of
elements should be included in the TPN. If transfusion‐associated GVHD. In addi-
possible, the patient should be transitioned tion, packed red blood cell (PRBC) trans-
to enteral feeds through a nasogastric tube. fusions should be CMV‐negative for HSCT
Once the patient is eating, it is vital that the recipients who are identified as CMV
patient’s family be aware of the bone marrow seronegative during the pretransplant
transplant low microbial diet of acceptable period. CMV seronegative platelets are not
foods and appropriate methods of handling, routinely required, as leukofiltered plate-
preparation, and storage in order to prevent lets are considered sufficiently leukore-
infection from food‐borne pathogens. Due duced to prevent transmission of CMV. In
to the lack of evidence on the need for a sig- order to diminish the risk of spontaneous
nificantly restricted diet in the setting of bleeding, the platelet count is kept above
306 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 6 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 immunosuppressive
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 Antipseudomonal cephalosporin (i.e., cefepime) or carbapenem (i.e.,
infection 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 Weekly to three times weekly blood culture if on steroids and afebrile
infection (controversial)
Pneumonia Weekly chest radiograph (controversial)
Deficient IgG Monthly quantitative IgG; replace with IVIG if IgG < 400 mg/dl

Abbreviations: PCP, Pneumocystis jiroveci pneumonia; TMP–SMX, trimethoprim–sulfamethoxa-


zole; 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.
* Based on institutional preference.

20 × 109/l following HSCT. This threshold bank maintain this information on file to
is increased to above 50 × 109/l in certain ensure proper ­transition of blood type pre-
cases, including patients with sickle cell transplant and post‐transplant.
disease, brain tumor patients, and patients
with severe mucositis, SOS, or problems
with hemostasis (e.g., epistaxis and GI Immune reconstitution
bleeding). A lower threshold of 10 × 109/l
can be utilized in stable patients who are Even with the recovery of white blood
out of the immediate post‐transplant cell counts and specifically lymphocyte
period. Hemoglobin levels are usually kept counts, HSCT patients take months to
above 8–9 mg/dl depending on institu- years to recover immune function. Loss of
tional practice. Platelet transfusions should memory B‐lymphocytes due to the pre-
be limited to 1 pheresed unit (see transplant preparative regimen leads to
Chapter 5). ABO blood types of both the loss of antibody secondary to vaccination
donor and recipient must be considered and lifetime environmental exposures.
when transfusing platelets and PRBCs. Immune recovery is variable but generally
Appropriate transfusion in ABO‐incom- occurs once the patient is off immunosup-
patible donor and recipient is summarized pressant therapy, and therefore is ­generally
in Table 24.13. It is critical that the blood earlier in patients undergoing autologous
Hematopoietic Stem Cell Transplantation 307

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, Measures response to CMV, HSV, and VZV,
VZV) and defines length of acyclovir prophylaxis
Mitogens (Concanavalin A, Concanavalin A measures T‐ and B‐cell
pokeweed, function
phytohemagglutinin)
Pokeweed measures B‐cell function
Phytohemagglutinin measures T‐cell
function, must be reactive before PCP
prophylaxis is discontinued
Antigens (tetanus toxoid, Lack of response to tetanus toxoid indicates
Candida albicans) 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 conjugated vaccines

Abbreviations: CMV, cytomegalovirus; HSV, herpes simplex virus; VZV, varicella zoster virus;
PCP, Pneumocystis jiroveci pneumonia; PRP, polyribose phosphate.

transplantation. Once there is sufficient begin once the absolute lymphocyte count is
evidence of immune reconstitution, HSCT >1 × 109/l and generally when the patient
patients will proceed with revaccination. is off immunosuppressive therapy, are
Studies of immune reconstitution, which summarized in Table 24.14.
308 Chapter 24

cefepime (a fourth‐generation cephalo-


Case study for review
sporin). Fungal infection must be seriously
considered with testing for Aspergillus by
A 14‐year‐old boy is on the bone marrow
serum galactomannan at a minimum.
transplant unit after receiving a HSCT for
Fluconazole will cover Candida albicans but
acute myelogenous leukemia (AML) in
not other candidal species or Aspergillus.
remission. His peripheral blood stem cell
You should consider discontinuing flucona-
reinfusion took place 10 days ago. He has
zole and starting an antifungal agent with a
been having daily fevers up to 39.5 °C for
broader spectrum of activity such as an echi-
3 days with negative blood cultures to date.
nocandin (i.e., micafungin). Micafungin has
He is not in any respiratory distress and his
a broader spectrum of activity than flucona-
physical exam is unremarkable except for a
zole, but only has moderate coverage against
mild rash. His complete blood count is
Aspergillus and does not cover most mold
remarkable for a WBC count that has been
species. Therefore, antimold coverage with
increasing, most recently 0.2 × 10 /l. He has
9
voriconazole or posaconazole is the better
a double lumen central catheter and is on
choice for empiric coverage in lieu of an
total parenteral nutrition. He has moderate‐
echinocandin. Finally, the addition of ster-
to‐severe mucositis that is improving. His
oids for acute graft‐versus‐host disease with
weight has been stable, and his chemistries
fever and rash should be a consideration if
are otherwise unremarkable. His medica-
infection is relatively well ruled out after
tions include cefepime as well as prophylac-
negative blood cultures and chest
tic fluconazole and acyclovir.
radiography.
a. What is the differential for fever at
After a few days, the patient defervesces
this point after HSCT?
but has developed an increase in his bilirubin.
Differential diagnosis:
The infectious workup has been negative to
●● Bacterial infection (Gram‐negative rods,
date. On looking at his fluid status for the
S. epidermidis, Streptococcus viridans species).
last 48 hours, you note that he is 2 liters posi-
●● Viral infection (CMV, herpes simplex I).
tive and his weight has increased by 2 kg
●● Fungal infection (Candida, Aspergillus).
from the time of his transplant.
●● Graft‐versus‐host disease.
c. What is the differential diagnosis for
●● Mucositis.
hyperbilirubinemia and weight gain at
b. What medications would you con-
this point after transplant?
sider adding based on the above men-
Differential diagnosis:
tioned differential?
●● Sinusoidal obstructive syndrome.
The patient is well‐covered for Gram‐­
●● Viral infection.
negative infections with cefepime. Gram‐
●● Acute graft‐versus‐host disease.
positive infections usually lead to a positive
●● Heart failure (secondary to infection,
blood culture, although vancomycin
fluid overload, or previous anthracycline or
should be considered if the patient is clini-
mediastinal radiation therapy).
cally worsening in the face of a negative
At this time point after transplant, with a
blood culture (i.e., respiratory distress or
negative infectious workup, SOS is the most
hypotension). The patient is at risk for S. epi-
likely diagnosis. If the patient is having
dermidis secondary to the central catheter
increasing WBC counts with rash and diar-
and S. viridans secondary to mucositis, nei-
rhea, acute GVHD is possible but should
ther of which is adequately covered with
Hematopoietic Stem Cell Transplantation 309

be a diagnosis of exclusion. Heart failure is a conditions. Although transplantation is


possibility secondary to toxicity from ther- required as part of the multi‐modal treat-
apy or an underlying infection and should ment for neuroblastoma, this can be done
be ruled out with echocardiogram. with autologous transplant rather than allo-
d. What changes in therapy should you geneic. The answer is d.
institute?
Most patients with SOS ultimately recover 2. All of the following are potential condi-
with supportive care alone. Fluid and tions requiring autologous transplantation
sodium restriction should be initiated. An (i.e., high dose chemotherapy with stem cell
abdominal ultrasound with elastography rescue) EXCEPT:
and Doppler flow should be undertaken to a. Relapsed Hodgkin lymphoma
look for hepatic fibrosis and changes in por- b. High‐risk medulloblastoma
tal venous flow. Antioxidants including c. Metastatic osteosarcoma
vitamin C, E, and selenium should be d. High‐risk neuroblastoma
added to the TPN for liver protection. If e. Relapsed Wilms tumor
the patient develops increasing weight Explanation: Many pediatric solid tumors in
gain and liver dysfunction (evidence of relapse or with very high‐risk disease bene-
developing SOS), early intervention with fit from high‐dose chemotherapy with
defibrotide should be instituted. stem cell rescue (i.e., autologous transplan-
tation). Sarcomas on the other hand includ-
ing osteosarcoma and rhabdomyosarcoma
Multiple choice questions have not shown benefit from autologous
transplantation. Although still somewhat
1. All of the following are potential indica- controversial most centers also agree that
tions for allogeneic hematopoietic stem cell transplant is not beneficial for Ewing sar-
transplantation EXCEPT: coma. The answer is c.
a. Relapsed acute lymphoblastic leuke-
mia (ALL) 3. All of the following are potentially concern-
b. High risk acute myelogenous leuke- ing for the development of vaso‐occlusive
mia (AML) disease (VOD; sinusoidal obstructive syn-
c. Severe aplastic anemia drome, SOS) EXCEPT:
d. High‐risk neuroblastoma a. Refractory thrombocytopenia
e. Sickle cell anemia b. White blood cell engraftment
Explanation: Conditions with underlying c. Weight gain
bone marrow disease or dysfunction should d. Hyperbilirubinemia
routinely undergo allogeneic rather than e. Hepatomegaly with reversal of portal
autologous transplantation in pediatric flow
patients. These conditions include leuke- Explanation: VOD occurs due to the pre-
mias as well as myelodysplastic syndromes, transplant chemotherapy which leads to
hemophagocytic lymphohistiocytosis (HLH), fibrosis at the sinusoidal level causing
and non‐malignant conditions including obstruction to returning blood flow which
aplastic anemia, sickle cell anemia, severe manifests as hepatomegaly, weight gain
congenital neutropenia, beta‐thalassemia due to decreased liver synthetic function
major, alpha‐thalassemia major, and of oncotic proteins, hyperbilirubinemia
severe immunodeficiencies, amongst other and transaminitis. Due to the reversal of
310 Chapter 24

portal flow there is resultant splenomegaly 5. Which of the following statements is


with trapping of platelets leading to refrac- FALSE?
tory thrombocytopenia. Patients with exist- a. Time to engraftment is shortest with
ing liver disease prior to transplant are at umbilical cord blood transplantation
higher risk of developing VOD. (versus peripheral blood and bone
Additionally, alky­lator therapy with medi- marrow)
cations including busulfan, cyclophospha- b. Risk of graft‐versus‐host disease
mide and melphalan put the patient at (GVHD) is highest after peripheral
higher risk. Generally supportive care with blood stem cell transplant (versus bone
fluid restriction, gentle diuresis with aldac- marrow and umbilical cord blood)
tone, platelet and red cell transfusion, urso- c. Risk of failure to engraft is highest
diol and antioxidants is sufficient to allow after umbilical cord blood transplant
the liver to heal. In rare cases a more novel (versus peripheral blood and bone
medication defibrotide must be utilized. marrow)
The answer is b. d. Risk of GVHD is directly related to
the amount of HLA mismatch between
4. All of the following are potential post- donor and recipient
transplant complications after autologous e. Risk of veno‐occlusive disease is
transplant EXCEPT: highest after alkylator preparative
a. Veno‐occlusive disease chemotherapy
b. Lack of engraftment Explanation: Stem cell source, stem cell size
c. Infection—viral, bacterial, fungal and mismatch between donor and recipient
d. Mucositis are important considerations when deter-
e. Infertility mining risk of posttransplant complica-
Explanation: All of the above are potential tions. Umbilical cord blood units have the
posttransplant complications, some though lowest total nucleated cell (TNC) count and
are specific for allogeneic rather than therefore time to engraftment is the longest
autologous transplantation. Veno‐occlusive (peripheral blood is the fastest due to the
disease is a complication specific to the pre- highest TNC count and highest CD34+
parative chemotherapy regimen, especially cells/kg). Goal stem cell dose for bone mar-
those that include alkylator therapy and row and peripheral blood is 5 × 106 CD34+
can be seen with both autologous and allo- cells/kg. Due to the maturity of the stem
genic transplant. Since the patient gets cells, GVHD is highest after peripheral
their own stem cells back at a high cell dose, blood stem cell transplant and lowest after
lack of engraftment is generally not an issue umbilical blood stem cells (while factoring
with autologous transplant. Infection is in the level of mismatch between donor and
more common with allogeneic transplant recipient). Risk of GVHD is lowest with a
but can be seen after autologous transplant. 10/10 matched sibling (matching entails
Mucositis is common after both allogeneic looking at MHC‐I antigens HLA‐A, ‐B, and
and autologous transplant depending on –C as well as MHC‐II antigens HLA‐DR
the intensity of the pretransplant chemo- and –DQ) and risk increases with mismatch
therapy regimen. Similarly, infertility is (in addition to peripheral blood stem cell
common for both autologous and allogeneic source). As explained previously, existing
transplant and dependent on the intensity liver disease and alkylator preparative
of the pretransplant chemotherapy regimen. chemotherapy are risk factors for VOD. The
The answer is b. answer is a.
Hematopoietic Stem Cell Transplantation 311

veno‐occlusive disease/sinusoidal obstruc-


Suggested reading tion syndrome with multiorgan failure. Int. J.
Hematol. Oncol. 6: 75–93.
Lanzkowsky, P. (2011). Hematopoietic stem cell Sahdev, I. and Abdel‐Azim, H. (2016).
transplantation. In: Manual of Pediatric Hematopoietic stem cell transplantation. In:
Hematology and Oncology, 5e. New York: Lanzkowsky’s Manual of Pediatric Hematology
Elsevier. and Oncology, 6e (eds. P. Lanzkowsky, J. Lipton
Ricahrdson, P.G., Grupp, S.A., Pagliuca, A. et al. and J.D. Fish). New York: Elsevier. 577–604.
(2017). Defibrotide for the treatment of hepatic
25 Supportive Care of the
Child with Cancer
Much of the dramatic improvement in chronically immunosuppressed as a direct
­outcomes in pediatric oncology over the last result of chemotherapy and immune‐modify­
50 years can be attributed to the develop­ ing therapies, and are at times severely myelo­
ment of novel chemotherapeutic agents and suppressed. While on the inpatient unit,
an increase in therapeutic intensity. Under­ patients are exposed to nosocomial organ­
standing, anticipating, and managing the isms as well. Many of these children have cen­
adverse effects and complications of therapy tral venous catheters and may intermittently
lead to optimal supportive care measures, a have mucosal breakdown secondary to chem­
practice now recognized to save lives and otherapy, both factors disrupting the integrity
improve quality of life during and after of the body’s physical defense barriers. Poor
­therapy. The practitioner should be familiar nutrition also plays a significant role in host
with the potential risks of chemotherapy susceptibility. Certain standards of care are
(Chapter 30), guidelines for transfusional indicated to minimize the risk of acquiring
supportive care (Chapter 5), and treatment for infection in these children. Infection prophy­
febrile neutropenia (Chapter 27). Additionally, laxis remains the cornerstone of supportive
practitioners should become familiar with the care in children with malignancies, decreas­
burgeoning novel therapies such as immuno­ ing morbidity and mortality.
therapeutics and their unique side effects such General measures for the prevention of
as cytokine release syndrome, neurotoxicity, infection include avoidance of crowded envi­
and pneumonitis. Here we discuss infection ronments, construction areas and soil (in the
prophylaxis, antiemetic therapy, and the use garden, playground), wearing a mask in pub­
of hematopoietic growth factors. Supportive lic when severely neutropenic (i.e., absolute
care for patients receiving hematopoietic stem neutrophil count [ANC] <0.5 × 109/l), and,
cell transplantation (HSCT) is discussed in most importantly, careful hand washing (by
Chapter 24. the patient and all those who have direct con­
tact). Good nutrition and proper dental
hygiene cannot be overemphasized. Oral
Infection prophylaxis hygiene should include daily brushing (with a
soft brush) and oral rinsing with tap water or
Children receiving chemotherapy for treat­ saline solution. Cleanliness of the perianal
ment of their malignancies are susceptible to area is important, especially in the neutro­
acquiring infection from bacterial, viral, penic state. Constipation should be avoided
­fungal, and protozoal organisms. They are with an age‐appropriate diet, and, if necessary,

Handbook of Pediatric Hematology and Oncology – Children’s Hospital & Research Center Oakland,
Third Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
© 2021 John Wiley & Sons Ltd. Published 2021 by John Wiley & Sons Ltd.
314 Chapter 25

a stool softener (e.g., docusate sodium) or suppression or transaminitis, rather than


laxative (e.g., MiraLAX). Rectal suppositories decreasing the dose of maintenance chemo­
and rectal temperatures should be avoided to therapy for children with acute lymphoblas­
decrease the possibility of a mucosal tear and tic leukemia (ALL).
infection with enteric organisms. Bacterial prophylaxis can be considered
Trimethoprim/sulfamethoxazole (TMP/ in patients receiving high‐intensity chemo­
SMX) prophylaxis is indicated in immuno­ therapy when the ANC is <0.5 × 109/l, though
suppressed children to reduce the risk of further data are required to prove benefit
acquiring Pneumocystis jiroveci pneumonia without significant risk of development of
(PCP) due to T‐lymphocyte dysfunction resistance. We utilize levofloxacin for
secondary to chemotherapy. Current pro­ ­children ≥6 months of age at a dose of 10 mg/
phylactic dosing is 5 mg/kg/day of the TMP kg/dose to a maximum of 250 mg for c­ hildren
component in two divided doses on two to under 5 years and 750 mg for older children.
three successive days per week. Evidence High‐intensity regimens that may benefit
indicates that this regimen also provides from bacterial prophylaxis include relapsed
good general antibacterial prophylaxis, leukemias, acute myelogenous leukemia
although probably more effectively in (AML), high‐risk leukemia during induction
patients given daily prophylaxis, which must therapy, infant ALL, ALL in Down syndrome,
be balanced with potential allergy or intol­ and intensive Head Start chemotherapy for
erance to TMP/SMX and especially second­ brain tumors.
ary to myelosuppression from the therapy. Fungal prophylaxis is indicated in
For patients unable to take TMP/SMX, severely myelosuppressed patients. Mouth
inhaled pentamidine or oral dapsone may care with brushing and oral rinsing is rou­
be given. Pentamidine is preferred, however, tinely recommended for the prevention or
due to the need for a cooperative patient treatment of mouth sores and yeast prolif­
who can appropriately inhale the medica­ eration. Voriconazole (see Formulary for
tion, is usually limited to children above the age‐based dosing) or posaconazole (200 mg
age of 7 years. The dose is 300 mg inhaled on suspension PO TID, three times daily, or
a monthly basis. Dapsone is given orally 300 mg BID, two times daily, tablets in
once weekly at 4 mg/kg, with a maximum patients ≥40 kg) is indicated in patients that
dose of 200 mg. Patients should commence are at very high risk of developing infection
prophylaxis at the initiation of immune sup­ due to periods of prolonged, severe neutro­
pressive therapy and continue until 3 penia (e.g., AML, relapsed ALL patients).
months after completion of therapy to The most common fungal infections in
ensure T‐lymphocyte immune reconstitu­ patients receiving intensive chemotherapy
tion. Prophylaxis with TMP/SMX should be include candidiasis and aspergillosis. Efforts
discontinued one day prior to the adminis­ to prevent invasive fungal disease (especially
tration of high‐dose IV methotrexate (doses Aspergillus sp., an airborne organism)
>1 g/m2) and restarted after the serum include respiratory isolation, laminar air
methotrexate level has fallen to below flow rooms, and high‐efficiency particulate
1 × 10−7 m due to competitive excretion air (HEPA) filters. Patients should not have
between TMP/SMX and methotrexate and live plants in the room, play in dirt or gar­
the risk for delayed methotrexate clearance. dens, or be in proximity to construction
Temporary interruption of TMP/SMX, or a work. Some patients may receive an echino­
change to an alternative therapy, may be candin (e.g., micafungin 1.5–2 mg/kg IV
necessary in the setting of prolonged marrow daily, 50 mg maximum prophylaxis dose) for
Supportive Care of the Child with Cancer 315

prophylactic fungal coverage while inpatient The child exposed to varicella or zoster
with prolonged fevers and severe neutrope­ with known negative varicella immune sta­
nia due to multiple potential drug interac­ tus (negative titers [IgG] and no history of
tions with voriconazole or posaconazole, varicella infection) should receive varicella
most notably vincristine for ALL patients zoster immune globulin (VariZIG) within
(see Chapter 27). Micafungin may also be 96 hours of exposure (of note, VariZIG
given at a treatment dose (i.e., 3–4 mg/kg IV remains an investigational agent in the
daily, max 150 mg) with prolonged fevers. United States and requires institutional
Prophylaxis for subacute bacterial endo­ review board approval and completion of an
carditis is recommended for patients with investigational new drug form). Parents
central venous catheters or surgical hardware should be counseled on the risk of exposure
undergoing invasive procedures that could for the child with negative immune status
cause transient bacteremia and seeding of the and the need for immediate evaluation and
catheter or heart valves. Such interventions treatment if exposed. The dosage is one vial
include dental cleaning or procedures and (125 units) per 10 kg, with a minimum dose
potentially surgery involving the gastrointes­ of 125 units and a maximum of 625 units (5
tinal or genitourinary tract (controversial). vials), intramuscularly. Administration of
The standard regimen is that recommended VariZIG extends the incubation period from
by the American Heart Association for children 14 to 28 days and decreases, but does not
with congenital heart disease: amoxicillin eliminate, the possibility of clinical infection
50 mg/kg (maximum 2 g) orally 1 hour prior with VZV (the incubation period generally
to the procedure. Azithromycin may be given is shortened in the immunocompromised
to penicillin‐allergic patients (15 mg/kg, max patient who does not receive VariZIG). If
500 mg, 1 hour prior to the procedure). VariZIG is not available, then intravenous
immune globulin can be given at 400 mg/kg.
Viral prophylaxis and treatment Myelosuppressive chemotherapy may need
Common viral infections may be particu­ to be stopped 7 days after the exposure and
larly virulent in immunocompromised held until the end of the incubation period.
children. These viruses include varicella
­ The decision to hold chemotherapy during
zoster (VZV), herpes simplex (HSV), cyto­ the incubation period should be based on
megalovirus (CMV), Epstein–Barr virus, the intensity of exposure, condition of the
hepatitis types A and B, respiratory syncytial patient, and intensity of the chemotherapy.
virus, and rubeola (measles). Infection with If >96 hours have passed since exposure, the
these viruses may result in prolonged viral patient should be given acyclovir at 80 mg/
excretion, increased morbidity, or death. At kg/day (max dose 800 mg/dose) divided
the time of diagnosis, an immunization and QID (four times daily) PO (orally) 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 7–10 days, until all
will result in different treatment recommen­ lesions are crusted and no new lesions have
dations depending on the potential for appeared for 24–48 hours. Monitor renal
primary infection versus reactivation. CMV function and fluid status daily due to poten­
serology should also be known in determining tial nephrotoxicity from acyclovir. Of note,
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
316 Chapter 25

healthy recipients rarely occurs. If skin chance of infection has decreased signifi­
lesions occur after vaccination, exposure cantly with herd immunity.
should be avoided until all lesions have Inactivated vaccines (e.g., DTaP, Tdap
crusted over. (both diphtheria‐tetanus‐pertussis vaccines),
Children with a history of recurrent her­ Hepatitis A, Hepatitis B, pneumococcal, Hib,
pes simplex infections are at an increased IPV (inactivated polio vaccine), meningo­
risk of reactivation with subsequent courses coccal, and influenza) can be safely given
of chemotherapy or during and after HSCT. during therapy, although response will be sig­
Acyclovir administered prophylactically can nificantly attenuated based on the level of
prevent or decrease the severity of recurrent immunosuppression. Yearly influenza vacci­
herpes infection; the recommended oral nation is reasonable during therapy, as the
dose is 200 mg TID or QID for children potential benefit of immunization outweighs
above 2 years of age. Immunocompromised the risks, especially for children with ALL in
patients with active HSV infection should maintenance. In order to increase the chance
receive IV acyclovir (30 mg/kg/day or of an appropriate antibody response, vaccina­
1500 mg/m2/day divided q8h). HSCT guide­ tion should be spaced out from chemo­
lines for HSV and VZV prophylaxis are therapy as much as feasible. Due to potential
summarized in Chapter 24. risks of pneumococcal and Hib infection in
patients with Hodgkin lymphoma (HL),
Immunization during some experts recommend vaccination
chemotherapy against these pathogens prior to starting
Patients receiving chemotherapy, or who are therapy. Although not immunocompro­
otherwise immunocompromised, should mised, untreated patients with HL have an
not receive live virus vaccines (Measles‐ underlying B‐lymphocyte dysfunction and
Mumps‐Rubella, MMR), live attenuated the benefits of immunization prior to therapy
influenza vaccine [LAIV; FluMist®], vari­ are often significantly diminished. Evidence
cella, rotavirus, and oral polio). Siblings or is lacking to make firm recommendations.
household contacts should not receive oral Catch‐up immunizations should occur
polio, although MMR, varicella, and rotavi­ after the completion of therapy. Most sources
rus are safe, as transmission is rare. Limited feel that waiting 3–6 months after therapy
information is available regarding risk of completion will produce sufficient immune
transmission with LAIV; since an ­inactivated reconstitution to allow an appropriate
form of influenza vaccination is available, response to inactivated vaccines. Patients who
this should be the preferred immunization had not previously completed their ­primary
in family members as well as health care vaccination series should restart from the
providers. Although varicella vaccination beginning. Patients who previously com­
has been shown to be safe in patients with pleted the primary series can have antibody
ALL in maintenance, the benefits and risks titers drawn to determine what protection
of this immunization should be weighed they may have lost, if any, or receive booster
carefully. Consideration should be given to vaccinations. Children above 5 years of age
the likelihood of developing a protective (not including patients with HL) have a small
response while still on chemotherapy and risk of developing significant disease from
the potential to develop active infection pneumococcus or Hemophilus influenza, but
with administration of a live attenuated vac­ revaccination against these pathogens should
cine. This must be weighed with the signifi­ be considered in all patients after therapy.
cant risks from natural varicella infection in Postvaccination titers can be considered,
the immunocompromised, although the although the likelihood of an insufficient
Supportive Care of the Child with Cancer 317

response is minimal in patients who are emesis occurs at least 24 hours after therapy
vaccinated more than 6 months after
­ has been completed. Poor control of nausea
chemotherapy has been completed. Little and vomiting may prolong, or result in, hos­
evidence exists as to the appropriate and safe pitalization and lead to dehydration and
timeframe to receive live vaccinations after electrolyte abnormalities. The single most
chemotherapy. In general, we recommend important factor in CINV is the emetogenic
waiting 6–12 months after therapy comple­ potential of a particular chemotherapeutic
tion before reimmunizing with MMR and agent, which, in pediatric patients, is also
varicella, depending on intensity of the dose dependent for some drugs (Table 25.1).
chemotherapy received. HSCT immunization Multiple agents are useful for the preven­
guidelines are briefly discussed in Chapter 24. tion and treatment of CINV. Recognition of
the chemoreceptor trigger zone and the
importance of the 5‐HT3 receptor have been
Prevention of chemotherapy‐ instrumental in better controlling CINV.
induced nausea and vomiting Cytotoxic chemotherapy appears to be
­associated with release of local mediators
Antiemetics are a vital component of the such as 5‐HT and substance P from the
supportive care regimen for patients receiv­ enterochromaffin cells of the small intestine.
ing chemotherapy or radiation. There are The release of these local mediators subse­
three types of chemotherapy‐induced nau­ quently stimulates vagal afferents that initi­
sea and vomiting (CINV): (i) anticipatory, ate vomiting. 5‐HT3 receptors and substance
(ii) acute, and (iii) delayed. Anticipatory P receptors (called neurokinin‐1) are located
emesis occurs before chemotherapy is both peripherally (vagal nerve terminals)
administered and may be a result of nausea and centrally in the chemoreceptor trigger
and vomiting experienced during previous zone; thus, 5‐HT3 and substance P antago­
cycles of therapy. Acute emesis occurs nists may have both peripheral and central
within the first 24 hours of therapy; delayed effects in inhibiting vomiting.

Table 25.1 Emetogenic potential of common pediatric chemotherapeutic agents.

High (>90%) Moderate–high Moderate (30–60%) Moderate–low Minimal


(60–90%) (10–30%) (<10%)

Cytarabine Cytarabine Carboplatin Cytarabine Bleomycin


(>1000 mg/m2) (250–1000 mg/m2) (<250 mg/m2)
Cisplatin Cisplatin Cyclophosphamide Etoposide Decadron
(>50 mg/m2) (<50 mg/m2) (<750 mg/m2)
Cyclophosphamide Cyclophosphamide Daunomycin Mercaptopurine Prednisone
(>1500 mg/m2) 750–1500 mg/m2)
Dactinomycin Doxorubicin Methotrexate Fludarabine
(<60 mg/m2) (50–250
mg/m2)
Doxorubicin Idarubicin Topotecan Methotrexate
(>60 mg/m2) (<50 mg/m2)
Methotrexate Ifosfamide Vinblastine Thioguanine
(>250 mg/m2)
Irinotecan Radiation therapy Vincristine
318 Chapter 25

The major pediatric antiemetics, and Granulocyte colony‐stimulating


their specific mechanisms of action and factor
dose, are summarized in Table 25.2. G‐CSF is a lineage‐specific cytokine that
Generally, based on the antiemetic potential stimulates the proliferation of neutrophils
of the chemotherapeutic regimen, a combi­ (granulocytes). As patients may have pro­
nation of antiemetics is utilized to prevent longed periods of myelosuppression after
and treat CINV. At a minimum, patients chemotherapy, G‐CSF is recommended for
receiving low emetogenic chemotherapy adult patients with: (i) the expectation for
should receive ondansetron. The addition of prolonged myelosuppression to prevent epi­
a NK1 receptor antagonist, such as aprepi­ sodes of febrile neutropenia and infection,
tant, is an excellent second‐line agent but (ii) a history of febrile neutropenia, (iii) the
should be used with caution with concomi­ history of a delay between chemotherapy
tant vincristine or ifosfamide due to syner­ cycles, and (iv) a diagnosis of febrile neutro­
gistic risk of neurotoxicity. Dexamethasone penia. Pediatric evidence is minimal com­
is an effective antiemetic if started prior to pared to adult studies but supports the use
chemotherapy onset, although should be of G‐CSF for patients with two of these four
avoided in patients with brain tumors due to scenarios: (i) prevention of febrile neutrope­
theoretical risk of causing decreased influx nia and infection (primary prophylaxis),
of systemic chemotherapy beyond the and (ii) treatment of febrile neutropenia.
blood–brain barrier. Dexamethasone should Although infection‐related morbidity has
also be avoided in patients receiving steroids not been shown to decrease with G‐CSF
as part of their chemotherapeutic regimen usage in these scenarios, risk of infection
(i.e., acute lymphoblastic leukemia and lym­ and length of hospitalization have been
phoma). The dose must also be halved if shown to be significantly reduced in meta‐
used concomitantly with a NK1 receptor analyses, with potential benefits for quality
antagonist. of life and decreased cost from shorter hos­
pitalization. GM‐CSF has shown similar
results to G‐CSF but has not been shown
Hematopoietic growth factors to be superior in clinical studies. In general,
in children with cancer G‐CSF is the colony‐stimulating factor of
choice in pediatric patients. Clinical studies
Several hematopoietic growth factors have have also established that the appropriate
been approved for clinical use in children doses of G‐CSF and GM‐CSF are 5 mcg/kg/
including granulocyte colony‐stimulating day and 250 mcg/m2/day, respectively, given
factor (G‐CSF, filgrastim or pegylated G‐CSF, SC or IV. It should be noted that the IV dose
pegfilgrastim) granulocyte–macrophage col­ may be less effective than the SC dose. A
ony‐stimulating factor (GM‐CSF, sargra­ longer lasting pegylated form of G‐CSF
mostim), and erythropoietin (EPO, epoetin (pegfilgrastim) has been shown to be equally
alfa). Thrombopoietin (TPO) receptor ago­ efficacious in adult studies with less fre­
nists (i.e., rivaroxaban) are discussed in quent dosing; pediatric randomized con­
Chapter 12 and have not shown benefit to trolled studies are lacking but pegfilgrastim
date in chemotherapy‐induced thrombocyto­ can be prescribed in children at a dose of
penia. The indications for each of these 0.1 mg/kg.
growth factors are based on limited evidence
in pediatric patients and are generally derived Primary prophylaxis
from adult data. Here we summarize general G‐CSF should be given to patients receiving
recommendations for the use of these agents multiagent chemotherapy who are expected
in pediatric oncology. to experience a high incidence of febrile
Table 25.2 Common pediatric antiemetic agents.

Agent Mechanism of action Dose Comments

Ondansetron 5‐HT3 receptor 0.15 mg/kg Well‐tolerated; common side


(also granisetron, antagonist q8h IV/PO effects include headache,
palonosetron) (max 32 mg/day) fatigue, constipation,
diarrhea. Can cause QTc
prolongation. Also available
as orally disintegrating tablet
Lorazepam Interaction with 0.25–0.5 mg Used as adjunctive; can be
GABA (gamma q4–6 h IV/ PO, utilized for anticipatory
aminobutyric acid) max dose 2 mg nausea. At higher doses has
receptor; poorly more sedation/anxiolytic
understood effect than antiemetic effect
antiemetic effects
Diphenhydramine H1 histamine 0.5–1 mg/kg Used as adjunctive; can be
receptor q6 h IV/PO, utilized for anticipatory
antagonist max dose 50 mg nausea. Higher dose used
for prevention of dystonic
reaction with
metoclopramide
Metoclopramide Dopamine 1 mg/kg q4–6 h IV/ Used as adjunctive; higher dose
antagonist PO, max dose required for antiemetic effect
50 mg; may also as compared to prokinetic.
use 0.075 mg/kg Must be given with
PO q6h after 1 mg/ diphenhydramine at higher
kg loading dose dose
Dexamethasone 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 Must be changed q72 h; patient
for adolescents/ must be advised to not touch
adults patch and then rub eyes, as
this will lead to mydriasis
Dronabinol Cannabinoid; 5 mg/m2 q2–4 h, No established pediatric
agonist max dose dosing. Teens and young
antiemetic effect 15 mg/m2 in adults should be advised to
adults not smoke cannabinoids
that can contain impurities
or increase the risk of fungal
infection
Aprepitant Neurokinin‐1 80–125 mg daily PO Must halve decadron dose if
receptor in adults; 3 mg/kg used concomitantly; use
antagonist on day 1, 2 mg/kg with caution with ifosfamide
on days 2 and 3 in
pediatric patients
Olanzapine Antipsychotic; 0.1 mg/kg PO Used as adjunctive for
5HT3‐receptor nightly, max dose anticipatory, delayed, or
antagonist 10 mg refractory nausea; can cause
sedation, weight gain, QTc
prolongation
320 Chapter 25

neutropenia or severe, prolonged neutrope­ etin alfa (rHuEPO) has been approved in
nia (i.e., ANC <0.5 × 109/l for 7 or more pediatric patients, although administration
days). G‐CSF should not be given on the of rHuEPO to oncology patients with
same days patients are given myelosuppres­ chemotherapy‐induced anemia remains
sive chemotherapy or radiation therapy. G‐ controversial. A second recombinant, dar­
CSF is generally not administered to patients bepoetin alfa, which has a twofold to three­
with myeloid leukemia due to the theoreti­ fold longer half‐life, has been approved in
cal risk of stimulating proliferation of the adult patients only.
leukemic clone. 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 thrombo­
ment should be considered in patients with embolism 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 addition,
venous thromboembolism has not been
Duration seen in pediatric patients. Concern remains
G‐CSF should be started between 1 and 5 that the decrease in adult survival rates with
days after the last dose of myelosuppressive rHuEPO may be secondary to the ubiqui­
chemotherapy or radiation. G‐CSF should tous expression of the EPO‐receptor on
be continued until the ANC is greater than tumor cells and therefore tumor upregula­
1.5 × 109/l for 1–2 days following the tion with rHuEPO usage. Conclusive in vivo
expected neutrophil nadir from chemother­ data are lacking.
apy. Specific protocols may call for different Pediatric guidelines for rHuEPO there­
ANC thresholds. fore include the following: (i) use of
rHuEPO is not recommended in pediatric
Monitoring oncology patients, and (ii) rHuEPO should
Once G‐CSF is initiated, a complete blood be considered on a case‐by‐case basis
count (CBC) and differential should be in special populations where blood
monitored at least weekly. pr­oduct usage is relatively contraindicated.
Specifically, Jehovah’s Witnesses forbid
Adverse Effects blood product transfusion and therefore
The most common side effects of G‐CSF are the potential risks and benefits of rHuEPO
bone pain and elevation of uric acid, LDH, should be discussed with the practicing
or alkaline phosphatase. Occasionally G‐ patient and their family. A candid discus­
CSF has been reported to cause fever, nausea sion should occur between the provider,
and vomiting, diarrhea, splenomegaly, and patient, family, and potentially patient
erythema at the injection site. advocate, to discuss this complex situation.
Although rHuEPO may ameliorate some
Recombinant human transfusion need, it may not be able to
erythropoietin (rHuEPO) eliminate this need completely. Many hos­
Erythropoietin induces proliferation and pitals have minimal blood use policies to
differentiation of red blood cell progeni­ assist in this situation. Additionally, the
tors. The recombinant product erythropoi­ patient and family should be made aware of
Supportive Care of the Child with Cancer 321

the theoretical risks regarding survival with first cycle of chemotherapy. We utilize loraz­
rHuEPO usage. epam, diphenhydramine, and scopolamine
as initial additional as needed medications.
The practitioner can also consider the use
Case studies for review of metoclopramide and dronabinol. For
patients with more refractory nausea,
1. You are following a patient with a new aprepitant and olanzapine can be of bene­
diagnosis of AML. The patient started stand­ fit. Generally, we utilize dexamethasone in
ard therapy with cytarabine, daunorubicin, non-central nervous system solid tumors.
and etoposide. The patient has been overall Dexamethasone should not be given with
well appearing to date without fever. brain tumors given the theoretical decre­
a. What are some initial considerations ment in influx of chemotherapy due to seal­
to prevent infection? ing of the blood-brain barrier and should be
Given the high risk of infection with treat­ avoided in AML secondary to the increased
ment of AML, patients should remain hos­ risk of infection.
pitalized until count recovery in a HEPA c. What are the considerations for utili­
filtered immunocompromised unit. Patients zation of GCSF?
(and caregivers) should be instructed on Although some AML regimens utilize GCSF,
appro­priate oral care as well as appropriate it is generally avoided in leukemia treatment
sterile technique regarding central venous given the theoretical (though generally dis­
catheter care to prevent introduction of proven) belief that GCSF can stimulate the
infection. Patients should be started on leukemic clone. GCSF is utilized regularly in
TMP/SMX on the weekends (i.e., 2–3 days a solid tumors with high rates of neutropenia.
week) to prevent PCP. For patients with Patients with solid tumors and a previous
severe risk of infection such as the patient history of febrile neutropenia or delayed
receiving treatment for AML, antifungal count recovery may also be prescribed GCSF
and antibacterial prophylaxis should be following chemotherapy. For the patient
instituted once the ANC <0.5 × 109/l. Either with AML and serious infection or symp­
an echinocandin or an extended spectrum toms of sepsis, GCSF should be utilized.
azole (i.e., posaconazole in the older patient)
can be utilized for fungal prophylaxis. 2. You are now seeing the above mentioned
Levofloxacin is generally utilized for anti­ patient with AML who has completed ther­
bacterial prophylaxis until the patient has a apy successfully. Treatment went relatively
fever requiring initiation of parenteral anti­ smoothly beyond need for transfusion (a
biotics (i.e., cefepime and vancomycin in total of 14 packed red blood cell [PRBC]
this case due to risk of Streptococci viridans transfusions) as well as two episodes of bac­
with cytarabine therapy). teremia. Total anthracycline (doxorubicin
b. What are some initial considerations equivalents) was 492 mg/m2.
to prevent CINV? a. What are your recommendations
Reviewing the emetogenic potential of the regarding infection prophylaxis?
chemotherapeutic agents, there is a moder­ With removal of the central venous catheter,
ate risk of CINV. Patients should be on risk of infection decreases dramatically. With
scheduled ondansetron at the minimum for ANC recovery after the last cycle of therapy,
prevention of CINV. Given the interpatient the patient can slowly resume normal activ­
variability, it is difficult to fully determine ity, though should be advised that normal
risk of CINV prior to the initiation of the immune function will take months to
322 Chapter 25

recover. TMP/SMX should be continued for device (SQUID). Patients with confirmation
3 months after the completion of therapy. of iron overload with increased liver stores
b. What are your recommendations should receive regular phlebotomy until
regarding immunization? normalization of the ferritin. Young children
Patients after AML therapy will be immune and menstruating females often can normal­
suppressed for some period of time and also ize their ferritin and iron overload with time,
are highly likely to have lost immunity to especially if the ferritin level and imaging
some prior vaccines. Patients can receive studies are borderline.
booster vaccines (or restart the primary series
if not previously completed) or can have vac­
cine titers checked to determine which anti­ Suggested reading
bodies are lacking prior to immunization. For
the patient with AML, we recommend wait­ Feusner, J. (2009). Guidelines for Epo use in children
ing 6 months after completion for therapy with cancer. Pediatr. Blood Cancer 53: 7–12.
for inactivated or dead immunizations and Feusner, J.H., Hastings, C.A., and Agrawal, A.K.
(eds.) (2015). Supportive Care of the Child with
12 months for live viral vaccinations.
Cancer: A Practical Evidence‐Based Approach.
c. What are your recommendations Springer. New York.
regarding cardiac screening? Freifeld, A.G., Bow, E.J., Sepkowitz, K.A. et al.
Patients that have received this total (2011). Clinical practice guideline for the use
anthracycline dose should have biannual of antimicrobial agents in neutropenic patients
echocardiogram per the current Children's with cancer: 2010 update by the Infectious
Oncology Group guidelines for monitoring Diseases Society of America. Clin. Infect. Dis.
of late effects. Risk of long-term left ventric­ 32: e56–e93.
ular dysfunction is more likely with time, Hesketh, P.J., Bohlke, K., Lyman, G.H. et al.
with increasing risk over 10–20 years and in (2016). Antiemetics: American Society of
Clinical Oncology focused guideline update.
situations such as pregnancy. Age at time of
J. Clin. Oncol. 34: 381–386.
treatment also affects development of late
Seelisch, J., Sung, L., Kelly, M.J. et al. (2018).
cardiotoxicity. Dexrazoxane prophylaxis can Identifying clinical practice guidelines for the
be considered in young children at the ini­ supportive care of children with cancer: a
tiation of anthracyclines if the total planned report from the Children’s oncology group.
anthracycline dose equivalent is 300mg/m2, Pediatr. Blood Cancer 66: e27471.
and for older patients once the total dose Sung, L. (2015). Priorities for quality care in pedi­
equivalent has reached 300mg/m2. atric oncology supportive care. J. Oncol. Pract.
d. Is there a risk of iron overload sec­ 11: 187–189.
ondary to PRBC transfusion? What are Sung, L., Aplenc, R., Alonzo, T.A. et al. (2013).
the next steps? Effectiveness of supportive care measures to
reduce infections in pediatric AML: a report
Patients that have received 4 or more PRBC
from the Children’s Oncology Group. Blood
transfusions are at risk for iron overload. 121: 3573–3577.
Once the peripheral blood counts have recov­ Sung, L., Zaoutis, T., Ullrich, N.J. et al. (2013).
ered, patients should have a screening ferritin Children’s oncology group cancer control
level. Patients with a ferritin >1000ng/ml are and supportive care committee. Children’s
at risk for iron overload and should have Oncology Group’s 2013 blueprint for research:
additional screening with a hepatic R2 MRI cancer control and supportive care. Pediatr.
or superconducting quantum interference Blood Cancer 60: 1027–1030.
26 Central Venous
Catheters
Indwelling central venous catheters (CVCs) placed is based on multiple factors including
have revolutionized the care of children length and intensity of therapy; frequency of
with cancer by simplifying the administra- lab draws; the need for multiple lumens to
tion of therapy, increasing safety, decreasing support simultaneous administration of
pain, and improving quality of life by chemotherapy, hydration, pain medication,
­reducing physical and psychological stress. parenteral nutrition, antibiotics, and other
Peripheral venous access can become agents; age; and lifestyle. In general, external
increasingly difficult in young children. and multilumen devices are associated with
Additionally, peripheral administration of greater risk, primarily due to an increased
certain antineoplastic drugs may cause incidence of infection and thrombosis. Ports
injury due to extravasation. CVCs provide a are generally utilized with lower‐intensity
safe, accessible route for infusion of chemo- therapies and in older children and adoles-
therapy, total parenteral nutrition, blood cents. Ports are cosmetically more satisfac-
products, antibiotics, pain medications, tory, do not require routine care, cannot be
hematopoietic stem cells, and other medica- accidentally self‐removed, have less impact
tions and infusions. External devices also on body image, and allow for easy bathing
allow for frequent and convenient blood and swimming. However, ports may be dif-
sampling, which can be taught to home care ficult to place and access in obese or very
providers. CVCs are now an integral aspect thin or small patients. Ports require special
of management of cancer or chronic illness needle (i.e., Huber) access through the skin,
in children. The optimal type and timing of and topical anesthetics are used routinely to
placement of a CVC are not standardized. decrease or eliminate pain with access.
Many types of CVCs are available and are When left accessed for prolonged periods,
divided between temporary (peripherally the Huber needle needs to be replaced every
inserted central catheters) and more perma- seven days. Subcutaneous ports also require
nent, tunneled catheters. This chapter monthly access with heparinization. The use
focuses on the permanent catheters, placed of ports for routine blood draws is limited
to provide access for months to years. due to their need to be accessed prior to use,
Tunneled catheters can either be entirely therefore an external CVC should be used in
under the skin (i.e., Mediport or Portacath) patients requiring frequent laboratory mon-
or have an external portion that allows itoring. External CVCs and ports may be
access (i.e., Hickman, Broviac, Powerline). used immediately after placement. In the
The decision about the type of CVC to be case of a port, the surgeon should be asked

Handbook of Pediatric Hematology and Oncology – Children’s Hospital & Research Center Oakland,
Third Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
© 2021 John Wiley & Sons Ltd. Published 2021 by John Wiley & Sons Ltd.
324 Chapter 26

Table 26.1 Type of central venous catheter to be placed by diagnosis.

CVC type Number of lumens Diagnosis

External Double AML


Neuroblastoma, advanced‐stage
Brain tumors on Head Start therapy
Solid tumors, advanced‐stage (potentially)
Patients requiring HSCT
Internal Single ALL, standard and high‐risk
Hodgkin and non‐Hodgkin lymphoma
Solid tumors except as mentioned above

Abbreviations: ALL, acute lymphoblastic leukemia; AML, acute myelogenous leukemia;


CVC, central venous catheter; HSCT, hematopoietic stem cell transplantation.

to access the device when it is placed, prior or lateral chest. Placement in other loca-
to development of postoperative edema. tions may be necessary for patients with a
This allows immediate use, decreasing history of thrombosis, multiple previous
patient discomfort and reducing the poten- CVCs, or in the patient with compression
tial for introduction of infection. Ultimately, of the upper venous system due to tumor.
in cases where an external catheter is not The location of the catheter tip should be
essential, the decision on the type of cathe- confirmed by fluoroscopy or chest radiog-
ter to utilize must include a discussion with raphy to be at the junction of the superior
the patient and family reviewing the pros or inferior vena cava and the right atrium
and cons of each type of device. Table 26.1 prior to the catheter being used.
summarizes the type of CVC to consider Ultrasonography or angiography may assist
based on diagnosis and subsequent therapy the surgeon in determining the most acces-
intensity. sible vein for CVC placement. External
CVCs have a Dacron cuff attached to the
catheter wall that is positioned 1–2 cm
Insertion of catheters from the skin exit site. This allows for the
growth of fibrous tissue (scar) into the cuff,
Central venous catheters are typically made preventing migration and loss of the line.
of plastics with flexible silicone rubber Surgeons will typically place sutures exter-
tubing. They are surgically placed (and
­ nally as well that may be removed after
removed) by experienced practitioners via healing. Ports are secured to the deep fascia
a percutaneous or cutdown technique. The on the anterior chest wall with sutures.
patient should be assessed for bleeding Ports are made of nonimmunogenic mate-
risk by having a platelet count and coagula- rials and some now allow for high‐pressure
tion studies checked preoperatively. CVCs infusion of contrast for radiographic imag-
are typically inserted into the subclavian, ing (i.e., PowerPort). The silicone membrane
­internal jugular, or external jugular vein of the reservoir is accessed by a special
and tunneled to an exit site on the anterior non‐coring needle (Huber) and is self‐sealing
Central Venous Catheters 325

Table 26.2 Common tunneled central venous catheters and related care.

Catheter type Saline flush Heparin Site care

Hickman/Broviac ≤10 kg: 5 ml/lumen; 10 units/ml Dressing change weekly


>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 catheter 10 ml each lumen 100 units/ml Dressing change weekly
q24 hours >30 kg: 2 ml/lumen or per institution policy
15–30 kg: 1.5 ml/ (or when wet, dirty)
lumen Do not use iodine or
<15 kg: 1 ml/lumen iodine‐based disinfectants
q24 hours and after
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; PRN, pro re nata, as necessary.

with needle removal. Topical anesthesia is conventional double lumen catheter (see
typically applied over the reservoir prior to Table 26.2).
access. Risks of CVC insertion include
pneumothorax, hemothorax, chylothorax,
malpositioning, and arterial puncture. As Maintenance
with all surgical procedures, risk and ben-
efits need to be assessed, discussed, and External catheters require daily heparin
consent obtained. flushes and periodic dressing changes (see
Single lumen ports and double lumen Table 26.2). Families require detailed educa-
external catheters are used most commonly tion on the care and potential complications
in children. Patients that require pheresis for of CVCs. They should be taught sterile
stem cell collection early in their course of technique to access the CVC for blood
therapy benefit from placement of a pheresi- draws or flushes and to provide external
ble catheter, such as a Medcomp. These lines care. Frequency of dressing changes may be
require meticulous maintenance and are not dependent on institution policy, type of
repairable, unlike the standard external tun- CVC, and local factors such as a wet, dirty,
neled catheters. The Bard PowerLine is a or loose dressing. At all times, the CVC exit
popular double lumen pheresible catheter site should be kept dry and clean. Patients
due to the higher flow rates that are possible are instructed to protect it during bathing
through it, and it can frequently be placed in and not immerse the dressing or exit site in
children who are too small to safely receive a a bath, hot tub, or pool. Should this occur,
326 Chapter 26

the dressing should be changed immediately the pathogen along the external catheter
with careful cleansing of the exit site. Ports surface, and (ii) contamination of the hub,
require less care, and heparin flushes, which leading to intraluminal colonization and
are required only monthly, are typically per- consequent seeding of the pathogen into
formed by the health care team. No dressing the circulation (see Chapter 27 for com-
is needed for implantable devices when mon pathogens and management of fever
they are not accessed. Each institution has a in patients with CVCs). Patients with CVCs
standardized approach to the care and require constant attention for prevention
maintenance of CVCs that is followed by and early recognition of infection. Despite
staff, family members, and home health care the meticulous techniques used by health
agencies (CVC bundle) to help minimize care providers and caregivers to maintain
the risk of central line‐associated blood sterility, many children with CVCs will
stream infection (CLABSI). develop a line infection or bacteremia.
Infection is often related to organisms that
are ubiquitous but can lead to true infec-
Complications: mechanical tion in the immunosuppressed patient
(such as gut and skin flora), or be due to
The use of CVCs can result in a number organisms introduced with venous access
of short‐ and long‐term complications. or with blood product or medication/fluid
Immediate complications related to CVC administration. Coagulase‐negative staph-
placement include malposition, hemor- ylococci, Staphylococcus aureus, aerobic
rhage, pneumothorax, chylothorax, arterial Gram‐negative bacilli, and Candida albi-
cannulation, cardiac dysrhythmia, and fail- cans most commonly cause catheter‐related
ure to place the line. Rarely, complications bloodstream infection. Additionally, devel-
may occur due to the anesthetic required for opment of a thrombus in the catheter may
surgical placement. Long‐term complica- serve as a nidus for infection. Occasionally,
tions include mechanical failure, catheter despite best efforts and appropriate antibi-
breakage, leakage, and port extrusion. otic therapy, the line may be colonized and
Mechanical failures have been reported in need to be removed. Recent techniques for
up to 10% of patients and may result in catheter sterilization include antibiotic and
removal of the CVC. Some types of external ethanol locks. Additionally, many manu-
CVCs may be repaired if the break is distal facturers have created antibacterial sub-
to the catheter exit site. Children with stances bonded to the catheters.
implanted ports are at risk for the Huber Risk factors for CVC infection include
needle becoming dislodged during infusion, the placement of an external catheter, young
leading to extravasation with resultant skin patient age, multilumen catheter, early
irritation or breakdown. These require care- placement (i.e., within 2 weeks of diagnosis
ful periodic assessment. of acute lymphoblastic leukemia [ALL]),
stem cell transplant, solid tumor, lack
of perioperative prophylactic antibiotics,
Complications: infectious high‐intensity protocol, and placement dur-
ing periods of neutropenia. These factors
Infection is the most common complica- must be weighed against the benefits of
tion of CVCs. Most catheter‐related infec- early line placement, type of line placed, and
tions arise by one of two mechanisms: (i) number of catheter lumens. The ­relatively
infection at the exit site with migration of higher infection rate seen with external
Central Venous Catheters 327

catheters favors the use of implanted cathe- ­culture results, but if the patient is neutro-
ters when possible. penic it should include both broad‐spec-
Approximately 50% of catheter‐related trum and directed antibiotic therapy. Site
infections occur locally at the exit site, along infections provide an opportunity to review
the tunneled catheter or as a “pocket” infec- meticulous local CVC management with the
tion around the implanted reservoir. Skin 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 irri- the patient will have repeatedly positive cul-
tation, skin breakdown, infection, and pain. tures with the same organism, often without
Patients with local catheter infections typi- systemic symptoms of infection. In cases of
cally have local signs and symptoms with colonization, cultures may become quickly
tenderness on palpation, erythema, puru- positive and may persist through appropri-
lent drainage, lack of healing, swelling, and ate antibiotic therapy. At times, catheters
pain. Severely neutropenic patients may may be reseeded from distant sites of occult
only have pain and tenderness as these infection (e.g., cardiac vegetations, osteo-
symptoms do not require the presence of myelitis, deep abscesses). Colonized cathe-
neutrophils. If drainage is present, the site ters must be removed.
should be cultured. Tunnel infections are The CVC should always be considered
characterized by a spreading cellulitis along the source of infection in a patient who pre-
the subcutaneous tract of long‐term cathe- sents with fever until proven otherwise.
ters. The catheter should be removed if the Fever, chills, or hypotension temporally
administration of appropriate parenteral related to line flushing increase the likeli-
antibiotics does not result in resolution of hood of a line infection. Broad‐spectrum
the signs and symptoms within 24–48 hours. empiric antibiotics should be administered
When evaluating a child with fever and a after drawing cultures from each lumen of
CVC, palpate along the tunnel to assess for the CVC. Persistent bacteremia despite
tenderness or to express drainage. Pocket appropriate antibiotic coverage (>72 hours),
abscesses may need to be surgically drained signs of sepsis (hypotension, chills, persis-
and packed and almost always will require tent fever, cool extremities, delayed capillary
removal of the reservoir and catheter. Site refill), or infection with fungus, mycobacte-
infections with minor exit site erythema and rium, S. aureus, or water‐borne bacteria
tenderness are treated with topical antibiot- (e.g., Pseudomonas aeruginosa) warrant
ics and monitored with careful, daily assess- CVC removal. If placement of a new CVC is
ment; systemic antibiotics should be added deemed necessary, the patient should first
for clinical worsening, lack of improvement, complete an appropriate course of antibiotic
or a positive blood culture from the line. therapy allowing for sufficient healing time
Tunnel and pocket infections should be and decreasing the risk of subsequent rein-
treated with both topical and systemic anti- fection of the new CVC. Patients who expe-
biotics, as well as a daily sterile dressing rience bacterial sepsis with hemodynamic
change. The patient should be treated sys- instability, septic thrombosis, or endocardi-
temically with broad‐spectrum antibiotics tis should have their CVCs removed.
providing good Gram‐positive coverage and CVC infection may be over‐diagnosed,
be monitored for the development of an resulting in either prolonged antibiotic
abscess requiring surgical drainage. The administration or unnecessary removal of
antibiotic regimen may be altered with the catheter. Differential time to positivity
328 Chapter 26

can be a reliable diagnostic technique to eter kinking or compression by a mass or


differentiate a true line infection from bac- other structure, ratio of the catheter size to
teremia. Paired blood samples (aerobic and the intraluminal vessel diameter, blood
anaerobic) from a peripheral vein and the flow rheology, genetic predisposition, and
central catheter are obtained and com- administration of prothrombotic medica-
pared with respect to time to positivity. If tions or infusions (e.g., asparaginase, ster-
the culture from the catheter turns positive 2 oids, total parenteral nutrition). Primary
or more hours before the peripheral cul- thromboprophylaxis is not currently recom-
ture, this is diagnostic of a catheter‐related mended as standard practice; however, it
infection. A positive differential time to may be warranted in certain situations with
positivity may not change management, documented increased risk.
although consideration should be given to Thrombosis should be suspected in
the type and duration of systemic antibi- patients whose catheter does not flush or
otic therapy, utility of an antimicrobial or draw easily or in the event of suggestive
ethanol lock, and the need for catheter clinical findings (pain or swelling in
removal. Addi­ tional factors include the extremity, prominent vasculature or swell-
type of catheter, prior history of infection, ing on side of neck/chest with CVC, inabil-
current neutrophil count, likelihood of ity to successfully access an implanted
short‐term recovery, and current chemo- port). It should be noted that a line that
therapeutic regimen. When a specific does not draw or flush might be occluded
pathogen has been identified, antibiotic by the vessel wall or a valve. Patients sus-
therapy should be targeted, with additional pected of having a thrombosis should
broad empiric coverage in the neutropenic undergo imaging to confirm this and deter-
patient (see Chapter 27). The possibility of mine the extent (see Figure 26.1). Based on
an infected catheter‐related thrombus the timing of line dysfunction or symptom
should be considered, and treatment may development, the age of the clot can be
also involve thrombolytic agents or approximated. Additionally, pertinent fam-
anticoagulation. ily history of thrombosis should be sought
to help guide the necessity of genetic evalu-
ation for thrombophilia (see Chapter 10).
Assessment and management Finally, environmental and medication pro-
of catheter‐related thrombosis thrombotic factors should be considered
(e.g., immobilization, steroids, and other
Venous thromboembolism is another com- medications).
mon complication of long‐term CVCs. The Treatment of thrombosis may require
extent of thrombosis may include the cath- thrombolytic therapy, anticoagulation, or
eter tip (ball‐valve clot), the length of the catheter removal. Guidelines are provided
catheter (fibrin sheath), or the catheterized here for the assessment and treatment of a
vessel (e.g., the upper limb with or without suspected CVC‐associated thrombus:
the central vasculature of the neck or medi- ●● Examine the catheter for any kinks in the

astinum). Catheter‐related venous throm- tubing.


boembolism can result in significant ●● Reposition the patient; flush the CVC

morbidity, and subclinical thrombosis may with normal saline (if possible) and again
occur in up to 50% of patients with CVCs. attempt to withdraw blood. Have the patient
The risk of thrombosis is influenced by hold their hands above their head, turn,
catheter location, insertion technique, cath- cough, or hold their breath.
Central Venous Catheters 329

Initial evaluation
Physical examination, including assessment of malfunctioning CVC
History (prothrombotic medications, family history)
Laboratory assessment (fibrinogen, PT, PTT, CBC)

Diagnostic imaging for confirmation of TE:


Ultrasound with Doppler and/or
CT with venography and/or
Catheter dye study

Is CVC essential for care?

no
yes

Thrombophilia risk assessment Occlusive: initiate thrombolysis,


(see Chapter 10) then anticoagulation
Nonocclusive: anticoagulation

Occlusive: initiate thrombolysis up to 48–72 hours;


anticoagulation
Nonocclusive: consider thrombolysis up to 48–72 hours;
anticoagulation

Functional CVC?

no
yes

Successful lysis Residual thrombus


of TE Continue therapeutic
Remove CVC
anticoagulation
Continue therapeutic anticoagulation
Consider CVC removal
if still with residual clot until clot
dissolution
Continue prophylactic anticoagulation
Therapeutic anticoagulation for 3 months for 6 weeks to 3 months after clot
followed by prophylaxis until CVC resolution
removed

Figure 26.1 Evaluation of suspected thromboembolism in patients with tunneled central venous cath-
eters. Abbreviations: CVC, central venous catheter; PT, prothrombin time; PTT, partial thromboplastin
time; CBC, complete blood count; TE, thromboembolism; CT, computed tomography.

●● Attempt a tissue plasminogen activator ⚪⚪ After the first dwell, attach an empty 3 ml

(TPA) dwell if the catheter can be flushed: syringe and attempt to withdraw blood. If
⚪⚪ Infants <3 months receive 0.25 mg successful, obtain blood work and flush line
(0.5 ml) per lumen; other children <10 kg per protocol. If unsuccessful, instill a second
receive 0.5 mg (1 ml) per lumen, while TPA dwell for an additional 30 minutes.
those 10–30 kg receive 1 mg and >30 kg ●● If the TPA dwell is unsuccessful, further

receive 2 mg. Dwell should be adminis- workup is required to determine the extent
tered for 30 minutes. of thrombus.
330 Chapter 26

⚪⚪ Obtain a chest radiograph to confirm line is removed. For the patient with a stable
appropriate line position. clot and a functional CVC, therapeutic anti-
⚪⚪ Obtain further imaging studies such as coagulation with LMWH should be contin-
a dye study of the line, ultrasound with ued until approximately 3 months after clot
Doppler flow, or CT with venography to dissolution and then switched to prophylac-
evaluate for thrombosis location and tic anticoagulation until line removal (see
extent. Assess for sleeve thrombus, Chapter 10 and Formulary for dosing guide-
mechanical failure of the CVC, or migra- lines). Additionally, if the patient is deter-
tion of the catheter outside the vessel. mined to have a genetic predisposition to
⚪⚪ If a thrombus is identified and the time thrombosis based on family history or a per-
from symptom onset is <14 days, throm- sonal history of previous thromboembolism,
bolytic therapy with low‐dose systemic a thrombophilia trait workup should be sent
TPA should be attempted if the catheter (see Chapter 10 for details). If the patient has
can be flushed. multitrait thrombophilia and therefore a
⚪⚪ Infants <3 months receive 0.06 mg/kg/h higher risk of rethrombosis, every effort
for 6–24 hours; older children receive should be made to remove the CVC once the
0.03 mg/kg/h. If no clinical improvement in risk of having an indwelling line outweighs
24 hours, double the dose to 0.06–0.12 mg/ the benefit (i.e., maintenance therapy in the
kg/h (max dose 2 mg/h). ALL patient).
⚪⚪ Systemic TPA can be given for up to If the patient on prophylactic anticoagu-
96 hours. lation develops rethrombosis, management
⚪⚪ Monitor the fibrinogen level and main- should be the same as with the initial clot
tain above 100 mg/dl with cryoprecipitate. with systemic low‐dose TPA followed by
●● Monitor the patient for signs of sepsis, as therapeutic anticoagulation with LMWH
bacteria can be released into the blood- until clot dissolution. If the patient is on
stream with dissolution of the thrombus. therapeutic anticoagulation with clot exten-
●● If the catheter becomes functional after sion, the CVC will need to be removed. In
administration of systemic TPA, obtain a cases where the clot cannot be dissolved or
follow‐up radiographic study to assess for stabilized and the line needs to be removed,
clot resolution. 3–5 days of anticoagulation is recommended
●● Remove the catheter if the thrombus for clot stabilization prior to catheter
­cannot be cleared after 24–48 hours. removal. Special circumstances will warrant
disruption in anticoagulation, such as lum-
If the thrombus dissolves, or is stabilized bar punctures to administer intrathecal
with normal CVC function, determination chemotherapy or other surgical procedures.
should be made as to the current necessity of In general, LMWH should be held for
the line and risk for recurrent thrombosis. If 24 hours in advance of these procedures and
the CVC can be safely removed, some practi- resumed 12–24 hours after procedure com-
tioners recommend short‐term prophylactic pletion, depending on the nature of the pro-
anticoagulation to mitigate the risk for cedure and risk of postoperative bleeding.
rethrombosis (usually 6 weeks to 3 months Additionally, LMWH should be held during
of low‐molecular‐weight heparin [LMWH]). periods of moderate thrombocytopenia (i.e.,
If the line needs to be maintained and the <50 × 109/l) to minimize additional risk of
clot has resolved, the patient should continue bleeding. Longer‐acting medications such as
on therapeutic LMWH for 3 months and fondaparinux should be held for a ­minimum
then switch to prophylactic LMWH until the of 48–72 hours prior to lumbar puncture. If
Central Venous Catheters 331

the CVC is removed and it is necessary to catheter‐related thrombosis in children:


­
place a new catheter, consideration should a retrospective analysis. Blood Coagul.
be given to anticoagulation prophylaxis to Fibrinolysis 27: 384–388.
prevent recurrence of thrombosis. Hord, J.D., Lawlor, J., Werner, E. et al. (2016).
Central line associated blood stream ­infections
in pediatric hematology/oncology patients
Suggested reading with different types of central lines. Pediatr.
Blood Cancer 63: 1603–1607.
Bundy, D.G., Gaur, A.H., Billet, A.L. et al. (2014). Mermel, L.A., Allon, M., Bouza, E. et al. (2009).
Preventing CLABSIs among pediatric hema- Clinical practice guidelines for the diagnosis
tology/oncology inpatients: national collabo- and management of catheter‐related infec-
rative results. Pediatrics 134: e1678–e1685. tion: 2009 update by the Infectious Diseases
Chen, K., Agarwal, A., Tassone, M.C. et al. Society of America. Clin. Infect. Dis. 49:
(2016). Risk factors for central venous 1–45.
27 Management of Fever
in the Child
with Cancer
Children with cancer are at an increased risk ●● Type of therapy with dose‐ or time‐inten-

for serious bacterial, viral, and fungal infec- sive therapies: administration of high‐dose
tions. Many factors contribute to this suscep- cytarabine, AML and ALL induction, and
tibility in immunocompromised children. HSCT conditioning regimens confer the
The two most important determinants of highest risk.
susceptibility to bacterial and fungal infec- ●● Nutritional status: malnutrition adversely

tion are the number of circulating neutro- affects immune function.


phils (absolute neutrophil count; ANC) and ●● Disruption of protective barriers such as

the duration of severe neutropenia. One of skin, mucocutaneous tissues, gastrointesti-


the most significant advances in decreasing nal tract, and exit sites of catheters.
mortality and improving survival for chil- ●● Defects in humoral immunity related to

dren and adolescents undergoing cancer therapy, impaired splenic function, or B‐cell
therapy is the recognition of fever as a medi- malignancies.
cal emergency and the prompt administra- ●● Defects in cellular immunity such as seen

tion of broad‐spectrum antibiotics. in patients with T‐cell malignancies or those


Risk factors for febrile neutropenia (FN) receiving steroids or radiation; persistence
include: of lymphopenia due to chemotherapy.
●● Underlying disease: leukemia, especially ●● Colonization from endogenous microflora.

acute myelogenous leukemia (AML) and ●● Presence of indwelling catheters or shunts

relapsed acute lymphoblastic leukemia (e.g., ventriculoperitoneal).


(ALL), advanced stage lymphoma, and
hematopoietic stem cell transplant (HSCT) The ANC is calculated by multiplying the
are at higher risk due to increased therapy percentage of neutrophils (segmented neu-
intensity. trophils + bands) by the total white blood
●● Remission status: patients not in remis- cell (WBC) count.
sion are at higher risk. Example: WBC = 1.0 × 109/l, segmented
●● Recent administration of chemotherapy neutrophils = 10%, bands = 10%.
anticipated to cause prolonged myelo- ANC = 20% × 1.0 × 109/l = 0.2 × 109/l (often
suppression. referred to as an ANC of 200).

Handbook of Pediatric Hematology and Oncology – Children’s Hospital & Research Center Oakland,
Third Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
© 2021 John Wiley & Sons Ltd. Published 2021 by John Wiley & Sons Ltd.
334 Chapter 27

Neutropenia: ANC < 1.5 × 109/l.


Fever and neutropenia
Moderate neutropenia: ANC < 1.0 × 109/l.
Severe neutropenia: ANC < 0.5 × 109/l.
FN is an emergent situation and the
Profound neutropenia: ANC < 0.1–0.2 × 109/l.
patient should be considered septic until
In general, patients with an ANC of proven otherwise. These children may
<0.5 × 109/l or those with dropping counts have fever as the first or only symptom of
after chemotherapy expected to be infection. It should be noted that neutro-
<0.5 × 109/l within 48 hours are considered penia will not prevent the patient from
severely neutropenic. The ANC at time of having a temperature spike with infection;
presentation with respect to the most recent one exception is the patient on steroids
chemotherapy gives important information. (such as in ALL induction and delayed
For instance, if a child is neutropenic with- intensification) who may not mount a
out recent myelosuppressive therapy, infec- fever with an underlying infection. A
tion (and less likely relapse of a hematologic careful clinical examination is critical, as
malignancy) should be considered as the the patient’s status may change quickly
most likely etiology due to marrow suppres- and dramatically. As neutropenic hosts
sion. In general, the expected nadir in ANC may have blunted inflammatory responses,
occurs 7–14 days from the start of myelo- even subtle signs and symptoms of infec-
suppressive chemotherapy. This timing tion should be considered significant.
should be considered when assessing the Neutropenic patients who develop clinical
patient and determining the management signs or symptoms suggestive of infection
plan. Practitioners may also utilize the abso- without fever should be managed in the
lute phagocyte count (APC), which is the same emergent manner as the neutropenic
summation of the ANC and absolute mono- patient presenting with fever.
cyte count, in determining the risk of bacte- Septic shock results from overwhelming
rial infection, as monocytes also possess infection with microorganisms in the
bacteria‐fighting ability. An increasing blood leading to central vascular dilation
monocyte count is often seen prior to and with resultant circulatory failure and inad-
often heralds neutrophil recovery. equate tissue perfusion. Symptoms of sep-
Fever is defined as a single temperature tic shock include hypotension, tachycardia,
from any source of 38.3 °C (101.0 °F) or tachypnea, clammy extremities (although
greater, two temperatures of 38.0–38.2 °C can be warm with initial sepsis), decreased
(100.4–100.9 °F) within a 24‐hour period, urine output, and deterioration of mental
or a temperature of 38.0–38.2 °C (100.4– status. Although septic shock can be seen
100.9 °F) persistently for 1 hour. Once a with any organism, the usual culprits with
fever is documented, it should be consid- acute and overwhelming sepsis are Gram‐
ered as real no matter what the underlying negative rods.
circumstances are, even if the patient The most common organisms causing
defervesces without any intervention. bacteremia and sepsis are:
Patients who develop a low‐grade fever ●● Gram‐positive: Staphylococci (coagu-
(38.0–38.2 °C) should be monitored with- lase‐negative, Staphylococcus epidermidis,
out antipyretics to determine if a fever Staphylococcus aureus including methicil-
spike will occur. Rectal temperatures lin‐resistant [MRSA]); Streptococci (α‐
should never be taken in potentially neu- hemolytic) including Streptococcus viridans
tropenic children. and Streptococcus mitis.
Management of Fever in the Child with Cancer 335

●● Gram‐negative: Enterobacteriaceae (Esche­ evidence of inflammation (faint ery-


richia coli, Enterobacter, Klebsiella, Serratia), thema, tenderness, or minimal discharge)
Pseudomonas aeruginosa, Stenotrophomonas may represent a source of infection, as
maltophilia, Acinetobacter sp. neutropenia diminishes classic inflam-
●● Anaerobic: Clostridium difficile, Bacteroides matory changes. Pain, however, is always
sp., Propionibacterium acnes. a concerning finding and should be con-
Other pathogens infecting cancer sidered a potential clue to site and source
patients include: of infection.
●● Fungi: Pneumocystis jiroveci, Candida sp., 2. Laboratory evaluation
Aspergillus sp., zygomycetes, cryptococci. ●● Complete blood count (CBC) with

●● Viruses: Herpes simplex virus (HSV), manual differential to determine ANC.


varicella zoster virus (VZV), cytomegalovi- ●● Blood cultures from each lumen of the

rus (CMV), Epstein–Barr virus, respiratory CVC; peripheral culture is necessary if the
syncytial virus, adenovirus, influenza, para‐ child does not have a central line or it is
influenza, human herpesvirus 6. unobtainable from the central line.
●● Other: Toxoplasma gondii, Strongyloides Simultaneous peripheral blood culture
stercoralis, cryptosporidium, Bacillus sp., and CVC blood culture can be considered
atypical mycobacterium. based on differential time to positivity
and may be helpful in differentiating
central line‐associated bloodstream
Initial evaluation of febrile infections (CLABSI) from the non‐
neutropenia CLABSI infection. It remains unclear of
the utility of this assessment in treat-
See Figure 27.1. ment‐related decision‐making in febrile
1. History and physical examination (PE) neutropenic patients (see Chapter 26).
●● Determination of vitals (temperature, Some practitioners do not routinely
blood pressure, heart rate, respiratory collect peripheral blood cultures due to
rate, oxygen saturation) and rapid assess- patient discomfort and possibility of
ment for shock. contamination. Do not flush the cathe-
●● The history should include a determi- ter after labs are obtained in the patient
nation of the presence of chills or rigor with a recent history of high fever, hypo-
following flushing of the central line, tension, chills, or rigors within 1 hour of
concerns for the site of the central venous flushing the catheter, suggestive of a line
catheter (CVC), recent chemotherapy or infection.
radiation therapy including dosage, ●● Serum chemistries to include electro-

medications (antibiotics, prophylaxis), lytes, liver, and renal function studies.


recent infectious exposures or travel, and ●● Clean catch urinalysis and urine cul-

prior history of infections including ture (no catheter sample for neutropenic
infected CVCs. patients) should be obtained, ideally
●● Meticulous physical examination with prior to the start of antibiotics, but
particular attention to sites of occult should not delay initiation of treatment.
infection such as the skin, exit sites of ●● Chest radiograph only if the patient

catheters (and the tunnel path of the has auscultatory signs or symptoms of
catheter), sites of any invasive procedure infection or respiratory compromise.
including bone marrow aspiration, oral The finding of a pulmonary infiltrate
cavity, and perianal areas. Even subtle should then prompt the practitioner to
336 Chapter 27

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

Initial Evaluation
History: Chills, rigors, fever within 1 hour of CVC flush
Recent immunosuppressive therapy
Symptoms of AGE, URI, pain
Physical examination: Vitals, CVC exit site(s), skin/mucosa, perineum
Laboratory assessment: CBC with differential
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, creatinine, 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, induction)
Diagnosis: ALL, NHL, solid tumors in AML, high risk ALL in consolidation or delayed intensification
remission High dose cytarabine
Neutropenia: Duration <7 days Prolonged neutropenia anticipated ≥7 days
Nontoxic appearance Profound neutropenia <0.1 x 10 9/L
No focal infection, mucositis, diarrhea Toxic appearance: Hypotensive, rigors, shock, tachypnea, hypoxia
Evidence of infection: Pneumonia, cellulitis, abdominal pain and
diarrhea, neurologic (mental status) changes
Known colonization with MRSA
Prior history of bacteremia/sepsis
Empiric antibiotic therapy; monotherapy Mucositis following chemotherapy
Consider outpatient management

Empiric therapy, broad spectrum, initiated promptly


Hospitalization
Treatment of co-morbidities as appropriate
Pain management as appropriate
Directed therapy as appropriate:
Vancomycin for recent high dose cytarabine, new fever in AML patient
Metronidazole 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.

consider further evaluation with chest causes with serologies and culture, if
computed tomography (CT) and possi- possible.
bly bronchoscopy to identify an organ- ●● Cultures and bacterial Gram stain or

ism (and sensitivity pattern). fungal stains from suspicious sites such
●● Patients with tenderness over the as the oropharynx, skin breakdown sites,
sinuses, perform diagnostic imaging to and catheter sites.
evaluate for sinusitis (sinus CT). ●● If diarrhea is present, send a stool sam-

●● Patients with symptoms of esophagitis ple for stool culture, rotavirus, and C. dif­
should be evaluated for viral or fungal ficile antigen.
Management of Fever in the Child with Cancer 337

●● In contrast to non‐oncology patients, for these organisms (including Pseudomonas)


lumbar puncture (LP) is not routinely should be initiated. Many antibiotic combi-
done as part of a serious bacterial infec- nations are effective, and the decision about
tion evaluation in oncology patients with the use of a specific agent or combination
FN. If LP appears clinically indicated, approach should take into consideration the
evaluation for increased intracranial pres- institution’s bacterial susceptibility patterns,
sure with appropriate imaging (i.e., head cost, toxicity, and local standards of care.
CT) should be considered and the pediat- Most institutions have adopted monother-
ric oncology attending notified prior to apy, as published meta‐analyses have not
the procedure. An adequate platelet count proven monotherapy to be noninferior to
and coagulation studies should be con- dual therapy. Common regimens include:
firmed in advance of the LP. 1. Fourth generation cephalosporin (anti‐
●● If another implanted device is pre- pseudomonal β‐lactam):
sent (e.g., ventriculoperitoneal shunt Cefepime 100 mg/kg/day intravenous
or Ommaya reservoir), do not attempt (IV) divided Q 8 hours:
to obtain a culture without speaking to 2. Carbapenems:
the appropriate attending physicians. Imipenem/Cilastatin 50 mg/kg/day (dosed
Intervention by a neurosurgeon may be on Imipenem component) IV divided Q
required. It is uncommon for these 6 hours.
devices to be the source of infection if 
Meropenem 60–120 mg/kg/day IV
it has been more than 2–4 weeks since divided Q 8 hours.
they were inserted. 3. Piperacillin/Tazobactam

(Zosyn) 300 mg/kg/day (dosed on
Piperacillin component) IV divided Q 4
Initial management hours.

Risk stratification models for pediatric Additional antibiotics used in specific cir-
oncology patients with FN have yet to be cumstances (see in the following text) are:
validated and widely accepted. Despite 1. Gram‐positive coverage:
­publication of numerous risk stratification Vancomycin 40–60 mg/kg/day IV
studies in FN, no single validated risk strati- divided Q 6 hours.
fication strategy has been adopted as stand- 2. Anaerobic coverage:
ard of care. It is generally accepted that  Metronidazole 30 mg/kg/day (loading
patients with high‐risk features (as deline- dose 15 mg/kg) IV divided Q 6 hours.
ated in the earlier text) should be hospital- Clindamycin 40 mg/kg/day IV divided Q
ized until count recovery, while certain 6–8 hours.
low‐risk groups may be able to be treated in
an outpatient setting after defervescence Gram‐positive organisms are frequent
(especially in the setting of a study). FN is an offenders in certain populations and
oncologic emergency and all measures empiric coverage is recommended in select
should be taken to ensure prompt evaluation situations. Patients with AML receiving
and treatment. Antibiotics should be chosen high‐dose cytarabine (Ara‐C) should have
based on microbial prevalence and antibiotic vancomycin as part of their empiric regimen
sensitivity patterns at each institution. In in combination with broad‐spectrum cover-
general, due to the more acute risk from age due to the high risk of S. viridans
Gram‐negative organisms, broad coverage ­infection (associated with septic shock and
338 Chapter 27

acute respiratory distress syndrome). f­ luids or medication administration initially


Cytarabine is known to alter mucosal integ- in this situation due to the risk of infusing a
rity and allow entry of this organism. Other large amount of bacteria into the blood-
indications for vancomycin in the empiric stream and worsening the signs of clinical
regimen include: sepsis. Patients should have central catheter
●● Presentation with hypotension or other cultures, placement of a peripheral IV,
evidence of shock. peripheral blood cultures obtained, and
●● Mucositis. receive broad‐spectrum antibiotics immedi-
●● Patients with prior history of α‐hemolytic ately in addition to fluid boluses (e.g., NS/
Streptococcus bacteremia. LR 20 ml/kg). The central catheter should
●● Catheter site infection or other skin not be flushed or utilized until the patient
breakdown. improves clinically with defervescence, res-
●● Patients colonized with resistant organ- olution of hypotension, and negative follow‐
isms treatable only with vancomycin. up blood cultures. If the line cannot be
●● Vegetations on echocardiogram. flushed, a tissue plasminogen activator
●● Severe pneumonia. (TPA) dwell can be attempted; if unsuccess-
ful, the line will need to be removed (see
Anaerobic therapy should be considered Chapter 26). It is advisable to give an IV
empirically in patients who have significant fluid bolus prior to reusing the catheter in
mucosal breakdown, perineal skin break- the event the patient again develops symp-
down, peritoneal signs, or in cases of neu- toms of bacteremia with hypotension fol-
tropenic colitis, or with presumed or proven lowing the flush. In the event the initial
C. difficile infection. blood culture from the catheter is positive,
Patients receiving vancomycin should the clinician will need to decide whether to
have trough levels monitored weekly (once remove the catheter or attempt to reuse it
therapeutic) due to the risks of nephrotox- depending on the patient’s clinical condi-
icity and ototoxicity, especially in those tion, ongoing need for central access, and
receiving other concurrent nephrotoxic or the organism’s identification and sensitivity
ototoxic drugs (e.g., furosemide). The goal pattern. Patients that have persistent fever
for a vancomycin trough level is 10–15 mcg/ and Gram‐negative organisms may do best
ml when treating a proven infection. Dose with removal of the line; patients that have
increases are often required in pediatric defervesced and are looking clinically well
patients to reach these levels (thus, gener- can generally have their line reused and ulti-
ally recommend starting at 60 mg/kg/day in mately salvaged.
the young patient). Monitor daily electro-
lytes, fluid balance, renal function studies,
and ensure adequate hydration. Patients Modification of initial
with renal dysfunction should receive renal antibiotic treatment
dosing with more frequent trough levels to
prevent toxicity. Subsequent modifications to the treatment
Any patient presenting with hypoten- plan are made on the basis of the clinical
sion, chills, rigors, or fever immediately fol- course, emergence of positive cultures, and
lowing, or within 1 hour, of CVC flushing estimated length and severity of neutrope-
should be evaluated emergently due to the nia. A careful daily clinical assessment
risk of bacteremia and septic shock. We elect includes a review of the vital signs, height
not to use the central venous line for IV and persistence of fever or change in the
Management of Fever in the Child with Cancer 339

fever curve, alterations in blood pressure, is a catheter site with accompanying signs or
and presence of any new symptoms or signs symptoms of inflammation (discharge, pal-
of infection on a meticulous physical exami- pable tenderness, local abscess), vancomy-
nation. The laboratory evaluation includes cin should be added for Gram‐positive
daily CBCs/ANC until count recovery, coverage. If the source is perineal, gingival,
review of prior cultures, and possible diag- or intra‐abdominal, anaerobic coverage
nostic imaging (see Figure 27.2). should be added to the regimen (metronida-
Modifications should be made to the ini- zole, clindamycin or meropenem).
tial broad‐spectrum antibiotic regimen if a Vancomycin is the most effective antibi-
source of infection is found that explains the otic to treat skin flora such as coagulase‐
initial fever, or the fever persists for more negative staphylococci (i.e., S. epidermidis) as
than 3–7 days. If the source of the infection well as mouth flora, most commonly

Empiric therapy

Negative cultures 48–72 hours, unknown


site of infection

Blood or site
cultures positive or site
organism suspected Persistent Afebrile ≥24 hours
fever/neutropenia

Evidence of neutrophil
Broaden coverage: Continue 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 consecutive days)
Mucositis/perineal breakdown: add metronidazole or 24 hours with PE Discontinue antibiotics
meropenem and cultures
Esophagitis: add acyclovir, consider antifungal
Pulmonary infiltrates: add triazole and/or
echinocandin after BAL/bronchoscopy
Catheter associated bacteremia >72 hours or Persistent neutropenia:
fungemia: remove CVC Continue antibiotics, until neutrophil
recovery*
If fever recurs, reassess with PE and
cultures, broaden antibiotics, and
consider antifungal therapy
*see text; consider outpatient management
for “low-risk” patients per institutional
guidelines
Persistent fever 3–5 days with neutropenia:
Imaging for infection if with expected
continuation of neutropenia: CT chest, other
areas based on symptoms
Urine cytospin for hyphae, fungal culture
Continue broad spectrum antibiotics,
broaden as indicated clinically
Initiate empiric antifungal 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.
340 Chapter 27

Streptococcal species (S. viridans, Pepto­ sensitivity patterns are reported. Once
streptococcus). Vancomycin is also effective microbial sensitivities are determined, fur-
against bacillus non‐anthracis species that ther narrowing of the antibiotic coverage
can be true pathogens in the immunocom- may be done. If daily cultures are persis-
promised patient. Finally, though rarely tently positive with the same organism for
seen, MRSA should be a consideration in 72 hours after initiation of appropriate anti-
the very sick patient. Vancomycin should be biotics, it will be necessary to remove the
used judiciously due to emerging resistance catheter. In addition, several organisms
patterns such as vancomycin‐resistant ente- including S. aureus, Pseudomonas, Bacillus
rococcus (VRE). Vancomycin should not be cereus, Corynebacterium, Mycobacterium
part of the empiric antibiotic regimen unless sp., Stenotrophomonas, and fungi usually
the institutional experience and susceptibil- require line removal. General care measures
ity patterns require it or there are special cir- to assess and manage a CVC with possible
cumstances warranting its use (as mentioned infection are as follows:
in the preceding text). ●● Hand washing with clean or sterile gloves

Protracted fever may signify the presence prior to manipulation of CVC.


of an additional or previously untreated ●● Monitor CVC site on a daily basis; change

infection. Patients should be reassessed daily CVC dressing weekly.


for new signs or symptoms of infection and ●● Assess site for skin infection (including

appropriate interventions and studies per- tunnel infection) with any fever, and daily.
formed. Antibiotic‐induced colitis may ●● For multilumen devices, the antibiotic

occur after any antibiotic. C. difficile over- infusion should be rotated among the
growth may be asymptomatic, lead to mild lumens, as infection may not be limited to
diarrhea, or may have moderate‐to‐severe one lumen. If one particular lumen has the
diarrhea and abdominal pain. The patient positive blood culture, it is reasonable to run
may also develop pseudomembranous colitis the majority of the antibiotic(s) through this
with peritoneal signs, mucosal erosions, and lumen. If more than one antibiotic is being
bloody diarrhea. Since C. difficile is a normal given, they should each be rotated to opti-
bowel inhabitant, the toxin must be docu- mize exposure of the lumens.
mented to diagnose this condition. If oral ●● Daily blood cultures should be obtained

therapy is possible, vancomycin (40 mg/kg (from each lumen of CVC) for the continu-
divided q8h) or metronidazole (30 mg/kg ally febrile patient or if growth is present,
divided q6h) may be given; in the patient until afebrile and negative cultures for 3 days.
unable to tolerate oral medications, IV met- ●● Assess cardiac valves for vegetations with

ronidazole (same as PO dose) should be an echocardiogram for repeatedly positive


given. Abdominal pain may also be due to cultures with Gram‐positive organisms.
infection with aerobic Gram‐negative bacte- ●● If bacteremia persists for more than 3 days

ria, enterococcus (which no cephalosporin with appropriate therapy, the catheter


covers in vivo), or anaerobes. should be removed.
If the patient is determined to have a
catheter‐associated bacteremia with a spe- For patients with a history of multiple posi-
cific organism, broad‐spectrum antibiotics tive cultures or a difficult‐to‐eradicate
should be continued (as long as the patient organism, consideration should be made for
remains neutropenic) with the addition of the use of ethanol or vancomycin lock ther-
antibiotics as needed to ensure adequate apy to possibly prevent the need for line
coverage of the identified organism until removal. Ethanol locks should not be used
Management of Fever in the Child with Cancer 341

with polyurethane catheters (PowerLine, Patients that defervesce but do not have
MedComp), as ethanol can damage the recovery of the ANC should continue on
catheter material. broad‐spectrum antibiotics to prevent
secondary infection. Some practitioners
may discontinue antibiotics after a period
Duration of therapy of time if the patient continues to be neutro-
penic but afebrile, although our general
Daily determination of the ANC is impor- practice is to continue until there is some
tant for patients who have defervesced, and evidence of count recovery.
have negative cultures, to determine the
appropriate length of therapy. When the
neutropenic patient becomes afebrile (no Empiric antifungal therapy
fever for 24 hours) and cultures are negative
(obtained daily while febrile), broad‐spec- When FN persists for 4–7 days, empiric
trum coverage should be continued until treatment should be broadened to include
there is evidence of adequate marrow recov- fungal prophylaxis. The risk of fungal infec-
ery and an appropriate period of therapy for tion is directly related to the duration and
any initially positive cultures has been pro- severity of neutropenia, which is secondary
vided (typically 48–72 hours). Resolution of to the intensity of chemotherapy or radia-
neutropenia is defined as an ANC of tion cytotoxicity. Patients with fever for 7 or
>0.5 × 109/l and evidence of bone marrow more days and associated profound neutro-
recovery is an ANC of >0.2 × 109/l and rising penia (ANC <0.1 × 109/l) are at the highest
on two consecutive days. Some practitioners risk for developing invasive fungal infec-
also use an APC of >0.5 × 109/l as sufficient tion. Thus, patients undergoing more inten-
evidence of recovery from neutropenia, dis- sive therapy, such as for AML, relapsed
continue antibiotics, and monitor in the hos- ALL, and hematopoietic stem cell trans-
pital or outpatient setting. Should the patient plantation, are particularly susceptible. The
have a localized infection, such as diarrhea major causative fungi are Aspergillus and
or skin breakdown, tailored therapy should Candida, with mortality as high as 30–60%
continue, but may be able to be done in the with documented invasive disease. Fungal
outpatient setting. The decision to discharge infections can be difficult to detect due to
a patient from the hospital without resolu- the frequent absence of localizing signs or
tion of neutropenia must include considera- symptoms and lack of means of detection
tion of risk factors as well as logistics of by culture. Prompt diagnostic assessment
outpatient management such as reliable and initiation of antifungal empiric therapy
transportation and compliance with care. are paramount for improving outcomes in
When a pathogen has been identified, its these patients.
antibiotic susceptibility pattern must be Invasive fungal disease is not uncommon
determined and appropriate antibiotic cov- in the patient undergoing highly immuno-
erage provided along with broad‐spectrum suppressive therapy. Invasive aspergillosis
coverage until the patient is afebrile and has (especially Aspergillus fumigatus) may be
evidence of bone marrow recovery. Once the associated with isolated pulmonary disease,
child is afebrile and the ANC recovers, anti- with characteristic findings on chest CT
biotics can be tailored to the specific organ- imaging of cavitating lesion or nodules.
ism for a 10–14‐day course, with day 1 being Invasive candidiasis is often suspected fol-
the day of the first negative culture. lowing positive blood or urine cultures or
342 Chapter 27

suspicious skin lesions, and pan-CT imaging have continued or new fevers with count
as well as ophthalmic evaluation should be recovery that should alert the clinician to
done to assess for possible disseminated dis- the need for repeat CT imaging.
ease. Patients with sinus signs and symptoms Risk factors and signs of development of
may have infection with Zygomycetes sp. (i.e., invasive fungal disease include:
Mucor, Rhizomucor, Rhizopus). ●● Prolonged use of corticosteroids.

Empiric antifungal therapy prevents ●● Persistence of fevers.

fungal overgrowth in patients with pro- ●● Prolonged fevers, over 4–7 days, in the

longed neutropenia. It also provides early face of severe or profound neutropenia.


treatment of clinically occult infection. ●● Recrudescence of fever after neutrophil

Clinical trials have found that antifungal recovery.


treatment of children with persistent or ●● Active graft‐versus‐host disease (GVHD).

recurring fever reduced morbidity and ●● Development of a new, focal nodular skin

mortality from invasive fungal disease. rash or eschar.


This is especially true in patients with pro- ●● Development of new respiratory findings

found neutropenia not receiving antifungal or periorbital swelling.


prophylaxis. The current recommendation ●● Tenderness over the sinuses and concom-

is that high‐risk groups (i.e., those expected itant CT findings such as bony erosion.
to have neutropenia >7 days) should receive ●● New findings on chest CT (nodules, infil-

antifungal prophylaxis during episodes of trates, cavitation, halo or crescent sign).


prolonged FN. ●● Shoulder pain.

A meticulous physical examination, ●● Positive galactomannan (serum, BAL),

laboratory assessments, and radiographic especially serial.


studies in search of deep‐seated infections ●● Positive fungal culture(s) (blood, urine).

are also warranted in FN patients with


prolonged unexplained fever (i.e., 4–7 Treatment of invasive fungal disease includes
days). Particular attention should be paid the following antifungal medications:
to the skin examination, as it can reveal ●● Invasive aspergillosis

fungal nodules. Fungal cultures should be ○○ Voriconazole IV

sent from the blood and urine, as well as ■■ 2 to <12 years: 7 mg/kg loading dose

from any suspicious skin site. A serum Q 12 hours × two doses followed by
galactomannan is done to screen for 7 mg/kg Q 12 hours.
Aspergillus infection (also can be done on ■■ ≥12 years: 6 mg/kg loading dose Q 12

urine of fluid from a bronchoalveolar lav- hours × two doses followed by 4 mg/kg
age, BAL). Serial repetition of this bio- Q 12 hours.
marker in conjunction with corroborative ○○ Voriconazole PO

clinical and radiographic findings ■■ 2 to <12 years: 8 mg/kg loading


increases its sensitivity as a useful diagnos- dose × two doses followed by 7 mg/kg BID.
tic tool. CT imaging of the sinuses, chest, ■■ ≥12 years:

as well as imaging of other symptomatic ●● <40kg: 100mg BID (max. dose 150mg).

areas (abdomen or pelvis) should be per- ●● ≥40kg: 200mg BID (max. dose 300mg).

formed. It should be noted that in the ○○ Posaconazole

severely neutropenic patient, areas of ■■ >13 years: 400 mg PO BID with meals.

infection may not be appreciated on imag- ○○ Micafungin 3–4 mg/kg IV Q 24 hours.

ing until neutrophil recovery has occurred. ○○ Caspofungin 70 mg/m IV loading dose
2

In these cases, the patient will generally followed by 50 mg/m IV Q 24 hours.


2
Management of Fever in the Child with Cancer 343

●● Mucormycosis with certain viruses, particularly the herpes


○○ Liposomal amphotericin B (Ambisome) family of viruses. Primary varicella infection
3 mg/kg IV Q 24 hours for empirical cov- is associated with high morbidity and
erage and 5 mg/kg IV Q 24 hours for mortality. In transplant patients, primary
­
­documented infection. infection or reactivation with CMV is an
●● Invasive candidiasis important cause of interstitial pneumoni-
○○ Fluconazole 12 mg/kg IV loading dose tis, marrow aplasia, and other infections.
followed by 6–12 mg/kg IV Q 24 hours. Antiviral drugs are available for a limited
number of infections including HSV, VZV,
The echinocandins (micafungin, caspo- CMV, and influenza and should be utilized
fungin, anidulafungin) have largely replaced in the treatment of immunocompromised
amphotericin B as the empiric antifungal patients. Other patients may also be candi-
agent of choice due to their once daily dos- dates for prophylaxis (i.e., solid organ trans-
ing, significantly decreased side effect pro- plant patients and patients with a prior
file, and broad coverage. Echinocandins are history of infection on therapy).
fungicidal to most Candida species and at Patients with documented HSV infec-
least fungistatic against Aspergillus species. tion should be treated with IV or oral
They also have limited activity against ­acyclovir (or valacyclovir) (30 mg/kg/day
Fusarium species and Zygomycetes. The tria- if <12 years; 15 mg/kg/day for ≥12 years
zoles are derivatives of fluconazole with divided Q 8 hours for 7–14 days). Dosing for
broad‐spectrum antifungal activity and lim- valacyclovir is 500 mg per os (PO) twice
ited side effects. Of note, the triazoles are daily (BID) for patients under 35 kg and
fungicidal against Aspergillus species. In gen- 500 mg three times daily (TID) for patients
eral, we utilize the echinocandins as first‐line weighing over 35 kg. Infection with VZV is
empiric therapy and consider adjustment in potentially life‐threatening and therefore
patients that are worsening clinically or have chemotherapy should be stopped while
signs consistent with Aspergillus infection. treating with IV acyclovir (30 mg/kg/day
Possible drug interactions with extended‐ divided Q 8 hours for 7–10 days or until all
spectrum azole agents should be considered. lesions are crusted and no new lesions have
Patients on either an echinocandin or tria- appeared for 24–48 hours). It is important to
zole should be monitored closely for hepato- monitor renal function and ensure adequate
toxicity; voriconazole may also cause visual fluid intake due to potential nephrotoxicity
disturbance that is more common in adult from acyclovir.
patients. Posaconazole may provide better Patients with symptomatic primary CMV
coverage against Aspergillus species but is infection or reactivation can be treated with
only available as an oral medication and clin- ganciclovir (10 mg/kg/day IV divided Q
ical trial data are limited in younger pediatric 12 hours for 14 days, then 5 mg/kg/day for
patients. maintenance [beginning after clearance of
CMV PCR], until resolution of viremia by
polymerase chain reaction). Patients with
Viral infection persistent infection may require longer ther-
apy or may have resistant disease and require
Immunosuppressed children are able to tol- alternate therapy with foscarnet or cidofovir.
erate many viral infections without difficulty. Of note, ganciclovir can be myelosuppres-
However, defects in cellular immunity pre- sive, whereas foscarnet and cidofovir are
dispose them to unusually severe infections quite nephrotoxic. Concomitant therapy
344 Chapter 27

with intravenous immune globulin (IVIG) have rales. Hypoxemia is evident by pulse
may be beneficial especially in the setting of oximetry and arterial blood gas. Chest radi-
CMV pneumonitis. Data regarding prophy- ography or CT shows an interstitial pattern.
lactic therapy in this setting is lacking, Without appropriate therapy, patients rap-
although once daily dosing (or with oral idly progress to respiratory failure.
valganciclovir) can be considered in Diagnosis is made by demonstrating the
patients that complete a course of antiviral organism by Gomori methenamine silver
therapy and remain neutropenic or severely stain on a sample obtained by induced
immunocompromised. sputum, BAL, percutaneous needle biopsy,
Patients who develop influenza while on or open lung biopsy. Patients who are at risk
therapy should be treated with neuramini- and present with the classic signs and
dase inhibitors (i.e., oral oseltamivir or symptoms should be treated while awaiting
inhaled zanamivir, dosed as below): definitive diagnosis, given the rapidity of
●● Oseltamivir clinical deterioration.
○○ 3–11 months of age: 3 mg/kg once daily. For documented or highly suspected
○○ 1–12 years: infection with P. jiroveci, trimethoprim/sul
■■ <15 kg: 30 mg once daily. famethoxazole (TMP/SMX) is the treat-
■■ >15 kg to <23 kg: 45 mg once daily. ment of choice, at a dose based on the
■■ >23 kg to <40 kg: 60 mg once daily. TMP component of 20 mg/kg/day IV
■■ >40 kg: 75 mg daily. divided Q 12 hours for 14–21 days. If the
○○ >12 years: 75 mg once daily. patient fails to respond to TMP/SMX
●● Zanamivir within 72 hours, or develops this infection
○○ ≥5 years: two inhalations (10 mg) once while on prior TMP/SMX prophylaxis
daily. with good compliance, initiate therapy
with pentamidine, 4 mg/kg IV daily. In
Patients are treated within 48 hours of patients with moderate or severe proven
exposure for 10 days to reduce symptom infection, concomitant administration of
duration and severity of disease. Oseltamivir corticosteroids may be beneficial with IV
may also be given as daily prophylaxis for methylprednisolone or prednisone given
5 days to patients that have been exposed to two to four times per day for 5–7 days, fol-
the virus. lowed by a taper over 1–2 weeks.

P. jiroveci pneumonia New sites of infection

P. jiroveci (formerly Pneumocystis carinii) is New or persistent fever and neutropenia


an atypical fungus and is a ubiquitous organ- may be associated with the development of
ism that may cause severe or fatal pneumo- 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 ●● Burning retrosternal pain may be esophagi-

with tachypnea and other symptoms of res- tis from Candida sp. or HSV. Cytotoxic treat-
piratory compromise such as nasal flaring ment may also cause severe pain due to
and intercostal retractions. Lung sounds mucosal erosions. Empiric micafungin or
may be clear and typically patients do not acyclovir, or both, may be needed. Esophagitis
Management of Fever in the Child with Cancer 345

may also be bacterial, especially as a result of


Fever in the non‐neutropenic
Gram‐positive aerobes.
oncology patient
●● Pulmonary infiltrates may be due to

resistant bacteria, Pneumocystis, fungi, or


The febrile, non‐neutropenic patient remains
viruses. Neutropenic patients may not
susceptible to infection secondary to
show evidence of pulmonary infiltrates
immune dysfunction related to underlying
due to lack of an appropriate local inflam-
malignancy and its treatment, as well as the
matory response. However, with the recov-
presence of an indwelling CVC. As with the
ery of the ANC, these sites of infection
neutropenic patient, these patients should
may become evident. If FN has been less
have a careful assessment with history, phys-
than 7 days, pulmonary infiltrates are very
ical examination, and pertinent laboratory
likely to be of bacterial origin. If the neu-
studies including blood cultures from any
tropenia persists for greater than 7 days,
in‐dwelling catheters. Patients with localiz-
the patient is at particularly high risk for
ing findings should have appropriate diag-
fungal infection. BAL or induced sputum
nostic procedures (stool cultures in patients
may identify Pneumocystis, viruses, or
with diarrhea, brain imaging, and possible
fungi. The risks and benefits of open lung
LP for patients with mental status changes,
biopsy and pursuing an aggressive diag-
etc.). Clinical judgment must be utilized to
nostic evaluation must be considered. It is
determine the risk for the patient without an
important to remember that the child may
obvious source of infection. Issues in the
rapidly become very ill and this decision
social history such as family reliability, com-
must be made expeditiously after discus-
pliance, and ability to travel to the outpa-
sion among the oncologist, surgeon, radi-
tient center should be factored into a
ologist, pulmonologist, as well as the
decision plan. If the patient has recently
patient and family.
received chemotherapy and is expected to
reach a nadir in the next few days, consid-
Other supportive measures eration should be given to more frequent
assessment or hospital admission given
Granulocyte colony‐stimulating factor (G‐ impending neutropenia and a blunted
CSF) may be indicated in the setting of FN; response to infection.
however, there are no specific standards for As with the neutropenic patient, blood
its use. A standard dose of 5 mcg/kg/day sub- cultures should be drawn and empiric anti-
cutaneously (or IV) is recommended under biotics initiated. The risk for Pseudomonas
certain conditions in which the clinical is significantly decreased in the patient
course is likely to worsen before the antici- without severe neutropenia and therefore
pated marrow recovery (see Chapter 25). ceftriaxone IV/IM at 50 mg/kg (maximum
Patients who remain severely neutropenic 2 g) can be given daily while febrile in the
with life‐threatening infections should outpatient setting in the otherwise well‐
receive G‐CSF. Granulocyte transfusions appearing child for at least the first 48 hours,
may be considered in the setting of profound while awaiting results of the blood
neutropenia and sepsis, especially in the face culture(s). Hospitalization may be required
of an invasive bacterial or fungal infection in patients who are persistently febrile with
that is not improving on appropriate therapy a dropping ANC and in those with a posi-
with expected continuation of profound tive blood culture or the development of
neutropenia (see Chapter 5). localizing signs.
346 Chapter 27

improving care and focusing research. J. Clin.


Suggested reading Oncol. 30: 4292–4293.
Freifeld, A.G., Bow, E.J., Sepkowitz, K.A. et al.
Lehrnbecher, T., Robinson, P., Fisher, B. et al. (2011). Clinical practice guideline for the use
(2017). Guideline for the management of fever of antimicrobial agents in neutropenic patients
and neutropenia in children with cancer and with cancer: 2010 update by the infectious dis-
hematopoietic stem cell transplant recipients: ease Society of America. Clin. Infect. Dis. 52:
2017 update. J. Clin. Oncol. 35: 2082–2094. e56–e93.
Pulsipher, M.A. (2012). Pediatric specific guide-
lines for fever and neutropenia: a ­catalyst for
28 Acute Pain
Management in the
Inpatient Setting
Pain is a common problem for children with that physicians could use when developing
hematologic or oncologic conditions, as it treatment plans for cancer pain, paving the
can result both from the condition itself and way for significant improvements in its man­
from its management. A number of studies agement. Over the subsequent 30 years, it
have demonstrated that effective pain man­ has undergone much scrutiny and several
agement not only increases a patient’s com­ modifications have been proposed. A recent
fort level but can also effect long‐term adaptation is presented in Figure 28.1.
changes in a patient’s pain threshold, and, in The cornerstone of the ladder rests on
critically ill patients, it has been demon­ five simple recommendations that remain
strated to improve morbidity and mortality. valid today:
Despite this, other studies have demon­ 1. The oral form of medication should be
strated that pediatric pain management is chosen whenever possible.
often suboptimal. This results from percep­ 2. Analgesics should be given at regular
tions on the parts of both parents and care intervals. It is necessary to respect the
providers that pain is less frequent or less ­duration of the medication’s efficacy and to
severe than is the case, or from concerns of prescribe doses at specific intervals in
causing dependence or addiction. accordance with the patient’s level of pain.
The management of chronic pain is a fre­ The dose should be adjusted until the
quent challenge for pediatric hematologists patient is comfortable.
and oncologists, but is outside the scope of 3. Analgesics should be prescribed accord­
this chapter. For pain management at the ing to pain intensity as evaluated by a scale
end of life, see Chapter 29. For pain manage­ of intensity of pain. The prescription must
ment in patients with sickle cell disease, see be given according to the level of the patient’s
Chapter 3. pain and not according to the medical staff ’s
perception of the pain.
4. Dosing of pain medication should be
The World Health Organization adapted to the individual. The correct
analgesic ladder dosage is one that will allow adequate relief
of pain. The dosing should be adapted to
In 1986, the World Health Organization pre­ achieve the best balance between the analge­
sented the analgesic ladder as a framework sic effect and the side effects.

Handbook of Pediatric Hematology and Oncology – Children’s Hospital & Research Center Oakland,
Third Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
© 2021 John Wiley & Sons Ltd. Published 2021 by John Wiley & Sons Ltd.
348 Chapter 28

STEP 4
Neurosurgical
procedures Nerve block
Epidurals
STEP 3 PCA pump
Acute pain Neurolytic block therapy
Chronic pain without control Spinal stimulators
Strong opioids
Acute crises of chronic pain
Methadone
STEP 2 Oral administration
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,
nonsteroidal anti‐inflammatory drug; PCA, patient‐controlled analgesia.

5. Analgesics should be prescribed with a made its measurement a challenge. As a


constant concern for detail. The regularity result, a variety of pediatric pain assessment
of analgesic administration is crucial for the scales have been developed to assist in
adequate treatment of pain. this process.

It is important to remember that these are Neonates and infants


guidelines and that there are situations The Neonatal Infant Pain Scale (NIPS),
where a stepwise approach to pain is not developed at the Children’s Hospital of
the best choice for management. For exam­ Eastern Ontario, is a widely used method of
ple, management of sickle cell vaso‐occlu­ assessing pain in this patient population. It
sive crises typically involves early evaluates a series of six parameters, with a
introduction of step 3 medications without range of scores between 0 and 7. Scores
waiting for the patient to fail steps 1 and 2 above 3 are considered indicative of a pain
therapies. level sufficient to require intervention. The
scale is presented in Table 28.1. Its major
limitation is that it may underestimate the
Assessment of pain level of pain in infants who are too ill to
respond appropriately or are receiving a
The accurate and reproducible assessment paralyzing agent.
of pain is essential to effective management,
allowing the provider to determine when Preverbal/Nonverbal children
intervention is necessary and to more Pain assessment for children who are una­
objectively measure the response to inter­ ble to describe their pain or use an interac­
vention. However, the subjective nature of tive pain assessment scale due to young age,
pain and the impact of developmental stage cognitive dysfunction, injury, or medical
­
on the best method of assessment have interventions such as intubation requires an
Acute Pain Management in the Inpatient Setting 349

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 —

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
University of Michigan as a tool to assess Figure 28.2. Developed in the early 1980s,
postoperative pain, it has been shown to it remains one of the most widely used
reliably measure pain in a variety of clinical tools for assessing pain in both children
situations. It is presented in Table 28.2. The and adults.
range of possible scores is from 0 to 10, con­ When pain assessment scales are used
sistent with other commonly used numeri­ appropriately, they can be very valuable in
cal rating scales used for older children. helping the care team know whether their

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
350 Chapter 28

0 2 4 6 8 10
No pain Little pain Mild pain Moderate pain Severe pain Worst pain
Figure 28.2 The Wong–Baker FACES Pain Rating Scale.

pain management interventions are appro­ can be relatively decreased up to 1 year of


priate or need to be adjusted. However, there age. Hepatic enzymes are initially immature,
is no “magic number” that indicates when but these mature quickly. Children 2–6 years
interventions need to be initiated. Studies of age develop a larger relative hepatic size
consistently show that there is little correla­ for weight and often metabolize drugs more
tion between a child’s pain score and their quickly than younger or older children; this
perception as to whether or not they need can translate to a need for larger doses of
pain medication. Therefore, in situations drug given more frequently to achieve ade­
where patients are likely to have pain, it is quate analgesia. Variations in age, weight,
essential to question them regularly regard­ blood flow, and organ function can all affect
ing their need for medication. effective drug dosing.

Pain pharmacology Step 1 therapy: nonopioid


analgesics
Pain can be subdivided into two categories,
nociceptive and neuropathic. Acute pain in Drugs that are commonly used for step 1
children is most often nociceptive, and a vari­ therapy of pain in children are presented in
ety of drugs can be used to treat this pain. Table 28.3. These should be first‐line ther­
Much of the information regarding this ther­ apy for pain unless the clinical situation
apy is empiric, as there are few formal drug dictates that therapy should begin at a
studies for many of these agents, especially in higher step.
children less than 12. Nonsteroidal anti‐ The use of NSAIDs in children with
inflammatory drugs (NSAIDs), opioids, cancer is generally discouraged due to the
muscle relaxants, local anesthetics, and tram­ common occurrence of thrombocytopenia
adol can be used. Some other adjuvant thera­ in this population and concern for an
pies can also be utilized, such as psychological increased risk of bleeding due to an inhibi­
intervention, distraction, and biofeedback. tion of platelet function. However, there
When treating pain in neonates, it is have been no large, controlled trials dem­
important to remember that several factors onstrating the validity of this concern.
can modify their response and clearance of Several clinical trials have demonstrated
drugs. They have increased body water con­ that ibuprofen can safely be used following
tent and decreased fat, which can alter drug dental surgery, tonsillectomy, and neuro­
distribution. A relative increase in blood surgery in healthy children and in vitro
flow to the brain and a somewhat “leaky” data suggest that ibuprofen causes no sig­
blood–brain barrier can cause a prolonga­ nificant prolongation of the platelet func­
tion of drug effect. Renal clearance of drugs tion assay, PFA‐100.
Acute Pain Management in the Inpatient Setting 351

Table 28.3 Step 1 pain therapy medications.

Drug Dose Comments

NSAIDs • Block conversion of


Ibuprofen 1–3 months: 5 mg/kg 3–4 times daily arachidonic acid into
3 months–1 year: 50 mg 3 times daily prostaglandins and
1–4 years: 100 mg 3 times daily 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 • Can be hepatotoxic
4 grams/day • Avoid rectal dosing in
neutropenic patients
• Parenteral form recently
licensed in United States

Abbreviations: NSAIDs, non‐steroidal anti‐inflammatory drugs; IV, intravenous;


IM, intramuscular.

pruritus, constipation, physical tolerance,


Step 2 therapy: weak opioids
and dependence. Although the Food and
Drug Administration (FDA) classifies them
Patients whose pain is inadequately con­
as Schedule III medications, the risks of
trolled with step 1 therapy or who are being
abuse and diversion still exist. In addition,
weaned from step 3 therapy require treat­
in 2013, the FDA added a black box warning
ment with step 2 agents. In the United States,
contraindicating the use of codeine after
the most common agents used are those that
tonsillectomy.
combine acetaminophen with a weak opi­
oid. These include codeine or hydrocodone,
although there is some disagreement about
the inclusion of hydrocodone in the weak Step 3 therapy: strong opioids
opioid category. Commonly used agents in
this category are presented in Table 28.4. Patients not responding to step 2 therapy or
It is important to remember that the des­ whose condition indicates the need for
ignation “weak opioid” does not translate to stronger pain therapy at the outset are can­
“without side effects.” The use of codeine didates for step 3 therapy. All of the medica­
and hydrocodone can cause the same side tions in this category are strong opioids.
effects seen with stronger opioids: respira­ They act by binding to μ‐receptors in the
tory depression, hypoxia, nausea, vomiting, spinal cord and central nervous system
352 Chapter 28

Table 28.4 Step 2 pain therapy medications.

Drug Dose Comments

Codeine 0.5–1 mg/kg q4–6 hours • 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 hours 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 • Tablet: 5 mg/500 mg
hydrocodone hydrocodone • Liquid: 7.5 mg/500 mg per
<40 kg: do not exceed 5 mg 15 ml
hydrocodone per dose • Elixir contains 7% alcohol
>40 kg: do not exceed 7.5 mg
hydrocodone per dose

(CNS). These receptors are also found with chronic opioid utilization can be miti­
throughout the body, and binding to periph­ gated through utilization of methadone or,
eral sites accounts for many of the side potentially, ketamine. However, these steps
effects seen with opioid therapy. Opioids should be performed under the guidance of
vary both by their duration of action and by individuals who are well‐versed in such
the emotional effect they produce. measures. Transdermal fentanyl has not
Meperidine and oxycodone typically cause been included; practitioners familiar with its
euphoria; conversely, morphine more com­ usage should transition patients to this agent
monly causes dysphoria. Equivalent doses of when deemed necessary.
the strong opioids and the pharmacokinet­
ics of their oral formulations are presented
in Table 28.5. Step 4 therapy
Oral starting doses of step 3 medications
are presented in Table 28.6. It is important to As can be seen in Figure 28.1, step 4 therapy
remember that patients with severe chronic does not introduce new medications.
pain will often be on doses much larger than Patient‐controlled analgesia (PCA) pumps
this due to tolerance. Tactics such as opioid are listed; this does not refer to short‐term
rotation can be used to deal with tolerance PCA pump use such as following surgery or
or with side effects resulting from large to deal with postchemotherapy mucositis or
doses. N‐methyl‐D‐aspartate (NMDA)‐ sickle cell vaso‐occlusive crisis. Rather, it
receptor upregulation leading to tolerance refers to long‐term, ambulatory PCA pump
Acute Pain Management in the Inpatient Setting 353

Table 28.5 Strong opioid characteristics.

Equianalgesic doses (mg) Pharmacokinetic profile (oral


formulations unless specified)
Medication IV PO Onset Duration
Morphine sulfate 10 30 IV: 2–4 minutes IV: 2–4 hours
IR 20–30 minutes 3–6 hours
CR (MS Contin) 2–4 hours 8–12 hours
ER (Kadian) 1–2 hours 12–24 hours
Hydromorphone 1.5 4.5 20–30 minutes 2–4 hours
Oxycodone 20 20–30 minutes 4–6 hours
IR 2–4 hours 8–12 hours
CR (for regular, not as 2–4 hours 8–12 hours
needed (PRN) use)
Fentanyl 180 mg oral morphine/24 hours = 100 mcg transdermal fentanyl/hour TD: 12–16 hours TD: 48–72 hours
1 mg IV morphine = 10 mcg IV fentanyl IV: 1–5 minutes IV: 0.5–2 hours
Methadone Conversion ratios: • Interindividual variability exists;
• Oral:IV = 2:1 methadone should be used by
• Oral morphine: methadone based on 24‐hour morphine total experienced clinicians only
24‐hour oral morphine Oral morphine: methadone ratio • Doses may need to be decreased after
total (mg) several days of administration; monitor
vital signs daily and consult a specialist
<30 2:1
• May cause QT interval prolongation at
31–99 4:1 higher doses
100–299 8:1
300–499 12:1
500–999 15:1
>1000 20:1

Abbreviations: IV, intravenous; IR, immediate release; CR, controlled release; ER, extended release; TD, transdermal.
354 Chapter 28

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 hours
• Sustained release: for patient
taking >20 mg/day of oxycodone
can administer 10 mg q12 hours
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 hours (mg/ml): 1, 5
PRN for immediate release tablets • Oral solution (mg/ml): 2, 4, 20
or 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 hours; max: 5 mg/dose
Methadone 0.03–0.08 mg/kg/dose PO
q4–6 hours; max: 5 mg/dose

Abbreviations: IR, immediate release; ER, extended release.

use for severe chronic pain. With the developmentally appropriate terms, mini­
increased availability of highly concentrated mizing wait times once the patient arrives in
oral narcotics and novel delivery systems, the operating room, child‐friendly waiting
the use of home PCA therapy has declined spaces, and cognitive behavioral therapies
significantly. Additional step 4 therapies (CBT) such as directed imagery, deep
include nerve blocks, epidural injections, breathing, and role playing. These interven­
and other alternative interventions routinely tions are typically provided and coordinated
provided by a specialized pain service. by the child life program.
●● The use of CBT, relaxation therapy, and

biofeedback to decrease the perception of


Nonpharmacologic approaches pain among children suffering from chronic
to pain pain caused by cancer, headache, and
abdominal pain.
The effectiveness of psychological inter­ ●● The use of CBT, relaxation, self‐hypnosis,

ventions to decrease the perception of pain biofeedback, and social support groups to
and the anxiety associated with it, and to decrease painful episodes and improve qual­
improve the quality of life in patients with ity of life in patients with sickle cell disease.
chronic pain, has been clearly demon­ It is vital that child‐life specialists become
strated in a number of clinical settings. involved with children admitted with a pos­
These include: sible diagnosis of cancer very shortly after
●● Minimizing postoperative pain and anxi­ admission. Data indicate that psychological
ety through the use of preoperative inter­ interventions are most effective in decreas­
ventions before painful procedures. These ing anxiety related to painful procedures
include the explanation of the event in when initiated before the first painful
Acute Pain Management in the Inpatient Setting 355

experience. It is also important to obtain additional pathology, the next step is to


input from a psychologist early on for better understand his pain and what inter­
children with conditions causing chronic ventions have been effective in the past.
pain so that appropriate interventions can For the patient with frequent pain, there
be started. ideally would be a pain contract or pain
plan agreed upon as an outpatient when
well that would be available for the inpa­
Case study for review tient practitioner as a guide for pain
management.
You have been following a now 13‐year‐old, b. What pain medications would it be
40‐kg male with sickle cell disease (hgb SS) important to begin?
since he was a newborn. In the first year of Opioids remain the mainstay of pain man­
life, he had a few episodes of dactylitis which agement for vaso‐occlusive events (VOE). It
required pain medicine and hospital admis­ is important to determine which opioid has
sion. He then had a relatively asymptomatic been effective in the past with the least side
period of time until the last 24 months. Over effects. Many patients will be treated effec­
the last two years, he has had multiple tively with morphine, while ­others will pre­
admissions for pain control secondary to fer hydromorphone due to improved pain
chest and back pain. His admissions for pain control with decreased nausea. It is impor­
have become more complex and of longer tant to be aggressive at the outset to get on
duration during this time. He is now again top of the pain and help to break the pain
admitted for chest pain. cycle. This will allow for more rapid decrease
a. What are some initial considerations? in pain medication dosing and overall less
It is not uncommon for patients with pain medication requirement. In the non‐
sickle cell disease to have increasing pain opioid naïve patient, there is minimal to no
during the adolescent years after a period risk of respi­ratory depression with giving
of relatively less symptoms. This can be 0.1 mg/kg morphine equivalents as the ini­
exacerbated by multiple different factors. tial IV bolus doses in the emergency depart­
It would be first important to assess that ment (ED). In this particular case, the
there are no other concerning findings for patient should receive 4 mg doses of IV
his symptoms other than pain, therefore it morphine (or approximately 0.4 mg IV
is important to check a chest X‐ray to rule hydromorphone) based on his weight of
out acute chest syndrome as well as check­ 40 kg. Ketorolac should also be initiated as
ing a baseline CBC with reticulocyte an adjuvant in the ED. For patients with fre­
count and complete metabolic panel quent admissions for pain as well as chronic
(CMP) to ensure he is not having a hyper­ opioid ­utilization, we also utilize bolus dos­
hemolytic episode which would be repre­ ing of ketamine in the ED as well as low‐
sented by a drop in his hemoglobin from dose ketamine infusion on the floor to assist
baseline with compensatory elevation in with pain control and decrease need for opi­
his reticulocyte count as well as increase oids. Patients chronically using opioids have
in indirect bilirubin from baseline. WBC upregulation of the NMDA receptor making
count and platelets are often elevated with opioids less effective. Ketamine works syn­
acute vaso‐occlusive pain crisis. If these ergistically with opioids by antagonizing the
studies are reassuring and point to an NMDA receptor in addition to anti‐anxiety
acute vaso‐occlusive crisis rather than and antidepressant properties.
356 Chapter 28

c. What medications should be contin­ immersive virtual reality therapy, and acu­
ued when the patient is admitted to the puncture (as available).
inpatient floor? e. What other considerations are impor­
If the patient is not able to achieve pain tant in the patient with frequent pain
control after two to three doses of IV opi­ admissions?
oids in the ED, admission is likely required. This patient has developed chronic pain,
This should be a discussion between the which requires complex management by a
practitioner and the patient to help deter­ multidisciplinary team. Psychology should be
mine what is the best course. If the patient involved to help explore stressors and other
is being admitted, he should be transi­ pain exacerbators and how to mitigate these
tioned to patient‐controlled analgesia. We symptoms. Psychology may be able to provide
typically give 0.05 mg/kg of morphine biofeedback and cognitive behavioral therapy
equivalents as the push dose without a to help patients better understand their pain
basal infusion. We give a 10‐minute lock­ as well as methods to decrease their pain.
out based on the onset of action (i.e., Regular follow‐up is required. There may be
5–10 minutes for IV morphine) and two benefit to being on chronic pain medications
push doses per hour. This would be (i.e., methadone, Neurontin, amitriptyline,
reported out as 2/0/10/4 (i.e., push dose/ and others) or antidepressants. Involvement
basal dose/lockout time/hourly maxi­ of a pain specialist is also vital. Utilizing thera­
mum). Note these are guidelines and there pies which minimize opioid utilization as
must be individual adjustment as needed much as possible are important. In the patient
based on the patient’s prior history of with sickle cell disease, it is also important to
response as well as pain plan (if avai­ initiate medical therapies to aid in decreasing
lable). Adjuvant medications including pain such as chronic transfusion or hydroxyu­
acetaminophen and ketorolac should be rea therapy. Practitioners should be familiar
continued. We utilize oral diphenhy­ with the concepts of allodynia and hyperalge­
dramine which can treat pruritus with IV sia and recognize that chronic pain presents
opioids in addition to being synergistic for very differently than acute pain. There
pain control. remains significant implicit and explicit bias
d. What additional medications and in the treatment of patients with chronic pain,
therapies should be initiated? and the practitioner must be cognizant of
Consideration for side effects of pain med­ these issues which may impact the patient
ications must be considered and therapy from receiving ideal medical management.
initiated. This includes a bowel regimen
for prevention of opioid‐induced constipa­
tion as well as a H2 blocker for gastric pro­ Multiple choice questions
tection with toradol. The patient should be
started on incentive spirometry to pre­ 1. You are taking care of a 5‐year‐old boy
vent atelectasis and the development of with metastatic neuroblastoma. He has
acute chest syndrome (ACS). The patient diffuse bony metastases and is complaining
should be encouraged to get out of bed as of pain in his extremities which he describes
much as possible. Warm packs may be of as “throbbing and sharp.” Based on this
benefit. Complementary therapies should presentation and description, his pain is
also be initiated, as available. We typically most likely to be:
utilize transcutaneous electrical nerve a. Somatic
stimulation (TENS), massage therapy, b. Visceral
Acute Pain Management in the Inpatient Setting 357

c. Neuropathic in adult studies for diabetic neuropathy.


d. None of the above Ketamine is a NMDA receptor antagonist
Explanation: While there are several catego­ and low dose ketamine can be beneficial in
ries of pain, visceral and somatic pain are neuropathic pain. Strong pain stimuli acti­
two of the main subtypes. Pain from bony vate NMDA receptors causing hyperexcita­
metastases is typically somatic pain, often bility of dorsal root neurons. This induces
described as “sharp, squeezing, throbbing.” central sensitization, wind‐up phenomenon
On the contrary, visceral pain is not well and pain memory. NMDA‐receptor antago­
defined and the patient often uses the terms nism may prevent the induction of central
“dull, aching” to describe it. Visceral pain is sensitization caused by stimulation of
caused due to stretching, blockage or irrita­ peripheral nociception, as well as block the
tion of visceral organs in the chest, abdomen wind‐up phenomenon. The answer is f.
and pelvis. The answer is a.
3. You are seeing a 15‐year‐old boy with
2. You are taking care of a 7‐year‐old girl diffusely metastatic multiply relapsed rhab­
with rhabdomyosarcoma who is receiving domyosarcoma. The family has agreed to
chemotherapy with a regimen that includes allow natural death and is he is now receiving
vincristine. She has been increasingly com­ home hospice care. His pain is increasingly
plaining of “burning” pain in her hands and difficult to control. He has had chronic
legs. Which of the following are appropriate diffuse body pain and is on > 60mg of oral
treatment options for her? morphine a day. He is also having trouble
a. Venlafaxine swallowing pills and liquids. A good analge­
b. Amitriptyline sic option for him would be:
c. Gabapentin a. Subcutaneous morphine
d. Tramadol b. Oral methadone
e. Low dose ketamine c. Transdermal fentanyl
f. All of the above d. Intranasal fentanyl
Explanation: This patient is describing Explanation: Fentanyl is the only opioid
neuropathic pain most likely caused due to available in the transdermal formulation in
chemotherapy induced peripheral neuropa­ the United States. It is highly potent (about
thy. The most likely chemotherapeutic agent 80‐100 times more than morphine).
involved in her case would be vincristine. Transdermal fentanyl is not used as the ini­
Neuropathic pain in children with cancer tial opioid for pain management in the opi­
can be treatment‐related (such as due to oid naïve patient. It is a good option for
Vinca alkaloids) or due to the disease pro­ patients with chronic pain who are tolerant
cess itself. Neuropathic pain can be difficult to high doses of morphine. A child must be
to describe, especially for younger children. tolerant to 30–60 mg per 24‐ hours oral
Typical descriptions can include “burning, morphine equivalent before being able to be
numbing, stabbing.” Physical exam may rotated to the smallest fentanyl patch, 12.5
reveal signs of peripheral neuropathy. Treat­ mcg/hr. Due to pre‐set patch doses, it can be
ment options for neuropathic pain include difficult to titrate the dose of a fentanyl
anti‐epileptics (such as gabapentin), TCAs patch. Because of a long onset time, inability
(amitriptyline) and certain opioids such as to rapidly titrate drug delivery and long
tramadol. Serotonin‐norepinephrine recep­ elimination half‐life, transdermal fentanyl is
tor uptake inhibitors such as venlafaxine contraindicated for acute pain management.
(Effexor) have been shown to be beneficial The patient in the vignette clearly has
358 Chapter 28

chronic pain, high opioid tolerance and dif­ combinations of medical providers, nurses,
ficulty taking oral medications. Transdermal social workers, psychologists and spiritual
fentanyl patch will be a good analgesic option personnel. They are equipped to provide
in this situation. Oral methadone is the ideal comprehensive care for the child’s body,
choice in the patient still able to take oral mind and spirit. Providing support to the
medications. Intranasal fentanyl is very family is also a major function of a pediatric
short acting and is only beneficial in acute palliative care team. This support can also
pain scenarios. The answer is c. extend after the death of a patient with
bereavement care. The answer is a.
4. You are taking care of a 4‐year‐old boy
with newly diagnosed medulloblastoma. He 5. You are following an inpatient with
underwent a gross total resection with a VP chronic, worsening pain from bony metas­
shunt placement and is due to begin chemo­ tases. The patient is currently on high dose
therapy in a few weeks. His family requests a dilaudid PCA with a basal rate of 2 mg and
palliative care consult and has several pushes of 1 mg. The patient is pushing the
questions for you. All the following are true PCA about 15 times per day with tolerable
regarding pediatric palliative care EXCEPT: pain. The patient is starting to have worsen­
a. Palliative care is not an option when ing opioid side effects including myoclonus
a child is receiving disease directed from the high dose PCA. You are planning
treatment on switching to PO methadone so that the
b. Palliative care teams can provide patient can be discharged for home hospice.
comprehensive physical, mental and What is the most appropriate BID oral dos­
spiritual support to the child as well as ing of methadone given the patient’s daily
his family dilaudid requirement?
c. Palliative care can be initiated at the a. 10 mg
time of diagnosis of a chronic illness, b. 30 mg
clinical worsening of an illness or at the c. 50 mg
end of life d. 100 mg
d. Palliative care can be provided in an e. 200 mg
inpatient, outpatient or home setting Explanation: When calculating opioid con­
Explanation: WHO has laid down guiding version it is best to first consider the mor­
principles and definitions for pediatric pal­ phine equivalence and then re‐convert to
liative care. Pediatric palliative care be initi­ the new opioid. In this case, the patient is
ated at the time of diagnosis of a chronic or taking 63 mg of dilaudid per day (24 hours ×
life threatening health condition. Receiving 2 mg + 1 mg x 15 pushes). The morphine to
disease directed treatment is NOT a con­ dilaudid conversion is 6.67 (see Table 28.5).
traindication to simultaneously receive pedi­ Therefore, the daily IV morphine equivalent
atric palliative care. While it can be easy to is 420 mg of morphine, which is 1260 mg of
confuse pediatric palliative care with “end of oral morphine (IV to oral morphine conver­
life” or “hospice care,” these are in fact sub‐ sion of 1:3). Therefore, based on Table 28.5,
categories of pediatric palliative care. For the oral morphine of >1000 mg converts at a
patient in our clinical vignette, a pediatric ratio of 20:1 with methadone, which in this
palliative care consult can be initiated now if case is 1260 divided by 20 or 63 mg of oral
the family so wishes. The palliative care team methadone per day (note this is only coinci­
is usually multi‐disciplinary and can include dentally the same dose of IV dilaudid). Also
Acute Pain Management in the Inpatient Setting 359

note the 2:1 oral to IV conversion ratio for


Suggested reading
methadone. Methadone will concentrate in
fat over time allowing for steady state and
Friedrichsdorf, S.J. and Kang, T.I. (2017). The
effective daily dosing (somewhat less likely management of pain in children with life‐
in the cachectic cancer patient). Patients limited illnesses. Pediatr. Clin. North Am. 54:
should generally be started on twice or 645–672.
thrice daily methadone and then weaned to Verghese, S.T. and Hannallah, R.S. (2010). Acute
daily or twice daily methadone at the same pain management in children. J. Pain Res. 3:
total daily dose. The answer is b. 105–123.
29 Palliative Care

The World Health Organization defines whose cancers have an excellent prognosis
pediatric palliative care as care that “aims to with appropriate therapy. It also applies to
improve the quality of life of patients facing the large number of children with life‐
life‐threatening illnesses, and their families, threatening non‐oncologic conditions. The
through the prevention and relief of definition stresses the importance of “early
suffering by early identification and identification and treatment of pain and
treatment of pain and other problems, other problems.” Palliative care is not
whether physical, psychosocial, or spiritual.” synonymous with “end‐of‐life” care;
It is the active total care of the child’s body, palliative care must begin at the time of
mind, and spirit, and also involves giving diagnosis and continue on throughout the
support to the family. It begins when illness period of disease‐directed therapy. It also
is diagnosed, and continues irrespective of must include many resources from
whether or not a child receives disease‐ throughout the patient’s and family’s range
directed treatment. It requires that health of experience: physicians, nurses, social
providers evaluate and alleviate a child’s workers, psychologists, spiritual counselors,
physical, psychological, and social distress at and a variety of other support personnel
all times during their treatment. To be chosen because of each child’s unique
effective, it requires a broad multidisciplinary situation. During any hospitalization, an
approach that includes the family and makes important responsibility of the medical
use of available community resources. team is to ensure that each child has access
However, it can be successfully implemented to all needed services and is being supported
even if community resources are limited. to the greatest extent possible.
Palliative care can and should be provided in
tertiary‐care facilities, community health
centers, and at home. Individualized care planning
This definition contains several concepts and coordination
that deserve consideration. It contains the
broadly defined term “life‐threatening” that At the time of diagnosis, parents and car-
includes situations in which a cure is egivers of children with life‐threatening ill-
possible, instead of the more limited “life‐ ness have two goals: a care‐directed goal of
limiting” that includes only those conditions cure and a comfort‐directed goal of decreas-
for which there is no realistic hope of cure. ing suffering. Introducing palliative care
There is a role for palliative care for all principles early on in the care process is
children diagnosed with cancer, even those respectful and supportive of these goals. A

Handbook of Pediatric Hematology and Oncology – Children’s Hospital & Research Center Oakland,
Third Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
© 2021 John Wiley & Sons Ltd. Published 2021 by John Wiley & Sons Ltd.
362 Chapter 29

model developed by the Palliative and End‐ options are acceptable to the patient and
of‐Life Care Task Force at St. Jude Children’s family given the prognosis and goals of treat-
Research Hospital, the Individualized Care ment. Key concepts in this model include the
Planning and Coordination Model is establishment of a close and caring relation-
designed to facilitate this introduction ship with the patient and family, clarifying
(Figure 29.1). Although developed to assist patient and family values and priorities,
in making care decisions for children under- determining the goals of care based on the
going bone marrow transplantation, the patient’s and family’s understanding of prog-
model is useful in helping to define what care nosis, and allowing them to choose from

Eligibility

Step 1 : Relationship

Understanding Sharing
illness Relevant
Experience Information

Needs assessment

Step 2 : Negotiation

Prognosis Goals

Treatment
options

Step 3 : Plan

Medical plan Life plan

Comperhensive
Care Plan

Action
(Care Coordination)
Figure 29.1 The Individualized Care Planning and Coordination Model. Source: From Justin N. Baker,
Pamela S., Integration of Palliative Care Practices into the Ongoing Care of Children with Cancer:
Individualized Care-Planning and Coordination, Pediatric Clinics of North America Pediatric
Clinic, 2008. Reproduced with permission from Elsevier.
Palliative Care 363

available goal‐directed treatment options. bereavement process are not totally clear.
From this foundation, the members of the Studies have also demonstrated that
care team can coordinate the provision of ­significant levels of parental dissatisfaction
care to ensure the social, psychological, and with hospital staff can arise from confus-
emotional needs of the patient and family are ing, inadequate, or uncaring communica-
addressed at the same time that appropriate tion regarding treatment and prognosis as
medical care is provided. well as discrepancies between parents and
care providers in understanding the termi-
nal condition. When a language barrier
exists between family and staff, these prob-
End‐of‐life care lems are often exacerbated.

Despite the tremendous advances made in


the care of children with cancer over the
past 50 years, at the present time one in five Common symptoms at the end
children diagnosed with cancer will ulti- of life
mately die of their disease. The death of a
child affects the physical and psychological Pain
well‐being of family members for the rest of Parents and caregivers report that their
their lives. Events occurring around the child’s pain is the symptom they fear most. A
time of death, both positive and negative, large survey of parents whose children died
play a critical role in defining how family of cancer revealed that 75% of them felt their
members grieve and ultimately come to child suffered pain at the end of life, but only
terms with the event. Families who have lost 37% felt the pain was successfully managed.
a child have identified several needs regard- The principles of pediatric pain man-
ing end‐of‐life care. These include the need agement are presented in Chapter 28 and
to have complete information honestly com- pain management at the end of life follows
municated, to have easy access to essential these same principles. However, it becomes
staff members who will be supportive, to particularly important to clarify and
have assistance in coordinating necessary understand the goals of treatment as eluci-
services, to have their relationship with their dated by the patient and parents. The desire
child maintained as much as possible, and to to maintain alertness and interaction early
be allowed to feel that their child’s life and in treatment may give way to a desire to
death have meaning. keep the patient comfortable, even if it
Research studies involving pediatric means the patient will be more sedated and
palliative care are limited, but they have less aware. Acceding to the family’s wishes
identified several important shortcomings that the patient not be hooked up to moni-
in the level of palliative care provided to tors might outweigh concern that the doses
children. Several studies have demon- of pain medications being used could result
strated a lack of successful management of in respiratory depression. These decisions
pain and other distressing symptoms. often run counter to what physicians see as
Other studies have demonstrated that the their primary goal of care, to prolong life as
majority of children dying of life‐threaten- long as possible. Working through some of
ing diseases die in the hospital, often in the these difficult philosophical issues with the
intensive care unit (ICU). The impact of help of a mentor can be essential to work-
this outcome on the appropriateness of care ing comfortably and effectively in this type
provided and its effect on the family’s of setting.
364 Chapter 29

Nausea and vomiting A stepwise approach to the manage-


Nausea and vomiting at the end of life can ment of nausea and vomiting in the palli-
be triggered by many different stimuli. A ative care setting is outlined in Table 29.1.
variety of toxins such as medications and A key feature of this approach is to ensure
their metabolites, urea resulting from renal that the interventions selected are in
failure, or metabolic byproducts of hepatic keeping with the patient’s and family’s
failure can all stimulate the chemoreceptor goals of care.
trigger zone and cause nausea. Increased A number of medications can be tried to
intracranial pressure (ICP) and a variety of control nausea and vomiting. The ideal
emotional and sensory stimuli can mediate agent is one that targets the specific cause of
nausea and vomiting through cortical path- the patient’s distress if such an agent can be
ways. The gastrointestinal (GI) tract can identified. A list of useful medications is
stimulate the vomiting center in response to outlined in Table 29.2.
local receptor stimulation caused by Opioids frequently cause nausea and
obstruction, stasis, toxins, or drugs. In addi- vomiting due to a direct effect on the chem-
tion, pharyngeal stimulation by mucus or otherapy trigger zone or secondarily to their
mucosal breakdown can also trigger the effect on GI motility leading to gastroparesis
vomiting center. 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,
antibiotics, 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
consistent 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.


Palliative Care 365

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


dopamine antagonist) 6 hours
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
antagonist) or 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 three divided doses
Dexamethasone (anti‐inflammatory) Antiemetic: 10 mg/m2 IV or orally daily; maximum
dose 20 mg daily
Increased ICP: increase dosing frequency to two–four
times daily, with a maximum dose of 40 mg/day
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 activity, neurological dysfunction, and intes-


Constipation often causes pain and distress tinal obstruction due to presence of tumor.
for children at the end of life, both physically
and psychologically. Prevention of constipa- Assessment
tion should therefore be a primary goal of History
end‐of‐life care. ●● Stool frequency, consistency, and associ-

One of the most common causes of con- ated discomfort.


stipation are opioids, which decrease GI ●● Abdominal pain, nausea, vomiting, and

motility and fluid secretion, leading to hard, anorexia.


dry stools. Unlike other narcotic side effects, ●● Neurological symptoms: urinary prob-

constipation does not seem to decrease with lems, lower extremity numbness, weakness,
ongoing use. Patients and families should be and paresthesias.
informed ahead of time about this side ●● Diarrhea (signs of encopresis or
effect, as children in severe pain may not obstipation?).
consider regular bowel movements a prior- ●● Prior episodes, previously effective therapies.

ity. Once well established, constipation may


require quite aggressive and unpleasant Physical Examination
therapies to correct it. ●● Abdominal palpation.
Other factors that contribute to constipa- ●● External rectal exam (digital exam rarely
tion in terminal patients include poor oral needed and may be dangerous).
intake of fluids and food, decreased physical ●● Neurologic exam.
366 Chapter 29

Medications Table 29.3 Appetite stimulants for cancer‐


●● Stool softeners (docusate): rarely effective associated anorexia.
alone.
●● Osmotic agents (MiraLAX, magnesium Drug Dose
sulfate, and lactulose): effective for long‐
Cyproheptadine 2–6 years: 2 mg BID
term use; MiraLAX requires 4–8 oz. of fluid
(Periactin) >6 years: 4 mg BID
which can be a limitation. Megestrol acetate 7.5–10 mg/kg/day in 1–4
●● Stimulants (senna, bisacodyl): often use-
(Megace) divided doses
ful and necessary in the end‐of‐life setting Maximum 800 mg/day
when less concern exists regarding develop- or 15 mg/kg/day
ment of dependence. Dronabinol 2.5 mg/m2/dose every
(Marinol) 4–6 hours
Use with caution in
Anorexia and weight loss children with
depression; avoid in
children with
Anorexia or loss of appetite is a familiar
sensitivity to sesame oil
symptom in children with cancer. It can
occur at any point in treatment; institu-
tional studies indicate that it affects as many
as 40% of children with cancer at diagnosis Support for emotional distress.
and the prevalence increases to as high as Discontinue medications that might be
70% in children with advanced‐stage dis- causing anorexia.
ease. Early in treatment it is most often a
side effect of chemotherapy or radiation, or Step 3: more aggressive
a result of stomatitis, dysphagia, constipa- therapies (if consistent with
tion, pain, or depression. As the child’s can- plan of care)
cer becomes progressive, it is often a Medications (see Table 29.3).
component of the anorexia/cachexia syn-
drome, a complex set of central nervous Nutritional supplementation
system (CNS) and metabolic abnormalities If acceptable to patient and family, enteral
caused by a combination of tumor byprod- feeding with a nasogastrostomy (NG) or
ucts and host cytokine release. gastrostomy tube is often reasonably well
tolerated unless the patient has a GI obstruc-
Step 1: assessment tion or is chronically nauseated.
State of primary disease. Total parenteral nutrition (TPN) is
Diet and fluid history. rarely useful in the end‐of‐life setting
Nausea/vomiting/constipation. except for short‐term use in the patient
Oral lesions (including candidiasis). with GI obstruction.
Medication history.
History of prior eating disorder.
Sense of smell. Fatigue

Step 2: environmental changes The National Comprehensive Cancer


Small meals. Network defines cancer‐related fatigue as
Regular mouth care. “a distressing persistent, subjective sense
Comfortable environment. of physical, emotional, and/or cognitive
Palliative Care 367

tiredness or exhaustion related to cancer Psychosocial and activity‐based inter-


or cancer treatment that is not propor- ventions have been shown to be quite effec-
tional to recent activity and interferes tive in adults with cancer‐related fatigue;
with usual functioning.” It is pervasive, data in children are lacking. Exercise has
multidimensional, and incapacitating; as the strongest data regarding its value, but
such it significantly decreases health‐ this is largely in adult cancer survivors suf-
related quality of life in children. While it fering from fatigue after the completion of
can exist throughout the entire period of curative therapy. Its utility in ameliorating
cancer treatment, fatigue is most often fatigue in patients near the end of life is less
seen as significant as the child’s disease well‐established.
progresses. In several studies, parents
have identified fatigue as the most signifi-
cant symptom that moderately or severely Dyspnea
impacted their child’s well‐being toward
the end of life. These same studies have Dyspnea is the term used to describe the
also demonstrated that it is the symptom feeling of breathlessness that occurs when
most likely to be missed by care providers; the respiratory system fails to meet the
fewer than 50% of children whose parents body’s need for oxygen uptake or carbon
described them as suffering highly from dioxide removal. It occurs under normal
fatigue had this documented in their med- circumstances such as after brisk exercise,
ical record. but in pathological conditions it can cause
Cancer‐related fatigue is more common severe distress. It occurs very frequently at
and more severe in children with brain tumors the end of life, as respiratory failure occurs
than in those with other cancers. Pediatric very commonly as a terminal event.
patients tend more often to associate their feel- Therefore, recognition of dyspnea and its
ings of fatigue to sleep disturbance or side effective treatment are key components of
effects of therapy, whereas parents are more end‐of‐life care.
likely to include emotions and nutritional sta- Dyspnea can result from either an
tus as potential causes. A recent study sug- increased metabolic demand or an inability
gested that pain and dyspnea were frequently to maintain a normal minute ventilation.
associated with fatigue; the authors postulated The former can be caused by complications
that opioids and benzodiazepines, common of the disease or its treatment, such as infec-
therapies for pain and dyspnea, might have sig- tion, metabolic acidosis, or increased meta-
nificantly contributed to their patients’ fatigue. bolic demands caused by extensive tumor
In a review on the assessment and man- burden. The latter can result from conditions
agement of fatigue and dyspnea in pediatric such as decreased lung compliance due to
palliative care, Dr. Christina Ullrich defined tumor, infection, or fluid, or by other condi-
a conceptual model for understanding tions such as interstitial lung disease, ane-
fatigue in this population. This framework mia, or fatigue.
is outlined in Figure 29.2. It follows that treatment of dyspnea
Data are very limited regarding effective would involve steps to decrease metabolic
treatments for cancer‐related fatigue in demand or improve the efficiency of respira-
children, although a number of interventions tion. However, this can prove to be quite
have been shown to be effective in adults. challenging in practice. It is often difficult to
These are outlined in Table 29.4. determine whether the cause of dyspnea is in
368 Chapter 29

Psychosocial factors
• Depression • Familial stressors
• Anxiety • Spiritual concerns
• Fear • Caregiver status
• Boredom • Practical concerns

Sleep disturbance
• Unrelieved symptoms
• Change in environment Fatigue experience
• Circadian rhythm alteration
• Medication effect

Physical factors
• Underlying illness • Metabolic abnormalities
Type, site, stage • Nutritional impairment
Disease-directed treatment Cachexia
• Unrelieved symptoms Dehydration
• Side effects of symptom treatment • Comorbidities
• Changes in muscle Anemia
Deconditioning Infection
Abnormal muscle structure, function Hormone imbalance
Organ dysfunction

Figure 29.2 A model of fatigue in life‐threatening illness. Source: Modified from Fatigue in Children
with Cancer at the End of Life, Journal of Pain and Symptom Management.

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.


Palliative Care 369

fact respiratory or metabolic, and once dis- Support of respiration


covered, the unwillingness of the patient or When intubation and mechanical ventila-
family to accept certain interventions can tion are not appropriate or acceptable to the
create additional challenges for the treat- patient and family, noninvasive positive
ment team. pressure ventilation (NIPPV) can be useful
Appropriate therapies to decrease meta- in supporting respiration. However, the use
bolic demand depend on the cause of the of NIPPV in adult patients who have elected
increased demand. For the patient who to forgo intubation or other aggressive
becomes dyspneic with activity, it may be as respiratory support is controversial. Its use
simple as recommending that the patient use in children with cancer is unstudied; there
a wheelchair instead of walking or decrease have been a handful of papers published
physical activity in other ways. Increased regarding its use in children with neuro-
metabolic demand caused by infection or muscular disorders.
inflammation can be treated with aggressive From the adult literature, it appears clear
use of antibiotics or anti‐inflammatory that the most important issue when deciding
agents, as long as such therapies are in keep- on the use of NIPPV to support respiration
ing with the patient’s and family’s wishes. is a clear understanding of the goals of
Respiratory failure can result from two therapy among all involved. In 2007, the
general causes: lung parenchymal dysfunc- Society of Critical Care Medicine Palliative
tion and respiratory muscle fatigue or fail- Noninvasive Positive Pressure Ventilation
ure. It is important to distinguish between Task Force published its recommendations
these, as the treatments are different. An on the use of NIPPV in the palliative care
algorithm for distinguishing between these setting. While intended for adults, the con-
causes is presented in Table 29.5. It is impor- cepts can be applied to the pediatric care set-
tant to note that hypercarbia can be a late ting as well. The task force proposed a
finding in parenchymal dysfunction as well. three‐category approach to assist in
decision‐making regarding this technology.
Treatment An overview of this approach is presented in
Treatment for dyspnea can be divided into Table 29.6.
three general categories: mechanical sup- The key areas of distinction in this model
port of respiration, support of gas exchange, are goals of care, determination of success,
and therapies to decrease the sensation of appropriate endpoints, and response to
dyspnea. ­failure. It is imperative that each member of

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
• Pleural‐based nodules respiratory system; weakness
• Infiltration of interstitium by metastases caused by systemic disease
Blood gas Hypoxemia Hypoxemia, hypercarbia
findings
370 Chapter 29

Table 29.6 Approach to decision‐making regarding noninvasive positive pressure ventilation.

Approach Category 1 Category 2 Category 3

Definition Life support; no preset limits Life support; do not intubate Comfort measures only
Primary goals of care Assist ventilation and/or oxygenation Includes same as category 1 except Palliation of symptoms (relief
Alleviate dyspnea intubation declined of dyspnea)
Achieve comfort Also could include briefly prolonging
Reduce risk of intubation life for a specific purpose (e.g.,
Reduce risk of mortality arrival of family member)
Avoidance of intubation
Main goals to Goal is to restore health and use Goal is to restore health without using Goal is to maximize comfort
communicate with intubation if necessary and indicated endotracheal intubation and without while minimizing adverse
patient and family causing unacceptable discomfort effects of opiates
Determination of Improved oxygenation and/or Improved oxygenation and/or Improved symptoms
success ventilation ventilation Tolerance of NIPPV
Tolerance of NIPPV or minor Tolerance of NIPPV or minor
discomfort that is outweighed by discomfort that is outweighed by
potential benefit potential benefit
Endpoint for NIPPV Unassisted ventilation adequately Unassisted ventilation adequately Patient is not more comfortable
supporting life supporting life having NIPPV on or wants
Intolerance of NIPPV Intolerance of NIPPV NIPPV stopped
Patient becomes unable to
communicate
Response to failure Intubation and mechanical ventilation Change to comfort measures only and Palliate symptoms without
(if indicated) palliate symptoms without NIPPV NIPPV
Likely location of ICU but may include step down unit or Variable but may include ICU, step Acute care bed but could be
NIPPV acute care bed in some hospitals down unit, or acute care bed applied in hospice by
with appropriately monitored setting appropriately trained
and trained personnel personnel

Abbreviations: NIPPV, noninvasive positive pressure ventilation; ICU, intensive care unit.
Palliative Care 371

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‐naїve 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 4–8 hours for this purpose.
are useful in situations in which the pressure A variety of nonpharmacologic inter-
necessary to maintain airway patency is too ventions have been shown to decrease the
high to allow for efficient exhalation. distress associated with dyspnea. These
In the setting of respiratory failure with- include:
out hypercapnia, supplemental oxygen is the 1. Cool air blown on the face.
most appropriate intervention to facilitate 2. Chest wall percussion.
gas exchange. It is often better tolerated than 3. Relaxation therapy and counseling.
NIPPV devices, especially by young chil- 4. Proper positioning.
dren, facilitating pulmonary vasculature 5. Limitation of activities.
dilatation and inhibiting cerebral artery dil- 6. Cooler room temperature.
atation, a frequent cause of headaches. It is 7. Avoidance of respiratory irritants
important to remember, however, that (tobacco smoke and cleaning fluids).
patients with hypercapnic respiratory failure
frequently rely on a “hypoxic drive” to main- Most experts agree that these interventions
tain respiration. Provision of supplemental work best as an adjunct to pharmacologic
oxygen to these patients can lead to hypo- therapy; they are rarely effective alone.
pnea or apnea.

Therapies to decrease Conclusion


the sensation of 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 dis- destined to die before they reach adulthood.
tress resulting from the sensation of dyspnea These include practical issues such as how
as this occurs. This goal can be achieved with to best manage distressing symptoms such
a combination of pharmacologic and non- as those listed in the preceding text, how to
pharmacologic therapies. best communicate with children about the
The two most effective classes of drugs dying process and involve them in decisions
for minimizing the perception of dyspnea regarding their care, and how to avoid
are opioids and benzodiazepines. Morphine aggressive end‐of‐life therapies that only
is the opioid used most often for this pur- delay death and prolong suffering instead of
pose, but fentanyl and hydromorphone extending life. After the child’s death,
(Dilaudid) have been shown to be effective families must continue to move forward,
as well. The dose that is required is lower raising questions about how best to help
372 Chapter 29

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 disease?
will soon be available in the medical ●● How do you reconcile your responsibility

literature. The children we care for, but can- to be forthright with Julie while simultane-
not save, deserve no less. ously being respectful of her parents’ request?
●● If you believe that her parents’ approach is

not in Julie’s best interest, how might you


Case study for review proceed?
●● Should children with life‐limiting ill-

Julie, a 13‐year‐old girl with widely meta- nesses be involved in end‐of‐life decisions?
static alveolar rhabdomyosarcoma diag-
nosed 6 months earlier, continues to have Understanding familial adaptation toward
extensive bony disease, including possible the dying process is imperative. As health
new metastatic lung lesions. Julie’s treat- care providers, our goal is to anticipate and
ment has been complicated by chronic identify problems and to assist dying chil-
pain and a challenging family situation. dren and their families as they adjust to and
Although Julie lives with her mother, her cope with the emotional responses, which
father oversees her care. Her parents do are part of the dying process.
not get along with one another. They fre- Many children, parents, and physicians
quently argue and scream in her presence, are hesitant to discuss death and dying so
which clearly upsets her. that opportunities for planning how to cope
Since diagnosis, her father has adamantly are often missed. Failure to prepare ade-
refused to let caregivers have one‐on‐one quately for death can deprive families of the
conversations with Julie. He insists on chance to enjoy what time they have left
remaining the only one to relate to Julie any with one another.
disease‐related information, as he knows Each child and family grieve in their own
her best and can gauge how she will react. unique way, determined by personal experi-
He has stated that Julie need not know that ences with death, religious and cultural
she will likely die of her disease and he has backgrounds, and individual makeup.
forbidden the medical team from discussing Dying is a process and all parties need to
issues related to prognosis. learn to live with dying or, as stated by the
Julie is to undergo a CT‐guided lung biopsy American Academy of Pediatrics, “the goal
to determine the nature of the lung lesions. [of palliative care] is to add life to the child’s
Her parents state that she is already anxious years, not simply years to the child’s life” [1].
enough and do not want the medical team to Parents’ natural tendency to protect their
tell her about the biopsy or the possibility that children influences the amount of informa-
her disease has progressed. Instead, they ask tion children receive and the degree to
that Julie be told that she is to be sedated only which they are involved in decision‐making
for a “scan.” Moreover, should she prove to about their care [2]. Some parents feel that
have new disease, they do not want Julie to be by giving their child a choice their responsi-
informed and ask that her caregivers not dis- bilities are being usurped, while others per-
cuss end‐of‐life matters with her. ceive including their sick child in decisions
Palliative Care 373

as overly burdensome. For many parents, their terms. Often, rather than being direct,
the sicker their child, the more they assume a child’s question or statement may be sug-
decisional priority and attempt to minimize, gestive of something they are uncomfortable
veto, or even preclude their child from hav- asking and therefore it is vital to determine
ing a role in decisions at all. Seemingly, such what their underlying intention is and not to
an approach is understandable; however, give too much or too little information or
parents need to be aware of the conse- answer the wrong question. Finally, failure
quences of not preparing their children for to inform and involve children can lead to
medical outcomes that are inevitable, such feelings of isolation and distress [12].
as anxiety, stress, and confusion. Ideally, discussions should occur early
Children frequently know more about and routinely and physicians should reassess
their disease than for which they are cred- how the child and parents understand the
ited. Even when kept in the dark by parents plan and goals of care. This helps the dying
and providers, children often know that the child and parents make appropriate deci-
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 repeatedly. Doing so will help minimize
should be involved in decisions about their misunderstandings and defuse conflicts.
care so that they can voice their prefer- The challenge is to do so in a way that is
ences [6, 7]. Therefore, the question we ask both sensitive and respectful of the child’s,
ourselves should not be “should I tell?” parents’, and providers’ needs, needs that are
rather it should be “how do I tell?” often in conflict with one another.
Increasingly, research supports the need for According to the Institute of Medicine
direct communication between parents, (IOM), “conflicts may . . . be productive or
physicians, and children with life‐limiting beneficial” [11]. Confronted with a disa-
illnesses. This includes discussions relating greement, parties tend to engage in a more
to prognosis and even death [1, 2, 8, 9]. in‐depth discussion allowing for a greater
Hinds et al. [10] found that children with appreciation of each other’s position, and, in
life‐limiting cancer between ages 10 and the process, each side may become more
20 years were capable of participating in sensitive to the other’s values and concerns.
end‐of‐life decision‐making. Mack et al. [13] surveyed parents of chil-
Physicians must be aware of the unique dren with cancer and found that parents
barriers to a child’s ability to participate in rated the quality of care provided by physi-
decision‐making as it relates to the dying cians more favorably when physicians com-
process. The first question we must ask is, municated directly with children (when
“does this child have the ability to understand appropriate). Similarly, in a survey of more
that he/she is dying and what dying entails?” than 400 Swedish parents of children who
As children mature, their intellectual and died of cancer, Kreicbergs et al. [14] found
emotional understanding of serious illness that none of the parents who spoke with
and the prospect of death matures too. their child about death regretted doing so,
Discussions about death should be honest whereas more than one‐quarter of parents
and take into consideration the child’s emo- who did not speak with their child regretted
tional and developmental level. It is impor- not doing so. The latter parent group had
tant to follow the child’s lead [11]. In other higher levels of anxiety and depression than
words, answer what children ask and on parents who did speak to their children.
374 Chapter 29

Parents are not alone in valuing the Case resolution


importance of direct communication Since Julie’s diagnosis, her father has peri-
between children and physicians. Recent odically sought the guidance of the unit’s
research has shown that children with cancer social worker. Prior to the lung biopsy, Julie’s
consider direct communication between father agreed to meet with her oncologist
doctors and children more important than and the social worker in the latter’s office, a
any other aspect [15]. Nearly 40 years ago, place where he is comfortable speaking.
following parental approval, Nitschke Following a brief update by the oncologist of
et al. [4] began including children aged Julie’s current medical status, the social
5 years and older who were near death from worker asked her father to share his con-
cancer in end‐of‐life discussions. They found cerns about Julie not being included in dis-
that the majority of children and parents ease‐related discussions. Her father stated
found the child’s inclusion a positive experi- that Julie is “just a kid,” and that it was “his
ence. They also reported that some children job to protect, and to decide what is best for
from whom information was withheld expe- her.” He recounted his limited parental role
rienced fear and isolation prior to dying. in Julie’s life prior to her cancer diagnosis
and blamed himself for not being more pre-
Conclusions and practical sent in her life, admitting that Julie’s illness
suggestions and likely death “terrify” him and that he is
Situations like Julie’s do not lend themselves resolute not to abandon her again.
to easy solutions. Dishonesty, evading the The social worker asked Julie’s father
question at hand, inconsistencies, and white what he believes she knows of her disease
lies are to be avoided at all costs. Deception and its prognosis. He replied that Julie
is difficult to maintain. When the truth is knows she has cancer and that it is “treata-
ultimately discovered, the patient–parent– ble,” and nothing more. She then asked if he
physician relationship is often irrevocably would allow Julie to join them, which would
damaged, an unfortunate circumstance in permit the oncologist (in his presence) to
any situation, but particularly for a child answer any questions she may have about
such as Julie who is nearing the end of life. the “scan” and to clarify what she indeed
Such action may lead Julie to withdraw and knows. Hesitant at first, he ultimately
even to experience feelings of isolation and agreed. After greeting and thanking her for
fear, let alone profoundly disrespect her joining them, the oncologist asked Julie if
developing autonomy. she has any questions about the upcoming
By helping to facilitate, clarify, and CT scan. Julie asked why she has to have
resolve areas of contention, pediatricians another CT when re-evaluation scans,
can be extremely helpful. As appreciated by including a chest CT, were just completed
the IOM, the following steps may be useful: one week ago. Directing her words to the
(i) postpone decisions to allow for time to oncologist, Julie asked, “is something
think about concerns and to discuss goals; wrong?” Before the oncologist or her father
(ii) rather than making definitive decisions could reply, Julie stated, “I’ve been online
that offer no room for compromise, con- talking to other kids with cancer. A few of
sider temporary steps that accommodate them said that the only reason to repeat a
each party’s goals; and (iii) periodically scan so soon is to see if the disease is back.”
reevaluate each party’s views concerning the Without hesitation and still speaking to her
goals of care and the options to achieve oncologist, Julie said, “I know that most kids
those goals [11]. with my kind of cancer die.”
Palliative Care 375

The social worker encouraged Julie and Liben, S., Papadatou, D., and Wolfe, J. (2008).
her father to speak openly and freely. Paediatric palliative care: challenges and
Tentatively, and with the support of the emerging ideas. Lancet 371: 852–864.
oncologist and social worker, they began to Wolfe, J., Grier, H.E., Klar, N. et al. (2000). Symptoms
and suffering at the end of life in children with
speak to one another and share their respec-
cancer. N. Engl. J. Med. 342: 326–333.
tive concerns and goals. Following their
conversation, Julie’s father cautiously agreed
to include Julie in future discussions. The
References
oncologist assured both Julie and her father
that although he remains hopeful, her dis- 1. American Academy of Pediatrics (2000).
ease is difficult to cure and that she might Committee on bioethics and committee on
indeed die of her cancer. However, the med- hospital care. Palliative care for children.
ical team would continue to treat both her Pediatrics 106: 351–357.
cancer and her pain, all along adhering to 2. Bluebond‐Langer, M. (1978). The Private
her and her parents’ wishes. Worlds of Dying Children. Princeton, NJ:
Successful care of the dying child requires Princeton University Press.
that health care providers acknowledge fam- 3. Hilden, J.M., Watterson, J., and Chrastek, J.
(2000). Tell the children. J. Clin. Oncol. 18:
ilies’ emotions, assure families that their
3193–3195.
responses are normal, provide a balanced
4. Nitschke, R., Meyer, W.H., Sexauer, C.L. et al.
and honest perspective, and communicate (2000). Care of terminally ill children with
frequently and consistently. cancer. Med. Pediatr. Oncol. 34: 268–270.
We would like to acknowledge and thank 5. McConnell, Y., Frager, G., and Levetown, M.
Yoram Unguru, MD, MS, MA, for writing (2004). Decision making in pediatric pallia-
this case and instructor guide. Dr. Unguru is tive care. In: Palliative Care for Infants,
an attending physician in the Division of Children, and Adolescents: A Practical
Pediatric Hematology/Oncology, The Handbook (eds. B. Carter and M. Levetown),
Herman & Walter Samuelson Children’s 69–111. Baltimore: JHU Press.
Hospital at Sinai, and Berman Institute of 6. Informed consent, parental permission, and
assent in pediatric practice (1995).
Bioethics, Johns Hopkins University.
Committee on bioethics, American Academy
of Pediatrics. Pediatrics 95: 314–317.
Suggested reading 7. Spinetta, J.J., Masera, G., Jankovic, M. et al.
(2003). Valid informed consent and partici-
Feudtner, C. (2007). Collaborative communica- pative decision‐making in children with can-
tion in pediatric palliative care: a foundation cer and their parents: a report of the SIOP
for problem‐solving and decision‐making. working committee on psychosocial issues in
Pediatr. Clin. North Am. 207 54: 583–607. pediatric oncology. Med. Pediatr. Oncol. 40:
Freyer, D.R. (2004). Care of the dying adolescent: 244–246.
special considerations. Pediatrics 113: 381–388. 8. Wolfe, J., Hinds, P., and Sourkes, B. (eds.)
Himelstein, B.P., Hilden, J.M., Boldt, A.M., and (2011). Textbook of Interdisciplinary Pediatric
Weissman, D. (2004). Pediatric palliative care. Palliative Care. Philadelphia: Elsevier.
N. Engl. J. Med. 350: 1752–1762. 9. Himelstein, B.P., Hilden, J.M., Morstad Boldt,
Kazak, A.E., Rourke, M.T., Alderfer, M.A. et al. A., and Weissman, D. (2004). Pediatric pallia-
(2007). Evidence‐based assessment, inter- tive care. N. Engl. J. Med. 350: 1752–1762.
vention and psychosocial care in pediatric 10. Hinds, P.S., Drew, D., Oakes, L.L. et al.
oncology: a blueprint for comprehensive ser- (2005). End‐of‐life care preferences of pedi-
vices across treatment. J. Pediatr. Psychol. 32: atric patients with cancer. J. Clin. Oncol. 23:
1099–1110. 9146–9154.
376 Chapter 29

11. Field, M.J. and Behrman, R.E. (eds.) 13. Mack, J.W., Hilden, J.M., Watterson, J. et al.
(2003). When Children Die: Improving (2005). Parent and physician perspectives on
Palliative and End‐of‐Life Care for Children quality of care at the end of life in children
and Their Families. Committee on Palliative with cancer. J. Clin. Oncol. 23: 9155–9161.
and End‐of‐Life Care for Children and 14. Kreicbergs, U., Valdimarsdottir, U., Onelov,
Their Families. Board on Health Sciences E. et al. (2004). Talking about death with
Policy, Institute of Medicine. Washington, children who have severe malignant disease.
DC: National Academies Press. N. Engl. J. Med. 351: 1175–1186.
12. Sourkes, B. (1995). Armfuls of Time: The 15. Unguru, Y., Sill, A., and Kamani, N. (2010).
Psychological Experience of the Child with a The experiences of children enrolled in pedi-
Life‐Threatening Illness. Pittsburgh: atric oncology research: implications for
University of Pittsburgh Press. assent. Pediatrics 125: e876–e883.
30 Chemotherapy Basics

Chemotherapeutic agents have multiple


Arsenic trioxide (ATO)
mechanisms of action and many potential
side effects. In order to be prepared for the
ATO is a naturally occurring substance in
potential complications, we discuss the
the Earth’s crust as well as soil, rock, water,
most common pediatric agents, their
and air, and is also found in shellfish.
mechanisms of action, the types of malig-
nancies for which they are utilized, and
Mechanism of action
finally, the most likely side effects. We dis-
ATO impacts multiple signal induction
cuss how best to monitor for these adverse
pathways and specifically leads to degrada-
events, and how to manage them should
tion of the PML–RARα fusion protein in
they occur. Note that side effects from
acute promyelocytic leukemia (APL), lead-
chemotherapy are numerous and we detail
ing to differentiation of promyelocytes.
some of the more common ones rather
than provide a comprehensive list; further
Utilization
reading may be required in appropriately
APL
diagnosing and managing a patient. We
also include common immunobiological
Side effects, monitoring,
agents which are more targeted and there-
and treatment
fore may have less off‐target effects.
Patients with APL utilizing ATO may
Emetogenic potential of common pediatric
undergo differentiation syndrome in induc-
agents is included in Chapter 25. The
tion as well as elevated white count (i.e.,
majority of agents are dosed depending on
WBC count >10 × 109/l), which should be
the patient’s body surface area (BSA) rather
treated with early initiation of steroids.
than weight, especially for patients older
Differentiation syndrome leads to cardio-
than 1 year of age. Multiple formulas exist
pulmonary distress and may also include
for the calculation of BSA; we prefer the
hypoxemia, fever, rash, pulmonary infil-
Mosteller formula:
trates and effusion, pericardial effusion,
weight gain, peripheral edema, renal failure,
Height cm Weight kg heart failure, and hypotension. Patients
BSA
3600 should be monitored for prolongation of the

Handbook of Pediatric Hematology and Oncology – Children’s Hospital & Research Center Oakland,
Third Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
© 2021 John Wiley & Sons Ltd. Published 2021 by John Wiley & Sons Ltd.
378 Chapter 30

QTc interval. Allergic reaction to ATO may Side effects, monitoring,


occur as well as fatigue, pain, peripheral and treatment
neuropathy, fever, chills, nausea, vomiting, The most immediate side effect from asparagi-
constipation, and diarrhea. nase compounds is an allergic reaction that
can range from local irritation to anaphylaxis.
Premedication with diphenhydramine and
Asparaginase famotidine is becoming more routine as aspar-
aginase levels can be monitored to ensure a
Asparaginase comes in three forms in the therapeutic level and no silent antibody devel-
United States: L‐asparaginase (Elspar®), opment. Patients should be monitored closely
polyethylene glycol (PEG)‐asparaginase
­ for at least 1 hour after injection or infusion.
(Oncaspar®), and Erwinia (Erwinase®). Based on the level of allergic symptoms, treat-
Asparaginase is a naturally occurring ment can range from an antihistamine such as
enzyme produced by many microorganisms. diphenhydramine to treatment of anaphylaxis
In the L‐asparaginase and PEG‐asparaginase with steroids, an antihistamine, an H2 blocker
forms, the enzyme is produced by Escherichia (i.e., famotidine), and epinephrine. A com-
coli, whereas the Erwinia form is produced mon later side effect is coagulopathy second-
by Erwinia chrysanthemi. PEG‐asparaginase ary to decreased synthesis of antithrombin III
is a modified form of L‐asparaginase with a (ATIII), fibrinogen, and other clotting factors.
much longer half‐life resulting from its cova- Currently, routine monitoring of ATIII and
lent binding to PEG, allowing the same ther- fibrinogen and repletion of low levels are not
apeutic effect from fewer doses. recommended without clinical symptoms.
Intramuscular (IM) PEG‐asparaginase has Rare side effects that should be considered
been replaced with intravenous (IV) as the include pancreatitis, thrombosis, and seizures.
standard of care, as there has not been clear
increased risk of antibody formation or ana-
phylaxis. Since Erwinia is produced by a dif- Azacitidine/decitabine
ferent microorganism, it is immunologically
distinct and can be utilized in patients that Azacitidine and decitabine are pyrimidine
have had a hypersensitivity reaction to the E. analogs.
coli forms. Erwinia is only available in an IM
formulation and has not yet been pegylated. Mechanism of action
Azacitidine and decitabine block or decrease
Mechanism of action the methylation of cytosine leading to DNA
L‐asparagine is a nonessential amino acid hypomethylation.
that cannot be synthesized by malignant cells
of lymphoid and myeloid origin. Asparaginase Utilization
depletes L‐asparagine from leukemic cells by Acute myelogenous leukemia
catalyzing the conversion of L‐asparagine to Mixed lineage acute leukemias (MLL;
aspartic acid and ammonia. KMT2A), such as infant acute lymphoblas-
tic leukemia
Utilization
Acute lymphoblastic leukemia (ALL) Side effects, monitoring,
Acute myelogenous leukemia (as rescue and treatment
post‐cytarabine) Azacitidine and decitabine are generally
Non‐Hodgkin lymphoma well‐tolerated, though may lead to a decrease
Chemotherapy Basics 379

in blood counts as well as fatigue, nausea,


Bendamustine
vomiting, constipation, diarrhea, and injec-
tion site reactions with both intramuscular
Bendamustine is a unique alkylator that was
and IV routes of administration.
originally formulated in the former East
German Democratic Republic in the 1960s.

Bleomycin Mechanism of action


Bendamustine is an alkylator with similar
Bleomycin is obtained as a mixture of antibi- properties as cyclophosphamide and other
otics isolated from Streptomyces verticillus. alkylators, though also has a benzimidazole
ring similar to cladribine. This leads to dif-
Mechanism of action ferential mechanism of action compared to
Bleomycin leads to the formation of oxygen‐ other alkylators in terms of its effects on
free radicals that cause single‐strand and DNA repair, as well as activity on both apop-
double‐strand DNA breaks. In addition, totic and nonapoptotic pathways. This also
bleomycin leads to cellular degradation leads to potential benefit in patients that have
­
of cellular RNA. Bleomycin is cell‐cycle‐­ previously received other alkylator therapy.
specific, targeting the G2 and M phases, thus
inhibiting cell growth and division, especially Utilization
in rapidly dividing cells. Hodgkin lymphoma

Side effects, monitoring,


Utilization and treatment
Germ cell tumors Bendamustine has a significantly better side
Hodgkin lymphoma effect profile compared with other alkylators.
Bendamustine can cause an infusional reac-
Side effects, monitoring, tion, and is better tolerated with premedica-
and treatment tions and when given over 60 minutes.
Bleomycin can cause infusional fever and Bendamustine requires less prehydration,
chills followed later by mucositis, as well as though some amount of prehydration and
pruritus and excoriation leading to hyper- posthydration is reasonable. Common side
pigmentation. Raynaud’s phenomenon can effects include decrease in blood counts,
also commonly occur later, especially with fatigue, anorexia, headache, cough, as well as
combination chemotherapy. Patients may constipation, diarrhea, nausea, and vomiting.
occasionally complain of rash, dysgeusia,
and anorexia. Due to the formation of oxy-
gen‐free radicals, patients are at risk for Blinatumomab
dose‐dependent pneumonitis and rarely
pulmonary fibrosis, and therefore should Blinatumomab is a bispecific T‐cell engager
be monitored with pulmonary function which binds CD3 and CD19.
testing (PFTs), especially diffusing capacity
of lung for carbon monoxide (DLCO). An Mechanism of action
anaphylactoid‐type reaction may also Blinatumomab combines a CD3 binding site
rarely occur. with a CD19 binding site and thus induces
380 Chapter 30

direct cytotoxicity of CD19+ cells by engag- antibody. Brentuximab binds the CD30
ing cytotoxic CD3+ T‐cells. CD3 is part of receptor and is internalized via endocytosis
the T‐cell receptor, thus leading to potentia- with subsequent cleavage of the linker
tion of otherwise unstimulated T‐cells to attaching MMAE to the antibody. MMAE is
direct cytotoxicity against all CD19+ cells. released into the tumor microenvironment
leading to direct effects on the targeted
Utilization tumor cells.
Acute lymphoblastic leukemia
Utilization
Side effects, monitoring, Hodgkin lymphoma
and treatment
Due to an extremely short half‐life, blinatu- Side effects, monitoring,
momab is given as a 28‐day continuous infu- and treatment
sion. Due to activation of CD3+ T‐cells, As a targeted monoclonal antibody, brentuxi-
patients may have cytokine release syndrome mab is generally well‐tolerated. Brentuximab
(CRS), especially if with a higher tumor bur- is utilized pretransplant and posttransplant in
den. CRS manifests as fever, hypotension, upfront and relapsed Hodgkin lymphoma.
capillary leak, hypoxemia, and pulmonary/ Potential side effects include lymphopenia,
pericardial effusion. Patients may also pre- peripheral neuropathy, fatigue, nausea, diar-
sent with neurotoxicity, likely due to T‐cell rhea, fever, and rash.
stimulation within the central nervous sys-
tem (CNS). Neurotoxicity should be moni-
tored closely with a daily writing sample and Cisplatin/carboplatin
measuring dysmetria and dysdiadochokine-
sia. Patients may develop altered mental sta- Cisplatin was the first of the platinum‐based
tus, seizure, and encephalopathy; early chemotherapeutic agents. Carboplatin has
initiation of dexamethasone with recogni- less severe side effects, in particular
tion of subtle neurological changes is vital. decreased nephrotoxicity and ototoxicity. It
Other common side effects of blinatumomab is more myelosuppressive, however.
include anemia, nausea, fatigue, diarrhea,
and headache. Due to CD19 cytotoxicity, Mechanism of action
hypogammaglobulinemia can occur. The platinum agents, like the alkylators,
cause interstrand DNA cross‐linking. In
addition, they bind to replicating DNA
Brentuximab vedotin causing single‐strand breaks.

Brentuximab is a monoclonal antibody to Utilization


CD30. Brain tumors
Germ cell tumors
Mechanism of action Hepatoblastoma
Brentuximab consists of a chimeric anti- Hodgkin lymphoma
body specific to human CD30, the microtu- Neuroblastoma
bule disrupting agent MMAE (monomethyl Osteogenic sarcoma
auristatin E [vedotin]), and a protease‐ Soft tissue sarcomas
cleavable linker that attaches MMAE to the Wilms tumor
Chemotherapy Basics 381

Side effects, monitoring, urements of the glomerular filtration rate


and treatment and likely platinum dose reduction.
Infusional nausea and vomiting are common,
as platinum agents have an extremely high
emetogenic potential. This is followed later by
cis‐retinoic acid/all‐trans
myelosuppression. Electrolyte abnormalities
retinoic acid
including hypokalemia, hypomagnesemia,
hypocalcemia, and hyponatremia are com-
cis‐Retinoic acid (isotretinoin) and all‐trans
mon and the patient may develop Fanconi
retinoic acid (ATRA) are naturally occur-
syndrome (diminished reabsorption of solutes
ring analogs of vitamin A.
by the proximal tubule resulting in hypophos-
phatemia, metabolic acidosis, and secondary
Mechanism of action
hypokalemia). Nephrotoxicity and ototoxicity
Retinoid mechanism of action is not known,
(high‐frequency sensorineural hearing loss)
though is thought to contribute to matura-
occur occasionally with carboplatin, but are
tion/differentiation of tumor cells.
more common and severe with cisplatin.
Ototoxicity must be monitored with
Utilization
serial audiograms during therapy. Due to
APL
the risk of electrolyte abnormalities, chem-
High‐risk neuroblastoma
istry panels including magnesium should
be followed daily while receiving platinum‐
Side effects, monitoring,
based therapy. In addition, due to the risk of
and treatment
nephrotoxicity, the patient’s intake and out-
Retinoids commonly lead to dry skin/mucosa
put should be followed closely and be well‐
and may also lead to headache and the devel-
matched. If it appears the patient is
opment of pseudotumor cerebri. Consideration
developing a negative fluid balance, addi-
should be made for formal ophthalmologic
tional fluids should be provided. If the
exam in patients with moderate to severe head-
patient develops a positive fluid balance,
ache to rule out papilledema. Patients with
mannitol should be used to increase urine
APL utilizing ATRA may undergo differentia-
output. Renal function should be moni-
tion syndrome in induction as well as elevated
tored closely, as a reduction in the glomeru-
white count (i.e., WBC count >10 × 109/l),
lar filtration rate will necessitate a dose
which should be treated with early initiation of
reduction. In general, for pediatric patients
steroids. Differentiation syndrome leads to car-
with no previous history of nephrotoxicity,
diopulmonary distress and may also include
serum creatinine can serve as a measure of
hypoxemia, fever, rash, pulmonary infiltrates
renal function utilizing the Cockcroft–
and effusion, pericardial effusion, weight gain,
Gault equation:
peripheral edema, renal failure, heart failure,
and hypotension. Photosensitivity, arthralgias,
Estimated creatinine clearance elevated liver enzymes, triglycerides, and cho-
140 Age years Mass kg 0.85 if female lesterol, as well as hypercalcemia are additional
72 serum creatinine mg / dl potential side effects with retinoids. Retinoids
are thought to be teratogenic and cannot be uti-
lized in women of child‐bearing potential
Decrease in the estimated creatinine unwilling or unable to utilize standard contra-
clearance should lead to more formal meas- ceptive methods.
382 Chapter 30

Utilization
Cladribine/clofarabine
Acute lymphoblastic leukemia
Acute myelogenous leukemia
Cladribine and clofarabine are purine nucle-
Brain tumors
oside antimetabolites/analogs.
Ewing sarcoma
Germ cell tumors
Mechanism of action Hodgkin and non‐Hodgkin lymphoma
As purine analogs, cladribine and next‐gen- Neuroblastoma
eration clofarabine are resistant to deamina- Osteogenic sarcoma
tion by adenosine deaminase, leading to Soft tissue sarcomas
direct inhibition of DNA synthesis. Wilms tumor
Utilization Side effects, monitoring,
Relapsed/refractory acute lymphoblastic and treatment
leukemia Cyclophosphamide and ifosfamide com-
Relapsed/refractory acute myelogenous monly cause nausea, vomiting, and anorexia
leukemia with drug infusion. Myelosuppression,
Recurrent/refractory histiocytosis (Langer­hans immunosuppression, and alopecia are com-
cell histiocytosis, Rosai–Dorfman dis- mon later adverse effects. Sterility is dose‐
ease, juvenile xanthogranuloma) dependent (greatest with cumulative doses
of cyclophosphamide >7.5 g/m2) and occurs
Side effects, monitoring, more commonly in pubertal males (though
and treatment prepubertal males and females are also at
Cladribine and clofarabine are likely to cause risk). Due to the risk of nephrotoxicity, par-
cytopenias and therefore potential resultant ticularly with ifosfamide, patients should be
infection. Though otherwise generally well‐ well‐hydrated prior to and after infusion.
tolerated, these agents can lead to diarrhea, Though the syndrome of inappropriate anti-
nausea, vomiting, anorexia, fatigue, fever, diuretic hormone (SIADH) is rare, patients
hypotension or hypertension, arrhythmias, should be monitored for appropriate urine
liver damage, as well as headache. output by following their intake and output.
Electrolytes are also routinely followed for
hyponatremia. If there are signs of fluid
Cyclophosphamide/ifosfamide retention, the patient should initially be
given increased hydration followed by diu-
Cyclophosphamide (Cytoxan) and ifosfamide resis with furosemide or mannitol if hydra-
are alkylating agents and structural analogs. tion is ineffective.
They are related to nitrogen mustard. Excretion of acrolein into the urine can
cause hemorrhagic cystitis. The addition of
Mechanism of action MESNA for bladder protection when the
Cyclophosphamide and ifosfamide require cyclophosphamide dose exceeds 1.0 g/m2/
conversion by the hepatic P450 system to day has significantly decreased the fre-
their active form that ultimately leads to the quency of this adverse event. MESNA
intracellular release of two compounds, specifically binds to acrolein and other
­
acrolein and phosphoramide mustard.
­ toxic metabolites in the urine to detoxify
Phosphoramide mustard causes interstrand them and protect the bladder wall. The
DNA cross‐linking. urine should be monitored for the presence
Chemotherapy Basics 383

of occult blood during and after infusion pain. High‐dose IV cytarabine requires
and, if positive, should be examined micro- prophylaxis with dexamethasone eye drops
scopically. If red blood cells are noted on to prevent conjunctivitis. A flu‐like syn-
microscopy, hydration should be increased. drome with fever, chills, and rash occurs
Of note, MESNA can cause a false positive occasionally with cytarabine; infection must
test for ketones on urine dipstick. Finally, still be ruled out with bacterial cultures.
metabolism of ifosfamide leads to forma- Streptococcus viridans sepsis and acute res-
tion of chloroacetaldehyde, a byproduct piratory distress syndrome are possible after
thought responsible for nephrotoxicity high‐dose cytarabine, therefore the addition
as well as the neurotoxicity seen occasion- of vancomycin with fever or clinical deterio-
ally with ifosfamide (somnolence, depres- ration should be strongly considered.
sive psychosis, and confusion). Thiamine Intrathecal (IT) cytarabine similarly can
prophylaxis may have benefit in a patient cause fever, nausea, and vomiting in addi-
with prior neurotoxicity. tion to headache. More serious and immedi-
ate side effects including arachnoiditis,
somnolence, meningismus, convulsions,
Cytarabine (Ara‐C) and paresis are rare but should be consid-
ered as clinically indicated.
Cytarabine, also known as cytosine arabino-
side or Ara‐C (arabinofuranosyl cytidine), is
utilized in the treatment of hematologic Dactinomycin (Actinomycin‐D)
malignancies.
Dactinomycin is an antibiotic compound
Mechanism of action isolated from Streptomyces parvulus, similar
Cytarabine is an antimetabolite and pyrimi- to the anthracycline class.
dine analog that inhibits DNA polymerase.
In addition, it is cell‐cycle‐specific, killing Mechanism of action
cells during synthesis (S phase); it therefore Dactinomycin intercalates with DNA, inhib-
specifically targets rapidly dividing cells. iting RNA and DNA synthesis. In addition,
dactinomycin interacts with topoisomerase,
Utilization which is required for DNA replication, and
Acute lymphoblastic leukemia leads to single‐strand DNA breaks.
Acute myelogenous leukemia
Hodgkin and non‐Hodgkin lymphoma Utilization
Soft tissue sarcomas
Side effects, monitoring, Wilms tumor
and treatment
Cytarabine commonly causes nausea, vom- Side effects, monitoring,
iting, and anorexia immediately after infu- and treatment
sion. Later, myelosuppression, stomatitis, Infusional nausea and vomiting are com-
and alopecia are common. Although not mon, followed later by alopecia and myelo-
common, the patient should be monitored suppression. Anorexia, fatigue, diarrhea,
for Ara‐C syndrome that includes fever, and mucositis also occur occasionally.
myalgias, bone pain, malaise, conjunctivitis, Radiation recall can occur in patients who
maculopapular rash, and occasionally chest previously received radiation therapy.
384 Chapter 30

Daunorubicin/doxorubicin/ Side effects, monitoring,


idarubicin/mitoxantrone and treatment
Anthracyclines commonly cause nausea and
Daunorubicin and doxorubicin are both vomiting with treatment. Due to the color of
anthracycline antibiotics isolated from the infusion, the patient should be warned
Streptomyces species (Streptomyces coerule- that urine, saliva, tears, and sweat may all have
orubidus and Streptomyces peucetius, a pink or red coloring (blue with mitox-
respectively). Idarubicin is an analog of antrone). After treatment, myelosuppression,
daunorubicin used less commonly in pedi- alopecia, and mucositis commonly occur.
atric oncology. Mitoxantrone belongs to a Due to the production of free radicals during
separate structural class of antineoplastic electron reduction of anthracyclines, patients
agents called anthracenediones and was are at risk for dose‐dependent cardiotoxicity
produced synthetically. occurring as a late finding. Echocardiographic
monitoring is required during and after treat-
Mechanism of action ment at regular intervals based on the total
Anthracyclines as a group are cytotoxic to anthracycline dose received as well as con-
malignant cells due to nucleotide base inter- comitant radiation therapy to the chest.
calation and cell membrane lipid‐binding Cardioprotection with continuous versus
activity. Nucleotide intercalation inhibits rapid infusion remains controversial and is
replication as well as DNA and RNA poly- not yet routinely recommended in pediatric
merases. The anthracyclines also interact patients. Dexrazoxane, an iron chelator and
with topoisomerase II, which is vital for free radical scavenger, has shown benefit as a
DNA replication. Cell membrane binding cardioprotectant when patients receive high
affects a variety of cellular functions. In addi- cumulative doses of anthracyclines (i.e.,
tion, electron reduction of the anthracyclines ≥300 mg/m2 doxorubicin equivalents [see in
produces free radicals leading to DNA dam- the following text]) and is recommended to
age and lipid peroxidation. Daunorubicin commence in adults when they have received
differs from doxorubicin structurally, as the a cumulative dose of 300 mg/m2. Young chil-
side chain terminates in a methyl group dren (i.e., <4 years of age) anticipated to
rather than an alcohol. Idarubicin lacks a receive a cumulative dose >300 mg/m2 may
methoxy group which increases its lipophi- benefit with earlier addition of dexrazoxane.
licity as compared with other anthracyclines. Anthracyclines provide different cumulative
Mitoxantrone specifically has been shown to toxicity, and dose conversion based on doxo-
have cytotoxic effects on both proliferating rubicin isotoxic dose equivalents must be per-
and nonproliferating cells. formed as provided in the following text:
Doxorubicin: multiply total dose × 1
Daunorubicin: multiply total dose × 0.5
Utilization
Epirubicin: multiply total dose × 0.67
Acute lymphoblastic leukemia
Idarubicin: multiply total dose × 5
Acute myelogenous leukemia
Mitoxantrone: multiply total dose × 4
Hepatoblastoma
(though data are increasing that the con-
Hodgkin and non‐Hodgkin lymphoma
version factor may be 10)
Neuroblastoma
Osteogenic sarcoma Radiation recall is also a potential rare com-
Soft tissue sarcomas plication of anthracyclines when given after
Wilms tumor radiation therapy.
Chemotherapy Basics 385

Mechanism of action
Dinutuximab
Etoposide binds to topoisomerase II, which
is vital for DNA replication, leading to DNA
Dinutuximab is a monoclonal chimeric
strand breakage. Etoposide is cell‐cycle‐spe-
14.18 antibody to the glycolipid disialogan-
cific and appears to act mainly on the G2
glioside (GD2).
and S (synthesis) phases, thus targeting
rapidly dividing cells.
Mechanism of action
Dinutuximab binds to cell surface GD2 on
neuroblastoma cells leading to cell lysis Utilization
through antibody‐dependent cell‐mediated Acute myelogenous leukemia
cytotoxicity and complement‐dependent cyto- Brain tumors
toxicity. Dinutuximab has been found to be Ewing sarcoma
effective with immune stimulation with either Germ cell tumors
IL‐2 or GM‐CSF (granulocyte‐macrophage Hodgkin and non‐Hodgkin lymphoma
colony‐stimulating factor), and is also given Hemophagocytic lymphohistiocytosis
with the cell differentiator cis‐retinoic acid. Neuroblastoma
Osteogenic sarcoma
Utilization Soft tissue sarcomas
High‐risk neuroblastoma Wilms tumor

Side effects, monitoring, Side effects, monitoring,


and treatment and treatment
Dinutuximab is given over 10–20 hours daily Infusional nausea and vomiting are com-
for 4 days in combination with GM‐CSF. mon followed by myelosuppression and
Dinutuximab can lead to life‐threatening alopecia. Although rare, the patient should
infusion reactions and severe neurotoxicity be monitored closely for hypotension and
and pain. Dinutuximab can lead to CRS with anaphylaxis during the infusion. In addi-
fever, hypotension, capillary leak, edema/ tion to fluid support, the infusional rate
ascites, hypoxemia, and pulmonary/pericar- can be slowed if the patient develops
dial effusion. Patients should have weight hypotension.
monitoring twice daily in addition to contin-
uous low‐dose opioid support. Urinary
retention, hypertension, bleeding/clotting,
­ Gemtuzumab
anorexia, diarrhea, nausea/vomiting, periph-
eral neuropathy, and anemia are additional Gemtuzumab ozogamicin is a monoclonal
potential side effects. Institutions utilizing antibody to CD33.
dinutuximab generally will have a standard
monitoring and treatment protocol that Mechanism of action
should be reviewed when using dinutuximab. Gemtuzumab is a monoclonal antibody
which binds to the CD33 antigen on hemat-
opoietic cells which leads to internalization.
Etoposide Gemtuzumab is linked to a calicheamicin
cytotoxic agent, which exerts cell cytotoxic
Etoposide is derived from podophyllotoxin, effects through DNA double‐strand breaks
a toxin found in the American mayapple. once internalized.
386 Chapter 30

Utilization Common immediate effects include fluid


Acute myelogenous leukemia retention, nausea, and diarrhea. Later com-
mon effects include fatigue, muscle cramps,
Side effects, monitoring, rash, arthralgias, and myelosuppression.
and treatment TKIs may impact growth. Dasatinib can
Gemtuzumab can lead to cytopenias in cause heart dysfunction and echocardio-
addition to nausea and vomiting, headache, gram should be followed on a routine basis.
fever and chills, shortness of breath, ano-
rexia, fatigue, and hypokalemia. Infusion
reactions can occur and premedications Inotuzumab ozogamicin
should be given. Sinusoidal obstructive syn-
drome (veno‐occlusive disease) risk can be Inotuzumab ozogamicin is a monoclonal
increased posttransplant due to prior utili- antibody to CD22.
zation of gemtuzumab.
Mechanism of action
Inotuzumab is a monoclonal antibody which
Imatinib (gleevec®)/dasatinib/ binds to the CD22 receptor on B‐cells, which
ponatinib leads to internalization. Inotuzumab is linked
to a calicheamicin cytotoxic agent, which
Imatinib was the first successful targeted
exerts cell cytotoxic effects through DNA
drug therapy for oncology patients.
double‐strand breaks once internalized.

Mechanism of action Utilization


Imatinib is a selective inhibitor of the tyros- Relapsed/refractory acute lymphoblastic
ine kinase activity of the BCR–ABL fusion leukemia
protein, a product of the Philadelphia chro-
mosome (reciprocal translocation of chro-
Side effects, monitoring,
mosomes 9 and 22) seen mainly in chronic
and treatment
myeloid leukemia and rarely with acute
Inotuzumab can lead to cytopenias in addi-
lymphoblastic leukemia. Dasatinib is a sec-
tion to nausea and vomiting, headache,
ond‐generation inhibitor of the BCR–ABL
fever and chills, fatigue, transaminitis,
fusion protein, being more specific to the
mucositis, diarrhea/constipation, and
protein binding site. Ponatinib is a third‐
hyperbilirubinemia. Infusion reactions can
generation inhibitor which is effective in
occur and premedications should be given.
patients with the T315I mutation.
Sinusoidal obstructive syndrome (veno‐
occlusive disease) risk can be increased
Utilization posttransplant due to prior utilization of
Acute lymphoblastic leukemia (Philadelphia inotuzumab.
chromosome‐positive)
Chronic myelogenous leukemia
Irinotecan
Side effects, monitoring,
and treatment Irinotecan is a semisynthetic analog isolated
Imatinib and other tyrosine kinase inhibitors from the plant alkaloid Camptotheca
(TKIs) have multiple potential side effects. acuminata.
Chemotherapy Basics 387

Mechanism of action Utilization


Irinotecan in its active form is a potent Acute lymphoblastic leukemia
inhibitor of topoisomerase I which is vital Non‐Hodgkin lymphoma
for DNA replication. Inhibition of topoi-
somerase inhibits replication and also leads Side effects, monitoring,
to DNA damage. and treatment
Mercaptopurine is generally well‐tolerated,
Utilization although myelosuppression occurs com-
Brain tumors monly. Mercaptopurine is usually adminis-
Ewing sarcoma tered at night as patients may occasionally
Hepatoblastoma complain of anorexia, nausea, and vomiting.
Neuroblastoma Morning hypoglycemia may also rarely
Soft tissue sarcomas occur. Separation from food or milk is no
longer required; giving the medicine at the
Side effects, monitoring, same time daily is highly recommended for
and treatment efficacy. Diarrhea and an erythematous rash
Patients receiving irinotecan can suffer from (often perioral) may also occasionally occur.
cholinergic symptoms including intestinal Monitoring of mercaptopurine metabolites
hyperperistalsis that can lead to abdominal (thioguanine nucleotides) can be utilized to
cramping and early diarrhea. Patients should objectively measure questionable compli-
be monitored closely for early diarrhea; if it ance and ensure appropriate dosing for
occurs, loperamide should be given prior to patients on either subtherapeutic or supra-
an anticholinergic (e.g., atropine). Other cho- therapeutic dosing.
linergic symptoms that may occur with drug
administration include rhinitis, increased
salivation, miosis, lacrimation, diaphoresis, Methotrexate
and flushing. Common later side effects
include diarrhea, alopecia, transaminitis, Methotrexate was the first successful chem-
neutropenia, mucositis, and hyperbilirubine- otherapeutic agent utilized in children after
mia. Diarrhea is due to the active metabolite the observation that folic acid worsened leu-
of irinotecan SN‐38 which can be mitigated kemia and dietary deficiency of folic acid
with the use of a prophylactic antibiotic such could improve leukemia symptoms.
as cefixime or cefpodoxime.
Mechanism of action
Methotrexate inhibits folic acid by prevent-
Mercaptopurine (6‐MP) ing the reduction of folic acid by the enzyme
dihydrofolate reductase. This inhibition
Mercaptopurine is a purine analog. subsequently limits the synthesis of purines
and DNA. Some of the methotrexate metab-
Mechanism of action olites also lead to DNA damage.
Mercaptopurine is converted into several
active metabolites that inhibit RNA and DNA Utilization
synthesis. As a purine analog it can also inter- Acute lymphoblastic leukemia
fere with purine biosynthesis. Mercaptopurine Acute myelogenous leukemia
is converted to nucleotide metabolites, some Brain tumors
of which can lead to DNA toxicity. Non‐Hodgkin lymphoma
388 Chapter 30

Osteogenic sarcoma after delayed clearance and includes severe


mucositis and myelosuppression. Patients
Side effects, monitoring, may benefit from glutamine prophylaxis for
and treatment the prevention of mucositis with subsequent
Because of significant enterohepatic circula- methotrexate doses. IT methotrexate often
tion of methotrexate, transaminitis is com- causes nausea and headache. Arachnoiditis
mon. Nausea, vomiting, and anorexia may occasionally occurs and the patient should
also occur; therefore, oral doses are generally be monitored for symptoms including fever,
given at bedtime. Many side effects of metho- vomiting, and meningismus. Long‐term
trexate are due to delayed clearance after cognitive dysfunction and learning disabili-
high‐dose IV treatment. Aggressive hydration ties occasionally occur. Leukoencephalopathy
and alkalinization of fluids can improve renal and progressive cognitive deterioration
clearance. Drugs including trimethoprim/sul- rarely occur, especially in adolescents and
famethoxazole, penicillin, nonsteroidal anti‐ adults, with high‐dose IV methotrexate and
inflammatories (NSAIDs), and proton pump accentuated by cranial radiation therapy.
­
inhibitors (PPIs) can competitively inhibit Management of patients with neurotoxicity
renal clearance and must be held during high‐ including ­seizures, confusion, ataxia, cranial
dose therapy. Leucovorin (folinic acid) is used nerve palsies, speech disorders, and parapa-
to decrease many of the toxic effects of folic resis is controversial. Practitioners may rec-
acid antagonists such as methotrexate. ommend leucovorin rescue after future IT
Leucovorin can participate in metabolic reac- doses or replacement with IT cytarabine,
tions requiring folic acid without the necessity with or without hydrocortisone. Conclusive
of reduction by dihydrofolate reductase which evidence is lacking on the use of dex-
is inhibited by methotrexate. This mechanism tromethorphan as a neuroprotectant but
of action can also counteract the therapeutic may also be trialed. The majority of patients
effect of methotrexate; therefore, leucovorin will tolerate rechallenge of IT methotrexate
should not be started until 18–24 hours after without issue.
the methotrexate infusion has been com-
pleted. For this reason, patients should be
advised to not take folic acid supplements Nelarabine
during methotrexate therapy, as this may sim-
ilarly reduce efficacy. Nelarabine is a prodrug of Ara‐G.
Urine pH should be monitored closely
and kept above 7.5 during high‐dose therapy Mechanism of action
to facilitate renal excretion of methotrexate. Nelarabine is a substrate for adenosine
The patient’s intake and output should also deaminase which converts the prodrug to
be followed. Methotrexate levels are usually Ara‐G. Ara‐G is internalized into T‐lymph-
drawn starting 24 hours after the infusion is oblasts and subsequently phosphorylated
started. Leukemia protocols provide a meth- into the active nucleotide Ara‐GTP which
otrexate nomogram that allows the practi- competes in leukemic blasts cells for incor-
tioner to determine if methotrexate levels are poration into cellular DNA, inhibiting fur-
declining appropriately or are in a toxic ther DNA synthesis.
range requiring an increase in hydration and
leucovorin frequency or dose. Rarely, renal Utilization
failure occurs and can be managed with car- T‐cell acute lymphoblastic leukemia/
boxypeptidase G2. Toxicity is typically worse lymphoma
Chemotherapy Basics 389

Side effects, monitoring,


Rituximab
and treatment
Neurotoxicity is the most identifiable
Rituximab is a monoclonal antibody to
unique toxicity of nelarabine and can be
CD20.
either central or peripheral. Nelarabine is
otherwise generally well‐tolerated, though Mechanism of action
can lead to cytopenias, nausea, and fatigue. Rituximab has multiple proposed mecha-
nisms of action including direct signaling of
apoptosis, complement‐dependent cellular
Nivolumab/pembrolizumab toxicity, and antibody‐dependent cell‐medi-
ated toxicity.
Nivolumab and pembrolizumab are mono-
clonal antibodies to the receptor of pro- Utilization
grammed cell death‐1 (PD‐1) and therefore Non‐Hodgkin lymphoma
called immune checkpoint inhibitors.
Side effects, monitoring,
Mechanism of action and treatment
Nivolumab and pembrolizumab bind to Rituximab can cause an infusion reaction
the PD‐1 receptor which blocks interac- and should be given with premedications
tion with its ligands PD‐L1 and PD‐L2. per a standard protocol. Other common
This binding limits T‐cell exhaustion on side effects include nausea, fever, chills, and
activated T‐cells and leads to T‐cell persis- fatigue. Due to inhibition of CD20, infection
tence of these cells, increasing tumor cell and hypogammaglobulinemia are possible.
killing. Progressive multifocal leukoencephalopathy
(PML) has been reported in adult patients
Utilization but is unlikely in younger patients.
Hodgkin lymphoma
Renal cell carcinoma

Side effects, monitoring, Steroids


and treatment
Dexamethasone and prednisone are the
Immune checkpoint inhibitors are gener-
steroids used most commonly as chemo-
ally well‐tolerated. The most common
therapeutic agents.
adverse events include fatigue, anorexia,
diarrhea, nausea, cough, dyspnea, consti-
pation, vomiting, rash, fever, and headache. Mechanism of action
Immune‐related c­ omplications with these Steroids have a variety of actions on the
medications can occur, most notably pseu- body. Although not completely under-
doprogression after initiation of the medi- stood, steroids are thought to destroy
cation due to local inflammatory reaction lymphoblasts by binding to the cortisol
as well as pneumonitis due to this response receptor found on lymphoid cells and spe-
in the lungs. Pneumonitis may require ster- cifically in large number on lymphoblasts.
oids. Other potential immune complica- Steroids are immunosuppressive and target
tions include vitiligo, colitis, hepatitis, T‐lymphocytes, monocytes, and eosino-
hypophysitis (pituitary inflammation), and phils. Steroids may also function by halting
thyroiditis. DNA synthesis. Dexamethasone has better
390 Chapter 30

CNS penetration than prednisone and has Side effects, monitoring,


benefit in preventing CNS relapse in ALL. and treatment
Temozolomide commonly causes anorexia,
Utilization nausea, vomiting, and constipation followed
Acute lymphoblastic leukemia by myelosuppression. Occasionally, temozo-
Hemophagocytic lymphohistiocytosis lomide may cause abdominal pain, diarrhea,
Hodgkin and non‐Hodgkin lymphoma headache, and mucositis. Continuous low‐
Langerhans cell histiocytosis dose (metronomic) therapy may be effective
in some patients with a decreased side‐effect
Side effects, monitoring, profile.
and treatment
Steroid treatment results in a multitude of
side effects. Common ones include hyper- Thioguanine (6‐TG)
phagia, insomnia, personality changes, adre-
nal suppression, acne, immunosuppression, Like mercaptopurine, thioguanine is an oral
and Cushing’s syndrome. Occasional side antimetabolite and purine analog.
effects include gastritis, hyperglycemia, poor
wound healing, facial erythema, striae, thin- Mechanism of action
ning of the skin, proximal muscle weakness, Metabolites of thioguanine interfere with
osteopenia, and cataracts. Avascular necrosis purine synthesis and DNA replication. The
of various joints, most commonly hips, knees, intercalation of nucleotide metabolites into
and ankles, occurs rarely. It is more common DNA also leads to DNA strand breaks.
in adolescents, and females are at higher risk.
It appears to occur more commonly with Utilization
dexamethasone than with prednisone. Acute lymphoblastic leukemia
Hypertension occurs rarely; blood pressure Acute myelogenous leukemia
should be monitored closely in addition to Non‐Hodgkin lymphoma
blood glucose. Patients should be on an H2
blocker (i.e., famotidine) while receiving Side effects, monitoring,
daily steroids to prevent gastritis and peptic and treatment
ulcer disease. Potassium chloride may be tri- Thioguanine commonly leads to myelo-
aled to decrease mood effects from steroids. suppression and occasionally causes
fatigue, nausea, vomiting, diarrhea, and
anorexia. Although rare, the patient should
Temozolomide be monitored for transaminitis as well as
signs of veno‐occlusive disease (sinusoidal
Temozolomide (Temodar®) is an oral alkylat- obstructive syndrome) and hepatic
ing agent. fibrosis.
Mechanism of action
As with other alkylators, temozolomide
leads to DNA interstrand cross‐linking. Tisagenlecleucel (Kymriah)

Utilization Tisagenlecleucel is a chimeric antigen recep-


Brain tumors tor re‐engineered T‐cell (CAR‐T) targeting
Ewing sarcoma CD19+ cells.
Chemotherapy Basics 391

Mechanism of action
Topotecan
Patients with multiply relapsed or refrac-
tory CD19+ acute lymphoblastic leukemia
Like irinotecan, topotecan is a semisynthetic
may be eligible for tisagenlecleucel. Patients
analog isolated from the plant alkaloid C.
have apheresis of their mononuclear cells
acuminata.
which are sorted such that cytotoxic and
helper T‐cells can be re‐engineered to spe-
Mechanism of action
cifically target CD19+ cells. These re‐engi-
Topotecan is a potent inhibitor of topoi-
neered CAR‐T cells are subsequently
somerase I that is vital for DNA replication.
reinfused to the patient after lymphodeple-
Inhibition of topoisomerase inhibits replica-
tion. Generally, there is rapid elimination
tion and also leads to DNA damage.
of the CD19+ cells by the CAR‐T cells.
There can be loss of the CAR‐T cells due to
Utilization
an immune response or secondary to anti-
Brain tumors
gen escape, which is usually due to a CD19–
Neuroblastoma
CD22+ clone. A surrogate marker of the
loss of persistence of CAR‐T cells is the loss
of B‐cell aplasia. Side effects, monitoring,
and treatment
Utilization Topotecan can commonly cause nausea,
Relapsed acute lymphoblastic leukemia vomiting, diarrhea or constipation, fever,
pain and later myelosuppression, fatigue,
Side effects, monitoring, and alopecia. The patient should also be
and treatment monitored for the occasional findings of
CRS is common after CAR‐T cell infusion headache, rash, hypotension, transaminitis,
and presents as unremitting fever with and mucositis.
associated symptoms including hypoten-
sion, capillary leak, ascites, respiratory dis-
tress, hypoxemia, and pleural/pericardial Vincristine/vinblastine/
effusions. Patients with CRS are at increased vinorelbine
risk of neurotoxicity (ICANS; immune
effector cell associated neurotoxicity syn- Vincristine and vinblastine are alkaloids iso-
drome) which can manifest as severe lated from the Madagascar periwinkle plant
headache, altered mental status, seizure, (Vinca rosea, now Catharanthus roseus) and
encephalopathy, and coma/death. therefore often referred to as vinca alkaloids.
Treatment of CRS includes early diagnosis Vincristine and vinblastine are structurally
based on clinical and laboratory criteria identical except for a single substitution (a
and rapid initiation of tocilizumab (anti‐ formyl group in vincristine is replaced by a
IL6) and potentially dexamethasone. methyl group in vinblastine), which leads
Neurotoxicity is managed with the use of an to significant differences in their cytotoxic
anti‐epileptic and supportive care. effects. Vinorelbine was subsequently pro-
Institutions utilizing tisagenlecleucel will duced semi‐synthetically, though can be
have detailed protocols to monitor and produced from byproducts of C. roseus and
manage potential side effects of CAR‐T is therefore considered to be in the family of
therapy. vinca alkaloids.
392 Chapter 30

Mechanism of action vincristine more commonly causes neuro-


The vinca alkaloids bind to microtubules toxicity including constipation and loss of
especially in the mitotic spindle leading to deep tendon reflexes. All can lead to alope-
metaphase arrest, thus targeting rapidly cia. Jaw pain, peripheral paresthesias, wrist
dividing cells. They have other disruptive cel- and foot drop, and abnormal gait occa-
lular functions that may or may not be related sionally occur, especially with vincristine.
to their effects on tubulin and microtubules. Ptosis, vocal cord dysfunction, and dam-
age to the eighth cranial nerve (clinically
Utilization with dizziness, nystagmus, vertigo, and
Acute lymphoblastic leukemia hearing loss) are rare findings. Vinca alka-
Brain tumors loids are vesicants; therefore, extravasa-
Ewing sarcoma tion, if it occurs, can lead to local ulceration
Hepatoblastoma (see Chapter 32 for management of
Hodgkin and non‐Hodgkin lymphoma extravasation).
Langerhans cell histiocytosis
Neuroblastoma
Suggested reading
Soft tissue sarcomas
Wilms tumor Adamson, P.C., Blaney, S.M., Bagatell, R. et al.
(2016). General Principles of Chemotherapy, 7e
Side effects, monitoring, (eds. Pizzo and Poplack), 239–315. Wolters
and treatment Kluwer/Lippincott.
Vinblastine and vinorelbine more com-
monly cause myelosuppression, whereas
31 Guide to Procedures

Procedures yield critical diagnostic and documentation in the medical record with
treatment information for patients with the signed informed consent.
suspected or known oncologic or hemato- All procedures should be performed or
logic diseases. Traditionally, tumor biopsies supervised by practitioners with technical
(depending on tumor site) are performed expertise. For patients undergoing anesthe-
on patients while under general anesthesia sia, a skilled caregiver (i.e., anesthesiologist,
by pediatric surgeons, pediatric orthopedic nurse anesthetist, or critical care physician)
oncologists, and pediatric interventional should administer the sedation and monitor
radiologists. Common procedures per- the patient. In addition to the practitioners
formed by the pediatric hematologist/ performing the procedure and sedation,
oncologist include bone marrow aspiration another skilled caregiver (i.e., nurse or physi-
(BMA) and biopsy, lumbar puncture (LP) cian) should be present to assist with posi-
to obtain cerebral spinal fluid (CSF) and tioning, sterile transfer of chemotherapy per
administer intrathecal (IT) chemotherapy, institutional policy, and general patient care
as well as the administration of chemother- and monitoring. Many centers perform all
apy via a peripheral vein or Ommaya reser- LPs and BMAs under deep sedation or anes-
voir (another form of directed central thesia; however, some patients prefer to have
nervous system [CNS] therapy). these procedures performed awake, or with
Basic principles for performing proce- mild sedation.
dures include: (i) ensuring the procedure Prior to initiation of the procedure, a
is indicated for diagnosis, assessment of time‐out should be done to ensure proper
response to therapy, or to determine pos- identification of the patient (hospital wrist
sible relapse; (ii) ensuring the proper band, medical record, and labels on medi-
medication is being administered at the cations) and the planned procedure. All
proper time; (iii) providing a safe and chemotherapeutic medications should be
sterile environment; and (iv) obtaining brought into the room and checked by at
informed consent with proper documen- least two skilled practitioners. If multiple
tation. The patient’s medical record medications are to be administered, care
should be carefully reviewed prior to the should be taken to avoid an error in
procedure and the purpose and nature administration by bringing in one drug at
of the procedure be reviewed with the a time. An exemption to this is if the
patient and family. This discussion should patient is receiving multiple IT medica-
include anticipated risks and benefits with tions and the pharmacy dispenses these

Handbook of Pediatric Hematology and Oncology – Children’s Hospital & Research Center Oakland,
Third Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
© 2021 John Wiley & Sons Ltd. Published 2021 by John Wiley & Sons Ltd.
394 Chapter 31

separately. Standardized procedures should 2. Physical examination should be per-


be followed to minimize error. Procedures formed prior to the procedure with atten-
may be performed in an outpatient or inpa- tion on a focused neurologic evaluation
tient setting by individuals who have been including assessment for papilledema or
properly trained and supervised. other evidence of increased intracranial
pressure such as high blood pressure,
widened pulse pressure, or altered level of
Lumbar puncture/intrathecal consciousness. Evaluate vitals and assess for
chemotherapy any evidence of infection or metabolic
abnormalities to ensure safety for anesthesia.
Pediatric hematology/oncology physicians Evaluate site of procedure to assure no
and their trained staff perform LPs to assess localized infection or skin breakdown.
for involvement of the CSF by malignancy 3. Laboratory studies must be checked to
as well as to administer IT chemotherapy confirm that parameters are met for admin-
(drug delivery directly into the subarach- istration of chemotherapy per protocol
noid space of the spinal column). Drugs are (e.g., platelet count, absolute neutrophil
administered in this manner to bypass the count, and hemoglobin). Patients with
blood–brain barrier. The three agents typi- ­moderate thrombocytopenia (platelet count
cally given IT are: methotrexate, cytarabine <50 × 109/l) may need a platelet transfusion,
(Ara‐C), and hydrocortisone. although this varies with the skill of the
practitioner and institutional protocol. The
Indications literature recommends a platelet count
Patients with suspected leukemia or lym- ≥100 × 109/l for a diagnostic LP in a patient
phoma (diagnosis, staging), history of leuke- with suspected or newly diagnosed leuke-
mia or CNS lymphoma (treatment or new mia to decrease the possibility of a traumatic
signs or symptoms suggestive of relapse), tap with potential increased risk of CNS
CNS metastases, and therapy complications relapse. A highly skilled practitioner should
related to the CNS, specifically infection be performing all diagnostic LPs.
or neurotoxicity (suspected meningitis,
encephalitis, or change in mental status The chemotherapy to be administered
without evidence of increased intracranial should be checked to ensure the correct
pressure). drug, dose, mode of infusion, and patient
identification.
Pretreatment evaluation
1. Review patient history for use of antico- Materials
agulants (should be discontinued with appro- 1. Standard LP tray, 22‐gauge spinal needle
priate time interval prior to procedure), (1.5 in. for infants, 2.5 in. for older children,
seizures, or other CNS concerns including longer needles available as needed for larger
prior complications with LPs or IT chemo- patients), 25‐gauge needle for lidocaine
therapy. The review of systems should include injection if needed, providone iodine and
history of headaches, altered mental status, alcohol, mask, and sterile gloves. A Quincke
fevers, bleeding, back pain, or lower extremity needle is the standard needle used for LP;
weakness. Consideration should be given to however, the pencil point Whitacre needle
imaging with assessment of possible increased may be indicated in patients with prior
intracranial pressure or spinal compression ­history of severe spinal headache (see Post-
with any of these signs or symptoms. procedure monitoring and complications).
Guide to Procedures 395

2. Chemotherapy agent (s) to be adminis- 6. Administer local anesthetic with


tered. Errors have occurred in the past with preservative‐free 1% lidocaine with a 25‐
vincristine being inadvertently administered gauge needle, if desired. Awake patients may
intrathecally. This is almost uniformly fatal. prefer application of a topical lidocaine
This drug should never be brought into the anesthetic (LMX or EMLA) applied
room of a patient undergoing a LP with IT 20–30 minutes in advance of the procedure
chemotherapy. Many institutions now require or ethyl chloride cold spray at the moment
vincristine be made in small infusion bags to of the puncture. Local anesthesia is not
prevent this complication. Patients receiving necessary in the anesthetized patient.
double or triple IT therapy likely benefit from 7. Insert the spinal needle into the midline
these medications being mixed by the chem- of the interspace with the bevel parallel to the
otherapy pharmacy prior to instillation to spinal column, directed at a slight (10°–20°)
prevent missed medication as well as transfer angle toward the umbilicus. Ensure the nee-
in the operating room without a hood. dle is perpendicular to the surface. Advance
the needle; if bony resistance is noted, draw
Procedure back and reposition. The less experienced
1. Proper positioning is key to a successful practitioner should check for CSF flow every
procedure. The lateral decubitus position 2–3 mm by withdrawing the stylet and look-
may be used for a sedated or small patient. ing at the translucent window hub for visu-
The patient should be placed on a firm bed, alization of CSF. If blood is returned, the
head flexed with chin to chest, and legs needle should be removed and the procedure
maximally flexed toward the head. Ensure re‐attempted one interspace higher. The
the hips and shoulders are aligned and the most experienced practitioner should
back is straight. Alternatively, an awake assume responsibility for subsequent LPs.
patient may prefer the sitting position, 8. Once clear CSF return is established,
flexed forward, and supported by the some practitioners rotate the needle 90°
assistant or using a Mayo stand. counterclockwise (bevel in transverse plane)
2. Identify the landmark with palpation of for patients in the lateral decubitus position
the interspaces. The L4–5 interspace is to increase rapidity of CSF flow and allow
located by a perpendicular line at the top of for easier administration of IT chemother-
the iliac crests; either this space or the one apy. Patients in the sitting forward position
above (L3–4) may be used. do not usually need repositioning of the
3. The practitioner should wear a mask to needle. Obtain the desired amount of CSF in
decrease infection risk. Put on sterile gloves two or three collection tubes. Traditionally,
and set up the tray. the volume of CSF withdrawn should
4. Perform a sterile prep by cleansing the approximate the volume of medication
skin surface with providone iodine solution administered (minimum 50% of adminis-
beginning at the anticipated site of puncture, tered volume). CSF should be collected in
working outward with friction. Repeat for a one tube for a chamber count (red blood cell
total of three scrubs. The skin may then be and white blood cell counts) and a second
cleansed with alcohol. Some centers utilize a tube for cytology (assessment of morphol-
chlorhexidine scrub in lieu of providone ogy). Additional tubes may be necessary for
iodine. Place a fenestrated drape over the culture, immunohistochemistry, or any spe-
site and ensure an adequate sterile field with cial studies such as myelin basic protein,
additional drapes as needed. tumor markers, or research studies. Glucose
5. Open all CSF collection tubes. and protein are checked initially for all
396 Chapter 31

patients, though not routinely for leukemia anesthesia monitoring. Most centers direct
patients receiving IT chemotherapy. that patients remain in a supine position
9. For IT administration of chemotherapy, (without head pillow, mild Trendelenburg
attach the chemotherapy syringe to the spinalposition) for 1 hour postprocedure to mini-
needle, ensuring no advancement or with- mize the risk of spinal headache and assure
appropriate chemotherapy distribution within
drawal of the spinal needle and a tight fit to
avoid leakage. One can attempt to withdraw the CSF. More advanced Trendelenburg posi-
tioning is not routinely recommended.
CSF to assure proper placement (will see a mix
2. Patients should be monitored for signs of
in the syringe), but this may not be possible and
bleeding, pain at site, headache, nausea,
is not necessary. Slowly inject the chemother-
apy over 30–60 seconds. There should be no vomiting, or change in neurologic status. In
resistance. Most institutions do not perform addition to monitoring for postprocedure
complications, the patient’s cardiovascular
sterile transfer of chemotherapy (due to lack of
a hood), but rather break sterility with one and respiratory status should be monitored
hand which administers the chemotherapy along with frequent vital signs until
recovered from anesthesia.
while the second hand sterilely holds the hub of
the spinal needle. The system remains closed 3. Headache occurs following LP in a small
percentage of patients, primarily adoles-
so as to not put the patient at risk for infection.
10. Chemotherapy should not be adminis- cents and females. Typically, onset of head-
tered when any of the following situations ache is within 12 hours to 5 days of the
exist: the spinal fluid is bloody indicating procedure and is due to slow leakage of CSF
from the puncture site (though may not be
puncture of a vessel; the patient is moving pre-
visible externally). Other complications or
venting safe administration of the drug(s); the
chemotherapy does not advance easily and adverse effects include nausea, vomiting,
when the syringe is removed the flow of CSF dizziness, neck stiffness, light sensitivity,
has stopped or greatly diminished; the chemo-and diminished hearing or vision. These
symptoms may be worse when the patient
therapy drug or dose is incorrect; or the awake
patient experiences pain with injection. assumes a standing position.
11. Remove the needle. Apply gentle pres- a. Patients are instructed to take fluids
sure, cleanse as necessary with alcohol, and liberally in addition to caffeine (highly
place a spot bandage. caffeinated sodas or coffee), though this
12. Assess patient for any adverse effect of latter intervention is not evidence‐based.
the procedure. Have the patient in the supine b. Patients with severe headache or neu-
position. rologic symptoms should be evaluated
13. Label all CSF tubes and send to the lab. immediately. If a postdural puncture
14. Document the procedure in the medical headache is suspected, treatment is initi-
record. ated with caffeine, fluids, and analgesics.
15. Follow up with the patient and family as If these steps are ineffective, a dural blood
to the results of the procedure, including patch may be performed by an anesthesi-
interpretation of the CSF specimens. ologist to provide immediate relief.
c. Use of a pencil point spinal needle
Post-procedure monitoring (Whitacre) for subsequent LPs should be
and complications considered for these patients. These
1. Observe the patient for a minimum of needles are designed to spread the dural
1 hour (or longer if necessary) for recovery fibers and help reduce the frequency and
from anesthesia with appropriate post-­ severity of postdural headaches. They
Guide to Procedures 397

can be more difficult to use and may are placed infrequently, they are useful in the
require an introducer to puncture the management of patients for whom it is tech-
skin and soft tissues. nically difficult to perform LPs for any rea-
4. Fever may occur following administration son. Lumbar catheters have also been placed
of IT chemotherapy. Some drugs (e.g., cytara- in patients with unusual anatomy or in the
bine) have been more implicated in causing presence of obesity; however these catheter
fever; however, fever should never be assumed reservoir systems are more prone to dysfunc-
to be due to drug and the patient should be tion than the intraventricular systems.
assessed for presumed infection. Frequently, Ommaya reservoirs are also used in patients
patients have indwelling central venous cathe- who suffer a CNS relapse of their leukemia
ters (CVCs) and are receiving other immuno- and require frequent administration of IT
suppressive therapy. Evaluation should be chemotherapy. Practitioners may opt to
comprehensive with appropriate clinical, physi- administer chemotherapy in reduced dosing
cal, and laboratory assessments and may include compared to standard IT dosing (50–100%
hospitalization for observation with adminis- of IT dosing) or give small daily dosing for
tration of intravenous (IV) antibiotics. up to 4 days (based on the concept of con-
5. Patients may experience pain or bleeding centration × time to optimize therapeutic
at the LP site for several days. The patient benefit).
should be evaluated and, if experiencing
minor bleeding, be treated with local therapy Pretreatment evaluation
(dry sterile bandaging) and pain medication The same principles apply as in LP/IT
as needed. Prolonged or heavy bleeding chemotherapy. Patients are not sedated for
requires immediate evaluation including this procedure.
physical examination and imaging.
6. Neurotoxicity may occur related to the Materials
IT medication or related to nerve damage 1. Standard LP tray, two 5 ml sterile syringes
secondary to the procedure. Patients should for CSF collection, 25‐gauge butterfly nee-
be evaluated immediately and intervention dle, mask, razor, antibacterial soap, 4 × 4
be taken as appropriate with subsequent sterile gauze, providone iodine and alcohol,
neurologic assessments and possible and two sets of sterile gloves.
imaging. Decisions regarding further IT 2. Chemotherapy agent(s) to be adminis-
therapy are made pending resolution of tered. Do not bring vincristine into the room.
symptoms and findings on imaging, as well
as clinical protocol. Procedure
1. The patient is placed in a supine or sitting
position. If needed, a small area over the
Intra‐Ommaya reservoir tap reservoir is shaved.
and injection of chemotherapy 2. Topical anesthesia may be achieved with
lidocaine gels (EMLA or LMX) applied
Indications 20–30 minutes prior to the procedure.
The Ommaya reservoir is an intraventricular 3. While wearing a mask and sterile gloves,
catheter with a reservoir implanted under the LP tray is set up.
the scalp that allows for administration of 4. A double sterile preparation is routinely
chemotherapy or other medication directly done when accessing intraventricular reser-
into the ventricular system, in addition to voirs due to concern for infection. Prepare
facilitating sampling the CSF. Although they the skin surface overlying the reservoir site
398 Chapter 31

by cleansing with sterile gauze moistened Postprocedure monitoring


with an antibacterial soap (e.g., chlorhex- and complications
idine) in a circular fashion, three times. 1. Document in the patient’s chart details of
Change sterile gloves. Continue with a rou- the procedure, chemotherapy administered,
tine sterile prep with three providone iodine specimens collected, and the patient’s status
scrubs and alcohol. Place a fenestrated drape after the procedure.
over the site and ensure an adequate sterile 2. See Postprocedure monitoring for LP
field with additional drapes as desired. regarding complications such as headache,
5. Open all CSF collection tubes. fever, bleeding, or neurologic changes.
6. Holding the reservoir firmly with one
hand, puncture the reservoir site with a 25‐
gauge butterfly needle. The CSF is allowed to Bone marrow aspiration
drip (or is slowly withdrawn) from the but- and biopsy
terfly into a sterile tube. The total volume
collected should approximate the total vol- Indications
ume of chemotherapy and normal saline The purpose of a BMA or biopsy is to obtain
flush to be delivered. CSF should be collected tissue for diagnostic and staging evaluation
in one tube for a chamber (cell) count and a of malignancies, marrow infiltrative dis-
second tube for cytology. Additional tubes eases, or marrow failure states. Bone marrow
may be necessary for culture, immunohisto- examination is required for the diagnosis of
chemistry, or any special studies such as leukemia, lymphoma, bone marrow failure
myelin basic protein, tumor markers, or states, evaluation of pancytopenia of
research studies. Glucose and protein are unknown etiology, suspected storage dis-
checked initially for all patients, though not eases, and certain causes of anemia. Bone
routinely for leukemia patients receiving marrow studies are also done as staging eval-
intra‐Ommaya chemotherapy. uations in certain solid tumors including
7. The chemotherapy is injected slowly high‐risk neuroblastoma and Ewing sar-
after nonsterile transfer of the chemotherapy coma. The bone marrow produces the cellu-
syringe similar to the LP procedure, keeping lar elements of the blood including platelets,
one hand sterile at the hub of the butterfly red blood cells, and white blood cells in addi-
needle to ensure a closed system. Do not tion to the supporting matrix to allow for cell
give the chemotherapy if the CSF is blood‐ growth and maturation. Aspirates are rou-
tinged. A small amount of sterile saline or tinely obtained for morphologic as well as
the patient’s CSF is then injected to ensure immunohistochemical and flow cytometric
the tubing is flushed and all the medication evaluation in patients with suspected leuke-
is delivered (typically <0.5 ml, the volume of mia. Those with suspected or known solid
tubing and reservoir). tumors, storage diseases, aplastic anemia, or
8. The needle is removed, and firm pressure other marrow failure states require evalua-
is applied to the site. A spot bandage may be tion by bone marrow biopsy as well. A core
applied. section of the marrow matrix is obtained in
9. Assess the patient for any adverse effects order to assess the suitability of the marrow
from the procedure. environment for growth of normal cellular
10. Label all CSF tubes and send to the lab. elements, the cellularity of the marrow, and
11. Follow up with the patient and family as the presence of abnormal cells that may be
to the results of the procedure, including adherent to the trabeculae. Samples are typi-
interpretation of the CSF specimens. cally taken from more than one site when
Guide to Procedures 399

looking for evidence of marrow involvement Procedure


by solid tumors in order to increase the sen- 1. Position patient in a prone or lateral
sitivity of the test. The most common site for decubitus position with a small lift under the
sampling in the child or adolescent is the hip to accentuate the posterior iliac crests.
posterior iliac crest. At times, other locations Identify the posterior superior iliac spine.
for bone marrow specimen collection are Have assistant secure patient’s position. Put
necessary (i.e., infants, children on ventila- on gloves and set up sterile tray.
tors) and may include the anterior iliac spine, 2. Scrub the site with providone iodine,
tibia, sternum, or other sites. applying some friction, beginning at the site
and moving outward, and repeating for a
Pretreatment evaluation total of three scrubs. Repeat procedure with
1. Review of systems including recent alcohol swabs. Allow to dry and apply sterile
illnesses or back pain. drapes.
2. Physical examination should be 3. Administer local anesthetic with a 22–25‐
performed prior to the procedure, with gauge needle. This is often done in the
vitals and assessment for any evidence of sedated patient to minimize postprocedure
infection or metabolic abnormalities to discomfort. Local anesthesia is achieved with
assure safety for anesthesia. Evaluate site of lidocaine 1% (2–3 ml depending on size of
procedure to assure no localized infection or the patient) injected down to and including
skin breakdown. the periosteum.
3. Routine laboratory studies may include a 4. Prepare marrow aspirate and/or biopsy
complete blood count, chemistries, and needles, ensuring the stylets are freely
coagulation studies. No specific platelet removable. Prime syringes as per institu-
threshold is necessary for the procedure and tional protocol with heparin, EDTA, etc.
it can be safely done at counts of 1 × 109/l Ensure inner side of syringe is coated with
with pressure bandages applied. The anticoagulant, if being used.
anesthesiologist will want to ensure an 5. Holding skin taught with outstretched
adequate hemoglobin level for anesthesia fingers, insert the BMA needle (with stylet
and be aware of any metabolic abnormalities. in place) initially at an angle to the skin and
4. The technician to assist with the marrow then perpendicular to the iliac spine once
slide preparation should be present at the through the skin. With gradual, controlled
start of the procedure with appropriate pressure and a gentle twisting motion,
materials. insert the needle into the iliac spine. Once
through the cortex, the needle will “give” as
Materials it enters the marrow space and a crunching
1. Biopsy tray with providone iodine, alco- sound or feeling may be appreciated. The
hol, 4 × 4 gauze, 20 ml syringes (two to four stylet should then be removed, and the nee-
depending on samples to be collected), dle should remain firmly in place. A 20‐ml
15‐gauge bone marrow aspirate needle, syringe is then firmly attached to the hub of
11‐gauge Jamshidi biopsy needle (or simi- the aspiration needle. Holding onto both the
lar), 22–25‐gauge needles, sterile gloves, and needle and syringe, constant and strong
Elas­toplast adhesive or other pressure pressure should be applied to draw up mar-
dressing. row into the syringe. If awake, the patient
2. Ethylendiaminetetraacetic acid (EDTA) may experience a shooting sensation down
and heparin to anticoagulate samples (if the legs at the time of aspiration. Aspirate
done), lidocaine 1% for local anesthesia. approximately 1–2 ml of marrow, detach the
400 Chapter 31

syringe carefully, and hand the specimen site and secured with an elastic pressure tape
immediately to the technician for quick vis- such as Elastoplast.
ual inspection for marrow tissue (fat, spic- 8. Assess the patient for any adverse effect
ules). Once this has been confirmed, obtain of the procedure.
additional marrow as needed with the other 9. Document procedure(s) in the medical
syringes, obtaining not more than 3–5 ml record.
per pull. In order to obtain optimal speci- 10. Follow up with the patient and family to
men, repositioning of the bone marrow ensure completion of procedure, explanation
aspirate needle should also be considered of any complications, review care of wound,
between pulls and should always be and follow up on results of studies done on
attempted if the first specimen does not con- specimens obtained.
tain spicules. Once complete, remove the
needle. If spicules are not present even with Post-procedure monitoring
repositioning, a biopsy should be consid- and complications
ered for adequate sampling. 1. Patients may be discharged from the
6. A bone marrow biopsy may be obtained recovery area after appropriate monitoring
from the same side and skin puncture site as post-anesthesia.
the aspirate. The needle should be inserted 2. The pressure dressing, if used, should be
into a fresh spot on the iliac spine (although removed after 6 hours and the bandage after
should utilize the same skin puncture site). 24 hours. The parents should inspect the site
Holding the skin tight, insert the bone mar- for evidence of infection, bleeding, or other
row (trephine) biopsy needle (with cutting drainage. Pain medications including aceta-
trocar in place) holding at an angle until minophen, acetaminophen with codeine, or
through the skin, then placing perpendicular occasionally an IV narcotic may be adminis-
to the spine and inserting with strong, con- tered for local pain (especially following
trolled pressure until the needle is firmly biopsy). Patients may feel achy or bruised
anchored into the cortex. Remove the trocar for several days following the procedure.
and holding the forefinger along the needle Discomfort can also be alleviated with a
at the desired depth of the core biopsy warm pack.
(5–20 mm depending on size of the patient), 3. Patients may resume normal activities as
insert the needle with a firm twisting pres- desired.
sure. The needle is then rocked in four angles
(i.e., sideways as well as up and down) to
break off the core marrow biopsy sample at Administration of peripheral
the base (unless a cutting needle is used) and chemotherapy
then the needle is removed. The provided
push rod (no sharp edge) is inserted into the Indications
sharp end of the needle and the biopsy is IV chemotherapy may be given into a CVC
pushed out gently onto sterile gauze. It is or by peripheral vein administration in
then examined to ensure that adequate mar- patients without a CVC. Many patients may
row tissue is present and given to the have had their catheters removed due to
technician. infection, thrombosis, or electively in order
7. Apply pressure for several minutes, for the patient to resume more normal activ-
cleanse the site with alcohol swabs, and ities. In the hands of experienced practition-
apply a bandage. If needed, a dry 4 × 4 gauze ers, administration of push IV chemotherapy
folded into quarters may be applied over the is a safe alternative to the use of a CVC.
Guide to Procedures 401

Vincristine is frequently administered via in vasoconstriction and increase difficulty


peripheral vein in the outpatient setting dur- of access in some patients.
ing maintenance phases of treatment for
acute lymphoblastic leukemia. Peripheral Materials
access should not be used routinely for 1. Chemotherapy (premixed by pharmacy
longer infusional chemotherapy due to the in enclosed syringe).
difficulty in monitoring and ensuring con- 2. 25‐gauge butterfly needle, stopcock,
tinued patency of the IV during infusion. alcohol wipes, gauze (2 × 2), tourniquet,
Many chemotherapeutic drugs are vesicants 10 ml saline flush, bandage, and gloves.
and may cause significant injury with
extravasation (see Chapter 32). Procedure
1. Assemble equipment; attach butterfly
Pretreatment evaluation tubing to stopcock, attach chemotherapy
1. Review the patient’s chemotherapy regi- syringe to right side of stopcock, attach
men to determine what medications are to be saline flush syringe to remaining junction of
administered and at what dose. The patient’s stopcock, and flush stopcock and butterfly
height, weight, and total body surface area are with saline.
verified with the dosage calculations. 2. Select an appropriate vein, preferably on
2. Ensure that required laboratory criteria the dorsum of the hand or foot. Ask the child
have been met (e.g., absolute neutrophil to assume a comfortable position that also
count, platelet count, transaminases) as allows for easy access to the desired vein.
directed by the treatment protocol. Avoid the antecubital fossae or joint spaces
3. Complete a physical examination on the due to the possibility of deep extravasation of
patient and assess the adequacy of chemotherapy with resultant injury such as
venipuncture sites, avoiding joints. contractures. These deeper spaces also pre-
4. Verify the labels on the syringe(s) of the vent immediate knowledge of extravasation.
drug(s) against the patient’s chart and orders 3. Clean the venipuncture site with alcohol
to ensure accuracy. and apply a tourniquet. Let air dry or wipe
5. Prepare the patient for the procedure. dry with clean gauze. Insert butterfly needle
Explain the procedure, taking into account with bevel pointed up. Advance needle until
the patient’s age, developmental status, and blood returns. When blood returns, remove
prior experience with the procedure. Elicit tourniquet, connect stopcock, and flush line
the patient’s help by encouraging him/her with 2–3 ml of saline.
to hold as still as possible. Enlist the assis- 4. If no sign of infiltration occurs (e.g., pain
tance of the parent(s), staff (including child or swelling) and blood return continues,
life specialist if available), and other distrac- administer chemotherapy slowly via IV push,
tions (iPad, virtual reality) as available and checking intermittently every 5–10 seconds
needed. Explain each step as you go; be for blood return or swelling. When chemo-
honest, thorough, and patient. Establish a therapy administration is completed, flush
routine with each patient; many also like to with remaining saline.
have the same practitioner if possible (con- 5. Remove butterfly and apply pressure
fidence boosting). for 1 to 2 minutes with clean gauze. Apply
6. Patients desiring topical anesthesia bandage.
should have a topical lidocaine gel (EMLA 6. Document in the patient’s chart the indi-
or LMX) applied 20–30 minutes in advance cation for and details of the procedure, med-
of the procedure. This however may result ication, dosage, site, and any complications.
402 Chapter 31

Postprocedure monitoring should not be used for future administration


and complications of chemotherapy. At times, subclinical burns
If infiltration has occurred, see Chapter 32. and scarring may occur and can appear as
Areas that may have sustained tissue damage hyperpigmented areas.
32 Treatment of
Chemotherapy
Extravasations
In addition to potential acute and late clines are among the most important, both
adverse reactions to chemotherapy, acciden- because of their widespread use in various
tal extravasation is a major safety concern. chemotherapeutic protocols and because of
Preventative strategies to minimize compli- their ability to produce severe tissue damage
cations of administration, early recognition, and necrosis. The extent of tissue damage
and an understanding of unique manage- depends on the chemotherapeutic agent’s
ment approaches and antidotes are critical binding capacity to DNA:
to lessening the acute injury as well as long‐ ●● Extravasation of an irritant drug may

term consequences. cause an inflammatory reaction, with pain,


Extravasation is the leakage of an intra- burning, tightness, or phlebitis at the needle
venous (IV) drug into the surrounding tis- insertion site or along the vein. Clinical
sues. Local reactions from extravasation of a signs include warmth, erythema, and ten-
vesicant chemotherapy agent can range derness in the area of extravasation, but
from mild pain and erythema to tissue there is no tissue sloughing or necrosis.
necrosis, ulceration, and damage to tendons Symptoms are typically of short duration
and nerves. Cytotoxic drugs are classified as (days) without long‐lasting sequelae.
irritants or vesicants (causing blisters), ●● Extravasation of a vesicant drug may

based on their potential for local toxicity, cause tissue necrosis with a more severe or
though this distinction is not absolute. The lasting injury. Clinical signs and symptoms
tissue injury may also be related to the may be similar to extravasated irritants.
amount of drug extravasated. Vesicant extravasation may result in loss of
Although this complication is frequently the full thickness of the skin and, if severe,
encountered with antineoplastic agents, a underlying structures.
number of other drugs can also act as vesi- The true incidence of vesicant extravasa-
cants if extravasated into the surrounding tion injury is unclear, but it is estimated to
tissues. These noncytotoxic drugs include range from 0.5–6% with peripheral IV infu-
alcohol, aminophylline, digoxin, nafcillin, sions, and 0.3–4.7% with implanted venous
phenytoin, tetracycline, and total parenteral access port infusions. Realistically, the inci-
nutrition. In terms of cytotoxic drugs that dence is likely higher, as there is no central
cause extravasation injury, the anthracy- mechanism of reporting these events.

Handbook of Pediatric Hematology and Oncology – Children’s Hospital & Research Center Oakland,
Third Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
© 2021 John Wiley & Sons Ltd. Published 2021 by John Wiley & Sons Ltd.
404 Chapter 32

Treatment of an extravasation is determined recommendations (i.e., proper dilution


by the particular chemotherapy agent and specified administration time) with
involved, although the efficacy of such ther- proper verification and identification
apy may be modest. (patient and protocol specific).
Prevention of extravasation is key, and ●● Avoid multiple venipunctures in the same

every possible measure should be taken to area. Do not use a vein distal to a recent
avoid such a complication. If it is anticipated venipuncture site, as the vesicant may leak
that a patient will need frequent or pro- from one of these proximal sites.
longed infusions of vesicants, it is advisable ●● Instruct the patient to avoid movement.

to place a central venous catheter (CVC) for In young children, this may require addi-
safer drug administration by providing reli- tional physical assistance for holding or
able venous access, high flow rates, and coaxing with diversion techniques.
rapid drug dilution. Rarely, extravasation ●● Never use a previously placed IV access

may occur even with such a device due to device; a new IV cannula should be placed
injection technique or device failure, includ- immediately prior to delivery of the
ing catheter migration. Providing education medication.
to patients and families is important, and ●● Choose a large, intact visible vein with

they should be instructed to immediately good blood flow.


report any burning sensation related to the ●● Use the smallest needle or cannula possi-

drug infusion, swelling, redness, or pain. ble for venipuncture. Check the patency of
The causes of extravasation are multiple the device by aspirating blood, as well as
and largely preventable. Risk factors for patency of the vein by flushing with the car-
peripheral IV extravasation include: poor rier solution (normal saline), before admin-
vein selection, multiple venous punctures to istering the medication. Obtain a blood
establish a patent IV, obesity, dehydration, return prior to, and during, vesicant
inability to report pain at the injection site, a administration.
moving patient, and inexperience of the ●● The IV infusion should flow freely with-

individual administering the chemotherapy. out pressure. The local area should not swell,
Risk factors for extravasation from CVCs become erythematous, or cause pain. If this
include needle displacement, catheter occurs, immediately stop the infusion and
migration, or fibrin sheath formation and attempt to withdraw any medication in the
thrombosis. Mechanical occlusions may be needle/cannula.
due to thrombus formation, drug precipita- ●● After infusing the medication, flush the

tion, and positional catheter occlusion or vein with 3–10 ml of the carrier solution. Do
kinking of the line. Avoidance of extravasa- not continue to flush if resistance is met or a
tion depends on proper placement and local reaction suggesting a blown vein
maintenance of IV access and frequent occurs.
monitoring. ●● For ports, it is vital to ensure appropriate

Recommendations to prevent extravasa- securement of the Huber needle with dress-


tion are: ings that enable the provider to visualize the
●● Venipuncture and placement of the can- site of access.
nula or other IV access should be performed Each extravasation incident should be
by experienced personnel. documented and reported, according to
●● Vesicants should be administered in local institutional guidelines. Standards of
accordance with the manufacturers’ practice as well as documentation should be
Treatment of Chemotherapy Extravasations 405

in place and most centers utilize national permanent hyperpigmentation at the site of
guidelines (including management) based drug extravasation. Tissue damage can
on the ASCO (American Society of Clinical injure tendons, nerves, and joints in the
Oncology) or ONS (Oncology Nursing affected area. For this reason, it is recom-
Society) published safety standards for mended that peripheral access for IV chem-
chemotherapy administration. otherapy administration never be in an area
DNA‐binding agents including anthracy- near a joint or deep tissue site, as extravasa-
clines, antitumor antibiotics, platinum ana- tion may lead to contractures or serious
logs, and some alkylating agents can cause injury before the leakage is recognized.
tissue damage by propagating lethal DNA Extravasation sites should not be utilized for
cross‐linking or strand breaks caused by free subsequent administration of chemotherapy
radicals, which lead to cell apoptosis. As the or placement of an IV device.
cells die, the drug is released and enters Treatment should begin immediately
undamaged cells. This further increases the with discontinuation of the chemotherapy
area of damage and slows healing. and thermal application and possible dilu-
Anthracyclines have the greatest vesicant tion of the site. The line should not be
potential compared to other chemotherapeu- flushed and avoid applying pressure to the
tic agents and are characterized by immediate site. The needle/catheter should not be
burning or severe pain, and lesions may con- removed immediately, but should be left in
tinue to evolve slowly over weeks due to tis- place to attempt to aspirate fluid from the
sue retention of the extravasated vesicant. extravasated area with a 10‐ml syringe, as
Non‐DNA‐binding antineoplastics well as administer an antidote if appropriate
(vinca alkaloids, taxanes, and topoisomer- (see Table 32.1). Clinicians should work
ase inhibitors) also function as vesicants by quickly to reduce morbidity and avoid fur-
interfering with mitosis. These agents clear ther patient harm. Management of non‐­
more easily from extravasation sites and vesicant extravasation includes elevation
cause less damage than the DNA‐binding and cooling and does not usually include the
agents. The tissue often resembles a chemi- use of pharmacologic therapy. Cold com-
cal burn and blisters may develop. presses can reduce pain and local inflamma-
The signs and symptoms of extravasa- tion by causing vasoconstriction and
tion may be readily apparent with a burning reducing the potential for continued spread
sensation, swelling, pain, or erythema, of the drug. Cold compresses are contrain-
although it may take days for the full extent dicated however in the case of extravasation
of the epithelial damage to be evident. with vinca alkaloids, as it may cause further
Caregivers should be careful to never under- tissue damage. In these situations, warm
estimate the significance of any symptom compresses and heat can be applied to the
the patient may report related to the infu- affected area leading to vasodilation and
sion, and perform a careful assessment of absorption of extravasated drug from the
the infusion and site. Sometimes patients tissue site (see Table 32.1). Other interven-
are not symptomatic and the tissue damage tions under investigation include local
becomes evident days to weeks later. administration of granulocyte‐macrophage
Discoloration and skin induration may pro- colony‐stimulating factor or local injection
gress with the development of blisters or of normal saline. Removal of the CVC may
necrosis and possibly ulceration or deep tis- be indicated for extensive extravasations or
sue injury. Patients may develop scarring or in the case of device malfunction.
406 Chapter 32

Table 32.1 Extravasation treatment.

Drug Local care Pharmacologic treatment

DNA‐binding vesicants
Anthracyclines Cold pack, 20 min Dexrazoxane 1000 mg/m2 IV over
Doxorubicin 4 times/d × 48–72 h 1–2 h (within 6 h) on days 1 and 2;
Daunomycin Elevate 500 mg/m2, day 3
If dexrazoxane unavailable or minor
extravasation: DMSO 99% topical
4 drops per 10 cm2 over twice the size
of the affected area within 10–25 min,
then q8 hour × 7–14 d; allow to dry,
then apply nonocclusive dressing*
Do not use Dexrazoxane and DMSO
concurrently
Alkylating agents Cold pack, 20 min None
Nitrogen mustard 4 times/d × 48–72 h Sodium thiosulfate
Elevate 10% 2 ml in 6 ml sterile water for
IV/SC injection*
Other Cold pack, 20 min None
Dactinomycin 4 times/d × 48–72 h DMSO as mentioned above*
Mitomycin C Elevate Hydrocortisone 1% cream topically
Dacarbazine
Non‐DNA‐binding vesicants
Vinca alkaloids Warm pack, 20 min None
Vinblastine 4 times/d × 48–72 h Hyaluronidase 150 units in 1 ml injected
Vincristine Elevate SC in multiple sites with small gauge
Vindesine needle*
Taxanes Cold pack, 20 min Hyaluronidase as mentioned above*
Docetaxel 4 times/d × 48–72 h
Paclitaxel Elevate
Irritants
Alkylating agents
Carboplatin Cold pack as Hydrocortisone 1% cream topically
Cisplatin mentioned above
Topotecan None None
Ifosfamide No local care
Cyclophosphamide
Melphalan
Antimetabolites
Cytarabine, fludarabine None None
Methotrexate Cold pack as Hydrocortisone 1% cream topically
5‐fluorouracil mentioned above
Gemcitabine None None
Other
Bleomycin None None
Etoposide, irinotecan Cold pack as Hydrocortisone 1% cream topically
mentioned above

* Suggested as possible antidote in the literature; lack of prospective studies to currently advocate
as treatment. Abbreviations: DMSO, Dimethyl Sulfoxide; IV, intravenous; SC, subcutaneous.
Treatment of Chemotherapy Extravasations 407

The placement of extravasation kits con-


Suggested reading
taining syringes and cannulas, cold and hot
packs, gauze pads, sterile and chemoprotec-
Jacobson, J.O., Polovich, M., Gilmore, T.R. et al.
tive gloves, and medications to treat extrava- (2012). Revisions to the 2009 American Society
sation in locations where chemotherapy is of Clinical Oncology/Oncology Nursing
administered facilitate early treatment (see Society chemotherapy administration safety
Table 32.1). In addition to local care and pos- standards: expanding the scope to include
sible pharmacologic antidotes, topical hydro- patient inpatient settings. J. Oncol. Pract. 8: 2.
cortisone 1% and oral pain medications may Kreidieh, F.Y., Moukadem, H.A., and El Saghir,
alleviate local discomfort. Patients should be N.S. (2016). Overview, prevention and man-
instructed to continue to elevate the affected agement of chemotherapy administration.
area as possible for 24–48 hours and apply the World J. Oncol. 7: 87–97.
Neuss, M.N., Polovich, M., McNiff, K. et al.
appropriate thermal intervention four times a
(2013). 2013 updated American Society of
day for 20 minutes for 2–3 days. Monitoring
Clinical Oncology/Oncology Nursing Society
the area with daily photos may be helpful. chemotherapy administration standards
Clinical follow‐up is recommended, as the including standards for the safe administra-
extent of damage may not become apparent tion and management of oral chemotherapy.
until days or weeks following the injury. Oncol. Nurs. Forum. 40: 225.
Rarely, referral for surgical intervention, such
as debridement of necrotic tissue or skin
grafting, may be necessary.
Formulary

Sample entry: drug in pregnant women may be acceptable


Generic name despite its potential risk.
Trade and other names E. Studies in animals or humans demon­
Drug category strate fetal abnormalities or adverse reaction;
How supplied reports indicate evidence of fetal risk. The
Pregnancy category (see explanation in the risk of use in a pregnant woman clearly out­
following text) weighs any possible benefit.
Indications
Dosage ACTINOMYCIN D, see DACTINOMYCIN
Notes, including adverse events, monitor­
ACYCLOVIR
ing, and dose modification
Zovirax, generics
Pregnancy categories: Antiviral
Capsules: 200 mg
A. Adequate studies in pregnant women
Tablets: 400, 800 mg
have not demonstrated a risk to the fetus in
Suspension: 200 mg/5 ml
the first trimester of pregnancy, and there is
Injection: 500 mg vial
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
Indications:
studies in pregnant women; or animal stud­
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
Dosage:
on the fetus, but there are no adequate studies in
humans; or there are no animal reproduction Children over age 12 years:
studies and no adequate studies in humans. Mucocutaneous HSV
D. There is evidence of human fetal risk, Initial infection: 750 mg/m2/day IV divided
but the potential benefits from the use of the q8h or 15 mg/kg/day IV divided q8h for

Handbook of Pediatric Hematology and Oncology – Children’s Hospital & Research Center Oakland,
Third Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
© 2021 John Wiley & Sons Ltd. Published 2021 by John Wiley & Sons Ltd.
410 Formulary

5–7 days; 1000–1200 mg/day PO divided prevention of recurrent calcium oxalate cal­
into 3–5 doses for 7–10 days. culi; prevention of gouty arthritis and
Recurrence: 1000 mg/day divided into 5 nephropathy.
doses or 1600 mg/day divided q12h or
2400 mg/day divided q8h for 5–10 days Dosage:
(maximum pediatric dose 1000 mg/day). Children: 10 mg/kg/day PO divided q6–8h;
maximum dose 800 mg/day or 200–300 mg/
Immunocompromised host (all ages) m2/day IV q8–24h; maximum 800 mg/day
HSV: 750–1500 mg/m2/day IV divided q8h or 200 mg/m2/day IV divided q6–12h (max­
for 7–14 days or (≥2 years) 100 mg/day PO imum dose 600 mg/day).
divided into 3–5 doses for 7–14 days (maxi­ Adolescent to adult: 200–800 mg/day PO
mum child dose 80 mg/kg/day). divided q8–12h for the prevention of acute
HSV prophylaxis: 750 mg/m2/day IV divided uric acid nephropathy; or 200–400 mg/m2/
q8h or 600–1000 mg/day divided q6–8h day IV divided q6–12h (maximum dose
during risk period (maximum child dose 600 mg/day).
80 mg/kg/day).
Varicella or zoster: 1500 mg/m2/day IV Notes:
divided q8h or 20 mg/kg/dose IV divided q8h Reduce dosage in renal impairment; discon­
for 7–10 days (maximum PO dose 800 mg). tinue with rash (may be exacerbated with
ampicillin or amoxicillin). Risk of hypersen­
sitivity may be increased in patients receiving
Notes:
thiazides/angiotensin‐converting enzyme
Adjust dose in renal impairment. Adequate inhibitors. It may cause fever, neuritis, gastro­
hydration and slow IV (1 hour) administration intestinal (GI) disturbance, hepatotoxicity,
are essential to prevent crystallization in the bone marrow suppression, and drowsiness.
renal tubules. Oral absorption is unpredictable Avoid concomitant use of amoxicillin, ampi­
(15–30%), consider use of valacyclovir for bet­ cillin, mercaptopurine, cyclophosphamide,
ter absorption. Use ideal body weight for obese theophylline derivatives, and vitamin K
patients when calculating dosage. antagonists.

ADRIAMYCIN, see DOXORUBICIN ALTEPLASE


ALLOPURINOL Tissue plasminogen activator (TPA)
Activase, Cathflo Activase, TPA
Zyloprim, Alloprim, generics
Thrombolytic
Uric acid lowering agent, xanthine oxidase
Injection:
inhibitor, antigout agent
Cathflo Activase: 2 mg/2 ml vial
Tablets: 100, 300 mg
Activase: 50 mg (29 million units), 100 mg
Suspension: 20 mg/ml
(58 million units)
Injection (Alloprim): 500 mg vial
Pregnancy category C
Pregnancy category C
Indications:
Indications: Treatment of recent severe or massive
Prevention of uric acid nephropathy in mye­ deep vein thrombosis (DVT) or arterial
loproliferative neoplastic disorders that may thrombosis, pulmonary embolus, or
occur as a result of tumor lysis syndrome; occluded central venous catheter (CVC).
Formulary 411

Dosage: be followed daily and fresh frozen plasma be


Pulmonary embolus, DVT, central venous given for replenishment if level <50%.
thrombosis, superior vena cava syndrome:
Systemic thrombolytic therapy should be AMIFOSTINE
given in consultation with a hematologist. Ethyol
Safety and efficacy have not been estab­ Antidote for cisplatin, cytoprotective agent
lished in pediatric patients. Injection: 500 mg vial
<3 months of age: 0.06 mg/kg/hour for
Pregnancy category C
6–24 hours.
≥3 months of age: 0.03 mg/kg/hour (maxi­ Indications:
mum 2 mg/hour).
A cytoprotective drug that scavenges free
If no clinical improvement in 24 hours, dou­ radicals and binds to reactive drug deriva­
ble the dose to 0.06–0.12 mg/kg/hour. tives reducing toxicity of radiation and
Systemic TPA can be given for up to 96 hours. platinum‐containing and alkylating agents
In the case of a pulmonary embolus, bolus such as cisplatin, carboplatin, ifosfamide,
dosing of 1 mg/kg up to a maximum of carmustine, melphalan, mechlorethamine,
50 mg may be given. and cyclophosphamide. The safety and
effectiveness of amifostine have not yet
Maximum duration of therapy is 96 hours or
been established in pediatric patients and
based on the patient’s clinical course.
its use has been primarily in the setting of
Preferable to infuse thrombolytic agent dis­ clinical trials.
tal to (and as close to) the site of thrombus
as possible. Dosage:
Occluded CVC (dosage per lumen, treating Refer to individual protocol.
one lumen at a time): Usual dosage is 740–910 mg/m2 IV daily
Weight < 30 kg: instill volume to equal 110% over 15 minutes starting 30 minutes prior to
of internal volume of the catheter, not to the dose of a platinum or alkylating agent.
exceed 2 mg. Infusions over less than 15 minutes are asso­
Weight ≥ 30 kg: 2 mg per lumen. ciated with a higher incidence of adverse
reactions.
Instill a dose in each lumen of the CVC;
allow to dwell for 30 minutes. If unsuccess­
ful, repeat the dose. If the catheter remains Notes:
obstructed, begin systemic thrombolytic Avoid in hypotension or dehydration. Can
therapy as outlined in the preceding text. cause severe nausea and vomiting and usu­
ally requires antiemetic premedication.
Allergic reactions, severe cutaneous toxicity
Notes:
(more commonly when used as a radiopro­
Avoid central venous puncture and noncom­ tectant), and hypocalcemia have been
pressible arterial sticks during infusion. reported. Ensure adequate hydration and
Avoid use in excessive hypertension, within monitor calcium levels.
10 days of a cerebral vascular accident, with
gastrointestinal bleeding or trauma, within
1 week of surgery, in patients with a bleeding AMINOCAPROIC ACID
diathesis, or with suspicion of a subarach­ Amicar, generics
noid hemorrhage. Plasminogen levels should Hemostatic agent
412 Formulary

Tablet: 500, 1000 mg AMPHOTERICIN B (Conventional)


Syrup: 250 mg/ml Generics (previously available as Fungizone)
Injection: 250 mg/ml (20 ml) Polyene antifungal
Pregnancy category C Injection: 50 mg vial
Pregnancy category B
Indications:
Enhances hemostasis when fibrinolysis con­ Indications:
tributes to clinical bleeding. Excessive activ­
ity of the fibrinolytic system may be present Treatment of severe systemic infections and
following cardiac surgery, portacaval shunt, meningitis caused by susceptible fungi such
hepatic cirrhosis, aplastic anemia, and as Candida species, Histoplasma capsulatum,
abruptio placentae. Typically used to treat Cryptococcus neoformans, Aspergillus spe­
mucosal‐type bleeding in patients with cies, Blastomyces dermatitidis, Torulopsis
bleeding diatheses (i.e., von Willebrand dis­ glabrata, Coccidioides immitis, Mucormycoses,
ease, mild hemophilia, immune thrombocy­ and Rhizopus. Also used empirically to treat
topenia purpura). suspected invasive fungal disease in immu­
nocompromised hosts with prolonged fever
and neutropenia. May be given intrathecally
Dosage:
or via bladder irrigation for localized ther­
100–200 mg/kg IV/PO loading dose (maxi­ apy. Note, currently the lipid formulations of
mum 10 g), followed by 50–100 mg/kg amphotericin are used as first line.
q4–6h maintenance dose; maximum
30 g/24 hour. Treat until symptoms resolve
Dosage:
(1–14 days).
Optional test dose: 0.1 mg/kg to a maximum
Low doses (10 mg/kg) have been reported to
of 1 mg IV over 20–60 minutes.
control bleeding in patients with thrombo­
cytopenia for long periods of time (i.e., Initial dose: 0.5–1.0 mg/kg/day IV.
weeks to months). The daily dose is increased by 0.25 mg/kg
Continuous IV infusion: Loading dose of until the desired daily dose is reached. In
100 mg/kg, then 10–33 mg/kg/hour in 5% critically ill patients, rapid escalation of dos­
dextrose in water (maximum 1.25 g/hour). ing may be needed.
Empiric dose is 0.6 mg/kg/day IV.
Notes: Therapeutic dose for confirmed invasive
May accumulate in patients with decreased fungal infection is 0.5–1 mg/kg/day IV.
renal function and require decreased dos­ Daily infusion is over 2–6 hours, depending
ing. Avoid in patients with disseminated on the infusion tolerability. Every other day
intravascular coagulation or upper uri­ dosing can be given at 1.5 mg/kg/dose. Salt
nary tract bleeding (hematuria) due to loading with 10–15 ml/kg of normal saline
increased risk of forming clots in the col­ prior to each infusion may prevent hypoka­
lecting system. Increased risk of thrombo­ lemia and nephrotoxicity. Premedication is
sis with oral contraceptives, estrogens, frequently needed with acetaminophen and
and factor IX or prothrombin complex diphenhydramine 30 minutes prior to dose
concentrates (PCC). May cause nausea, (with repeat 4 hours later, if necessary).
diarrhea, malaise, headache, decreased Hydrocortisone (1 mg/mg of amphotericin,
platelet function, and false increase in uri­ maximum 25 mg) may be added to the bot­
nary amino acids. tle to possibly prevent immediate reactions.
Formulary 413

If the patient experiences rigors, meperidine AMPHOTERICIN B, LIPOSOMAL


hydrochloride may be given. Once therapy AmBisome
has been established, alternate‐day dosing Polyene antifungal
may be administered at a dose of 1–1.5 mg/ Injection: 50 mg vial
kg/IV every other day. For obese patients,
use total body weight for dose calculation. Pregnancy category B

Notes: Indications:
Administration of other nephrotoxic drugs Treatment of systemic or invasive fungal
may result in synergistic toxicity and should infection. Cerebrospinal fluid concentrations
be avoided if possible. Monitor daily electro­ are higher than with other amphotericin
lytes, renal and hepatic studies, and urine products as well as higher concentrations in
output. Common metabolic abnormalities the liver and spleen than with conventional
include hypokalemia, hypomagnesemia, amphotericin B.
and hypocalcemia. Other problems that
may occur are thrombocytopenia, hypergly­
Dosage:
cemia, diarrhea, dyspnea, back pain, and
increases in transaminases and bilirubin. Systemic fungal infections: 3–5 mg/kg/once
Common infusion‐related toxicities are fever, daily IV over 2 hours (may be reduced to a
chills, rigors, nausea, vomiting, hypoten­ 1‐hour infusion if well‐tolerated). Doses as
sion, and headache. high as 10 mg/kg/day have been used in
patients with Aspergillus sp.
AMPHOTERICIN B LIPID COMPLEX Empiric therapy for fever and neutropenia:
(ABLC) 3 mg/kg IV once daily.
Abelcet, ABLC Infusion may be shortened to 1 hour if
Polyene antifungal well‐tolerated.
Injection: 5 mg/ml (10, 20 ml)
Notes:
Pregnancy category B
See Amphotericin B.
Indications:
Treatment of aspergillosis or invasive fungal ANTI‐THYMOCYTE GLOBULIN (ATG)
infection. Higher concentrations are achieved Thymoglobulin (rabbit ATG, rATG,
in the spleen, lung, and liver, and therefore Genzyme)
may be more beneficial in the treatment of Atgam (equine ATG, eATG, Pfizer)
hepatosplenic candidiasis. Cerebrospinal Injection: 50 mg/ml
fluid (CSF) levels may be lower than with
Pregnancy category C
amphotericin B or the liposomal compound.

Indications:
Dosage:
Anti‐thymocyte globulin is an infusion of
Usual dose is 2.5–5 mg/kg IV once daily rabbit‐ or horse‐derived antibodies against
over 2 hours. Rate should not exceed 2.5 mg/ human T‐cells, which is used in the preven­
kg/hour. tion and treatment of acute rejection in
organ or hematopoietic transplantation and
Notes: therapy of aplastic anemia and refractory
See Amphotericin B. histiocytic disorders.
414 Formulary

Dosage: where intensive life support facilities are


Aplastic anemia: rATG 3.5 mg/kg/day IV on immediately available and with a physician
days 1–5 or hATG 40 mg/kg/day IV on days familiar with the treatment of potentially
1–4. Cyclosporine is given following the last life‐threatening allergic reactions in attend­
day of hATG or rATG infusion and adjusted ance. A systemic reaction such as a general­
to maintain a serum level of 200–400 ng/ml ized rash, tachycardia, dyspnea, hypotension,
or based on renal toxicity or tolerability. or anaphylaxis precludes any additional
Patients typically receive corticosteroids, administration of ATG.
usually methylprednisolone, at a dose of ATG administration very substantially
1 mg/kg per day, from day 1 until day 14; the reduces immune competence in patients with
dose is thereafter tapered down to prevent normal immune systems. rATG in particular
serum sickness. affects large reductions (through cell lysis) in
Skin testing before the first infusion is rec­ the number of circulating T‐lymphocytes,
ommended. Patients are tested with an hence preventing (or at least delaying) the cel­
intradermal injection of 0.1 ml of a 1:1000 lular rejection of transplanted organs or
dilution of ATG in sodium chloride injec­ hematopoietic cells. In the United States, it is
tion with a contralateral sodium chloride frequently given at the time of the transplant
injection control. The patient, and specifi­ to prevent graft‐versus‐host disease (GVHD).
cally the skin test site, should be observed ATG use can induce cytokine release syn­
every 15–20 minutes over the first hour after drome (CRS), and has been thought to
intradermal injection. A local reaction of increase the risk of posttransplant lymphopro­
10 mm or greater with a wheal or erythema, liferative disorder (PTLD); however, this asso­
or both, with or without pseudopod forma­ ciation may not apply when lower‐dosing
tion, and itching or a marked local swelling regimens are used. There is some evidence to
should be considered a positive test. suggest that inducing immunosuppression
with rATG at organ transplantation may cre­
Notes: ate conditions in the patient’s immune system
favorable to the development of immunologi­
Thymoglobulin and Atgam are currently
cal tolerance, but the exact basis for such a
licensed for use in the treatment of renal
development remains largely speculative.
allograft rejection; Atgam is additionally
Temporary depletion of the T‐cell population
licensed for use in the treatment of aplastic
at the time of the transplant also risks delayed
anemia. Both drugs are used in a number of
acute rejection, which may be missed and
off‐label applications.
cause severe damage to the graft.
The predictive value of the skin test has not
been proved clinically. Allergic reactions
such as anaphylaxis have occurred in APREPITANT
patients whose skin test is negative. In the
Emend
presence of a locally positive skin test to
Substance P/neurokinin 1 (NK1) receptor
ATG, serious consideration to alternative
antagonist
forms of therapy should be given. The risk
Solution: 125 mg
to benefit ratio must be carefully weighed. If
Capsules: 40, 80, 125 mg
therapy with ATG is deemed appropriate
Injection (fosaprepitant): 150 mg
following a locally positive skin test, treat­
ment should be administered in a setting Pregnancy category B
Formulary 415

Indications: regular dosing recommendations. Pediatrics:


Aprepitant is indicated, in combination with Administer 50% of the recommended dexa­
other antiemetic agents, in patients 6 methasone dose and regular 5‐HT3 dosing.
months of age and older for prevention of Moderately emetogenic regimen: Adults:
acute and delayed chemotherapy‐induced Dexamethasone 12 mg on day 1. Pediatrics:
nausea and vomiting (CINV) associated Administer 50% of the recommended dexa­
with initial and repeat courses of highly methasone dose and regular 5‐HT3 dosing.
emetogenic drugs including high‐dose cis­
platin and CINV associated with initial and Notes:
repeat courses of moderately emetogenic
Oral capsules and suspension can be admin­
cancer chemotherapy.
istered with or without food. Swallow cap­
Aprepitant is also indicated for prevention sules whole. Known hypersensitivity is a
of postoperative nausea and vomiting contraindication.
(PONV) in adults. It has not been studied
for treatment of established nausea and Aprepitant is a substrate, weak‐to‐moderate
vomiting. Chronic continuous administra­ (dose‐dependent) inhibitor, and an inducer
tion of aprepitant is not recommended. of CYP3A4. Concomitant administration of
the oral or IV form (fosaprepitant is rapidly
Dosage: converted to aprepitant) may result in
increased plasma concentrations of the con­
Prevention of CINV in adults and pediatric
comitant drug.
patients 12 years of age and older: 125 mg cap­
sule PO on day 1 and 80 mg capsule on days 2 Warfarin is a CYP2C9 substrate and con­
and 3. Fosaprepitant (prodrug), 115 mg IV comitant use may result in a decreased inter­
infused over 15 minutes, can be substituted national normalized ratio (INR); monitor
for 125 mg oral aprepitant on day 1 only. The INR over a 2‐week period, particularly at
oral suspension is available for pediatric 7–10 days, following initiation of aprepitant.
patients 6 months to less than 12 years of age There is concern for increased neurotoxicity
or pediatric and adult patients unable to with aprepitant and certain chemotherapeu­
swallow capsules. Administer 1 hour prior to tics including vincristine and ifosfamide.
chemotherapy on days 1, 2, and 3. If no Use with caution, consider prophylaxis with
chemotherapy is given on days 2 and 3, thiamine with ifosfamide.
administer in the morning. Adverse reactions may include:
Patients aged 6 months to <12 years:
Adults (≥3%): fatigue, diarrhea, asthenia,
Day 1: 3 mg/kg or 72 mg/m2 PO, maximum dyspepsia, abdominal pain, hiccups, white
125 mg. blood cell count decreased, dehydration,
Days 2 and 3: 2 mg/kg or 46 mg/m2, maxi­ and alanine aminotransferase increased.
mum 80 mg. Pediatrics (≥3%): neutropenia, headache,
Concomitant administration of dexametha­ diarrhea, decreased appetite, cough, fatigue,
sone and 5‐HT3 antagonist is recommended hemoglobin decreased, dizziness, and
as follows: hiccups.

Highly emetogenic regimen: Adults: Aprepitant does not affect the QTc interval.
Dexamethasone 12 mg on day 1; 8 mg on Aprepitant does not affect the pharmacokinet­
days 2, 3, and 4 with 5‐HT3 antagonist per ics of 5‐HT3 inhibitors such as ondansetron or
416 Formulary

granisetron. Corticosteroids: Dexamethasone refractory to, or have relapsed from, retinoid


and methylprednisolone are CYP3A4 and anthracycline chemotherapy, and whose
­substrates, and concomitant use with aprepi­ APL is characterized by the presence of the
tant leads to an increased area under the curve t(15; 17) translocation or PML/RAR‐alpha
(AUC) for these drugs, therefore doses should gene expression.
be reduced by approximately 50% when co‐
administered with a regimen of fosaprepitant Dosage:
followed by aprepitant, to achieve exposures of Newly diagnosed low‐risk APL:
dexamethasone similar to those obtained
Induction: 0.15 mg/kg intravenously daily in
when dexamethasone is given without aprepi­
combination with tretinoin until bone mar­
tant. The daily dose of dexamethasone admin­
row remission. Do not exceed 60 days of
istered in clinical CINV studies with oral
induction.
aprepitant reflects an approximate 50% reduc­
tion of the dose of dexamethasone. Consolidation: 0.15 mg/kg intravenously
daily 5 days per week during weeks 1–4 of
The co‐administration of fosaprepitant or
an 8‐week cycle for a total of four cycles in
aprepitant may reduce the efficacy of hor­
combination with tretinoin.
monal contraceptives during and for 28 days
after administration of the last dose of Relapsed or refractory APL:
either. Alternative or backup methods of Induction: 0.15 mg/kg intravenously daily
contraception should be used during treat­ until bone marrow remission. Do not exceed
ment with fosaprepitant or aprepitant and 60 doses for total induction.
for 1 month following the last dose. Consolidation: 0.15 mg/kg intravenously
Concomitant administration of fosaprepi­ daily for 25 doses over a period up to 5 weeks.
tant or aprepitant with strong CYP3A4
inhibitors (e.g., ketoconazole, itraconazole, Notes:
nefazodone, troleandomycin, clarithromy­ Refer to specific protocol for dosing infor­
cin, ritonavir, nelfinavir) should be mation and modifications. Do not adminis­
approached with caution. Co‐administra­ ter to patients with known hypersensitivity
tion of fosaprepitant or aprepitant with to ATO. The most common adverse reac­
drugs that strongly induce CYP3A4 activity tions (>30%) are leukocytosis, neutropenia,
(e.g., rifampin, carbamazepine, phenytoin) thrombocytopenia, nausea, vomiting, diar­
may result in reduced plasma concentra­ rhea, abdominal pain, hepatic toxicity,
tions and decreased efficacy. fever, rigors, fatigue, insomnia, tachycardia,
QTc prolongation, edema, hyperglycemia,
ARSENIC TRIOXIDE (ATO) hypokalemia, hypomagnesemia, dyspnea,
Trisenox cough, rash or itching, sore throat, arthral­
Injection: 12 mg in 6 ml vials gia, headaches, paresthesia, and dizziness.
Patients treated with ATO have experienced
Pregnancy category D
differentiation syndrome and cardiac con­
duction abnormalities. Symptoms of differ­
Indications: entiation syndrome, which can be fatal if not
Treatment of adults with newly diagnosed treated, may include fever, dyspnea, acute
low‐risk acute promyelocytic leukemia respiratory distress, pulmonary infiltrates,
(APL), as well as induction of remission and pleural or pericardial effusions, weight gain
consolidation in patients with APL who are or peripheral edema, hypotension, and renal,
Formulary 417

hepatic, or multi‐organ dysfunction, in the protocols may utilize different frequency or


presence or absence of leukocytosis. If differ­ patient‐specific dosing.
entiation syndrome is suspected, immedi­ In the setting of hypersensitivity, the replace­
ately initiate high‐dose corticosteroid therapy ment dose for Erwinia asparaginase is
and hemodynamic monitoring until resolu­ 25,000 IU/m2/dose IM or IV, given three times
tion of signs and symptoms. Temporary dis­ per week (every 48–72 hours or Monday–
continuation of ATO may be required. ATO Wednesday–Friday) for six doses to replace
can cause QTc interval prolongation, com­ one dose of pegaspargase. IV administration
plete atrioventricular block, and a torsade de may result in shorter duration of serum aspar­
pointes‐type ventricular arrhythmia, which aginase activity levels compared to IM admin­
can be fatal. Before initiating therapy, assess istration. When administered IM, maximum
the QTc interval, correct pre‐existing electro­ volume is 2 ml per injection site. Many
lyte abnormalities, and consider discontinu­ patients require multiple injections.
ing drugs known to prolong QTc interval. Do
Patients should be observed in a clinic or
not administer to patients with ventricular
hospital setting for at 1–2 hours after admin­
arrhythmia or prolonged QTc interval.
istration to monitor for a hypersensitivity
reaction. Appropriate agents for treatment
ASPARAGINASE of hypersensitivity should be readily availa­
PEG‐aspargase (Escherichia coli asparagi­ ble (oxygen, epinephrine, antihistamines,
nase, Pegaspargase, Oncaspar), Asparaginase intravenous steroids) in addition to resusci­
Erwinia chrysanthemi (Erwinaze) tative equipment.
Antineoplastic, results in asparagine deple­
tion in malignant cells
Injection: Notes:
10,000 unit vial (Erwinaze) Do not give any form of asparaginase to
3750 unit vial (Oncaspar) patients with prior life‐threatening throm­
Pregnancy category C bosis or hemorrhage or significant pan­
creatitis (grade 3 or 4) associated with
Indications: asparaginase.
Treatment of acute lymphoblastic leukemia Hypersensitivity to Oncaspar pegaspargase
(ALL). Current Children’s Oncology Group necessitates a switch to Erwinaze. Some
(COG) trials have adopted pegaspargase as the centers may consider desensitization or re‐
preparation of choice secondary to a longer challenge, though this should be done with
half‐life compared to the prior native E. coli caution. Use with caution in patients with
asparaginase. Erwinia‐derived asparaginase is hepatic impairment or in those receiving
FDA approved in the setting of hypersensitiv­ other hepatotoxic drugs as well as in those
ity to E. coli‐derived asparaginase. receiving anticoagulation or nonsteroidal
anti‐inflammatory agents (NSAIDs).
Asparaginase may cause hyperglycemia,
Dosage:
hyperuricemia, hyperammonemia, hypofi­
Refer to individual protocol. brinogenemia, thrombosis, hemorrhage,
Usual dose in ALL of pegaspargase is anaphylaxis, and hemorrhagic cystitis.
2500 IU/m2 IV over 1–2 hours every 2–4 Dose modifications related to hyperglyce­
weeks (Induction, Delayed Intensification, mia or hyperlipidemia are not recom­
and Interim Maintenance phases). Other mended; treat underlying conditions as
418 Formulary

medically indicated. Dexamethasone may baseline hematology values, is 75 mg/m2


alleviate allergic symptoms. daily for 7 days by SC or IV infusion.
Premedication for hypersensitivity is not Premedicate for nausea and vomiting.
recommended though is given at some insti­ Repeat cycles every 4 weeks or per protocol.
tutions. In the event of hypersensitivity dur­ After two cycles, may increase dose to
ing a pegaspargase infusion, substitution 100 mg/m2 if no beneficial effect is seen and
with Erwinia asparaginase should begin no toxicity other than nausea and vomiting
within 48 hours in patients who are clini­ has occurred.
cally stable. Up to six doses of Erwinia
asparginase may be given to replace the Notes:
incomplete administration of the IV pegas­ Azacitidine is contraindicated in patients
pargase dose. Erwinia doses are based on with prior hypersensitivity to this drug
nadir serum asparaginase activity detected or mannitol, or advanced liver tumors.
at 48 and 72 hours after dosing. May cause cytopenias; monitor complete
Asparaginase activity assays are commer­ blood count (CBC), liver, and kidney
cially available to determine activity, length function. Patients with severe preexisting
of asparagine depletion, and evaluate for the hepatic impairment are at higher risk for
presence of neutralizing antibodies. Some toxicity. Azacitidine and its metabolites
clinicians/treatment centers may elect to dis­ are primarily excreted by the kidneys,
continue pegaspargase and switch to Erwinia and patients with renal insufficiency
asparaginase based on lab evidence of silent should be monitored for increased risk of
antibodies or decreased asparaginase activity toxicity. The most common adverse reac­
suggesting silent inactivation in the absence tions by the SC route are: nausea, anemia,
of overt hypersensitivity. Similarly, patients thrombocytopenia, vomiting, pyrexia,
with a clinical hypersensitivity reaction but leukopenia, diarrhea, injection site ery­
without evidence of antibody development thema, constipation, neutropenia, and
can be considered for re‐challenge. ecchymosis. The most common adverse
reactions by the IV route also include
AZACITIDINE petechiae, rigors, weakness, and
hypokalemia
Vidaza
Nucleoside metabolic inhibitor; DNA hypo­
methylating agent BRENTUXIMAB VEDOTIN
Injection: IV, SC; 100 mg single dose vials Adcetris
Pregnancy category D Antibody–drug conjugate
Injection: 50 mg vials, 5 mg/ml solution
Indications: Pregnancy category D
Treatment of acute myelogenous leukemia
(AML) and myelodysplastic syndrome Indications:
(MDS). Being studied in infant ALL with Treatment of relapsed or refractory Hodgkin
hypermethylating KMT2A mutation as well lymphoma (HL) and systemic anaplastic large
as posttransplant in AML patients. cell lymphoma (ALCL). It selectively targets
tumor cells expressing the CD30 antigen, a
Dosage: defining marker of Hodgkin lymphoma
The recommended starting dose for the first and ALCL (a type of T‐cell non‐Hodgkin
treatment cycle, for all patients regardless of lymphoma)
Formulary 419

Dosage: impairment in wound healing has not been


1.2–1.8 mg/kg (maximum 120–180 mg) 30‐ determined. General recommendation is to
minute IV infusion every 2–3 weeks. allow 28 days prior to and following major
surgery. Use with caution in patients with
Notes: thrombocytopenia. An increased risk of
thromboembolic events has been reported
Brentuximab vedotin had been linked with
in combination regimens. May impair fertil­
two cases of progressive multifocal leukoen­
ity and have adverse effects on fetal develop­
cephalopathy, requiring the addition of
ment; adequate contraception must be used
a black box warning to the drug label regard­
during therapy. May cause infusional toxic­
ing this potential risk. Pneumonitis has also
ity (discontinue infusion until symptoms
been reported. Common adverse events
abate) or proteinuria. May potentiate car­
include cytopenias, nausea and vomiting,
diac effects of anthracyclines. Discontinue
constipation, decreased appetite, weight loss,
therapy in patients who develop fistulas,
abdominal pain, fever, as well as peripheral
hypertensive crises, encephalopathy, or
sensory neuropathy. Pediatric trials down to
nephrotic syndrome. Common adverse
2 years of age are ongoing.
effects include epistaxis, rhinitis, dry eye,
altered taste, headache, hypertension, and
BEVACIZUMAB rectal bleeding.
Avastin
Angiogenesis inhibitor; monoclonal anti­ BLEOMYCIN SULFATE
body to vascular endothelial growth factor
Blenoxane, generic
(VEGF)
Antineoplastic antibiotic
Injection: 25 mg/ml
Injection: 15 unit vial (1 unit = 1 mg)
Pregnancy category C
Pregnancy category D
Indications:
Indications:
Treatment of metastatic colon cancer, non‐
Treatment of Hodgkin and non‐Hodgkin
small cell lung cancer, metastatic renal cell
lymphoma, squamous cell carcinoma, tes­
carcinoma and recurrent glioblastoma mul­
ticular carcinoma, and germ cell tumor. Used
tiforme. Currently being investigated in
as a sclerosing agent to control malignant
pediatric protocols for the treatment of
pleural effusions.
brain tumors, refractory or recurrent solid
tumors, and neurofibromatosis.
Dosage:
Dosage: Refer to individual protocol.
Refer to individual protocol. 10–20 IU/m2 IV infusion over 10 minutes
5–15 mg/kg q2–3 weeks as a single agent or (may be given IM or SC) one to two times
in combination regimens. per week per protocol.

Notes: Notes:
Avoid use in patients with recent surgery, Do not give to patients with known
hemoptysis, gastrointestinal or central nerv­ hypersensitivity to bleomycin.
ous system bleeding, or any other serious Premedication with acetaminophen,
bleeding. The interval required between hydrocortisone, and antihistamine may
surgery and drug administration to avoid decrease infusional toxicity. Dose may
420 Formulary

need to be modified for renal or pulmo­ Notes:


nary toxicity. Monitor renal function stud­ Blinatumomab is contraindicated in patients
ies and pulmonary function, including with known hypersensitivity. Common
pulmonary diffusion capacity for carbon adverse effects include pyrexia, headache,
monoxide (DLCO). If symptoms of pneu­ peripheral edema, fever, nausea, hypoka­
monitis appear (i.e., fine rales, decreased lemia, and constipation. Severe effects
DLCO), assess CXR and pulmonary func­ include fever with neutropenia, pneumonia,
tion. Pulmonary fibrosis is a rare and sepsis, confusion, and encephalopathy.
potentially fatal complication. Direct instil­
Black box warnings exist for CRS and severe
lation into the pleural space may be used to
neurologic toxicity. Symptoms of CRS include
control malignant effusion.
pyrexia, hypotension, increased alanine ami­
notransferase and aspartate aminotransferase,
BLINATUMOMAB increased total bilirubin, headache, and nausea.
BLINCYTO Evidence of capillary leak syndrome and dis­
Antineoplastic, monoclonal antibody, seminated intravascular coagulation (DIC) may
bispecific T‐cell engager (BiTE) be seen rarely. Symptoms of CRS warrant imme­
Injection: 35 mcg/vial (includes IV solution diate cessation of the infusion and ­supportive
stabilizer) care as medically indicated. Approximately 50%
of patients may experience neurologic toxicity
Pregnancy category C with a median time of onset of 7 days, with 15%
resulting in grade 3 or higher (encephalopathy,
Indications: Treatment of Philadelphia chro­ convulsions, speech changes, altered conscious­
mosome‐negative relapsed or refractory ness, confusion or disorientation, or coordina­
CD19+ B‐cell precursor ALL. tion or balance disorders). The majority of
events resolve with interruption of blinatu­
momab, often require dose reduction and may
Dosage:
lead to treatment discontinuation.
Refer to individual protocol.
Given as a continuous infusion, generally for BUSULFAN
28 days. Goal dosing is 15 mcg/m2/day, as tol­ Busulfex, generic
erated. Often utilized as bridging therapy to Antineoplastic
hematopoietic stem cell transplant (HSCT) Tablet: 2 mg
in patients with refractory disease or low Injection: 6 mg/ml
minimal residual disease (MRD) positivity.
Pregnancy category D
Premedicate with dexamethasone (5 mg/m2;
per protocol) IV 30–60 minutes prior to
Indications:
start of infusion, change in dosing or when
restarting an infusion after an interruption Treatment of chronic myelogenous leukemia
of ≥4 hours. (CML) and for myeloablative conditioning
regimens prior to bone marrow or periph­
The blinatumomab solution for infusion must
eral blood stem cell transplantation.
be administered using IV tubing that contains
a sterile, non‐pyrogenic, low protein‐binding,
Dosage:
0.2 μm in‐line filter. Blinatumomab must be
changed every 48 hours to 7 days but can be Refer to individual protocol.
done as an outpatient infusion if tolerated in HSCT regimen: dose based on actual body
the initial week. weight (ABW); adjustments based on
Formulary 421

therapeutic drug monitoring per protocol Dosage:


and goal AUC). Refer to individual protocol. May require dose
>10 kg and ≤4 years: 4 mg/kg/dose IV daily calculation using the modified Calvert for­
over 4 consecutive days mula (total dose in mg) to achieve the appro­
<10 kg or ≥4 years: 3.2 mg/kg/dose IV daily priate AUC for concentration over time rather
over 4 consecutive days than dosing by body surface area (BSA).
Solid tumors: 400–560 mg/m2 every 3–4 weeks
Notes: or per protocol.
Do not give to patients with known hyper­ Brain tumor protocols:
sensitivity to busulfan; should not be used 175 mg/m2 weekly for 4 weeks, then 2‐week
in pregnancy or while nursing. May cause recovery; alternatively 540 mg/m2 q month.
severe bone marrow suppression, sinusoi­ Dose is adjusted based on suppression of neu­
dal obstructive syndrome (SOS) (hepatic trophil and platelet counts or renal toxicity.
veno‐occlusive disease [VOD]), or sei­
Bone marrow transplant preparative regi-
zures when given at high doses. Use with
men: 500 mg/m2/day for 3 days.
caution with other myelosuppressive
drugs or radiation. May cause hemor­
rhagic cystitis. Use with thioguanine may Notes:
increase hepatic toxicity. Patients should Do not give to patients with known hypersen­
be placed on a prophylactic antiepileptic sitivity to carboplatin, cisplatin, or other plati­
drug (i.e., Keppra, lorazepam) while num‐containing compounds. Anaphylaxis
receiving busulfan. may occur within minutes of administration.
Therapeutic drug monitoring is done with May cause hypotension, electrolyte abnor­
dose 1. To calculate the AUC, obtain levels malities (hypomagnesemia in particular),
before and after infusion, per protocol. nausea, hearing loss, and peripheral neuropa­
Metabolism of busulfan may be inhibited thy. Highly emetogenic; increasing the length
by antifungal agents (azoles), CYP3A4 of infusion or giving smaller doses on con­
inhibitors, dasatinib, and metronidazole secutive days may alleviate the severity. Severe
leading to increased levels and effect. marrow suppression may occur, and the
Busulfan metabolism may be potentiated postchemo nadir may be delayed. Anemia
by CYP3A4 inducers, deferasirox, and occurs from cumulative effects and transfu­
Echinacea, leading to decreased levels sion is likely with high or repetitive dosing.
and effect. Reduce dose in impaired renal function (cre­
atinine clearance <60 ml/minute) utilizing the
modified Calvert formula. Aminoglycosides
CARBOPLATIN may increase serum levels/effects of carbopl­
Paraplatin, generic atin and augment ototoxicity and nephrotox­
Antineoplastic, alkylating agent icity; nephrotoxic drugs may increase renal
Injection: 10 mg/ml; 50, 150, 450, 600 mg vials toxicity of carboplatin. Children are particu­
larly susceptible to ototoxicity and care should
Pregnancy category D be taken to avoid concomitant drugs with oto­
toxic potential. Recent study has shown oto­
Indications: protection with sodium thiosulfate; concern
Treatment of pediatric brain tumors, neuro­ still remains on worse outcomes in patients
blastoma, testicular tumors, relapsed leuke­ with high‐risk diseases. Avoid concomitant
mia, and solid tumors. administration of topotecan and taxanes.
422 Formulary

All patients should receive hydration prior CASPOFUNGIN


to and following administration with Cancidas
sodium‐chloride‐containing solution, with Antifungal agent, echinocandin
or without mannitol or furosemide, to Injection: 50, 70 mg
ensure good urine output and decrease risk
of nephrotoxicity. Reduce dose for young Pregnancy category C
children (<6 months) and with renal impair­
ment. Electrolytes and magnesium should Indications:
be monitored. Acute leukemia has been Treatment of candidemia, candidal intra‐
reported as a second malignant neoplasm. abdominal abscess, or esophageal candidia­
sis; empiric therapy of presumed fungal
CARMUSTINE infection in febrile neutropenic patients.
BCNU
BiCNU, Gliadel Dosage:
Antineoplastic, alkylating agent Preterm neonates to infants <3 months:
Injection: 100 mg vial 25 mg/m2/dose IV once daily.
Implant: Gliadel wafer Infants ≥3 months to adults: 70 mg/m2 loading
Pregnancy category D dose IV over 1 hour (maximum dose 70 mg),
followed by 50 mg/m2/dose IV once daily
Indications: over 1 hour (maximum dose 50–70 mg/day).
Treatment of brain tumors and Hodgkin Patients may benefit from increased daily
and non‐Hodgkin lymphoma. Wafer dosing to 70 mg/m2 once daily dependent on
implant (Gliadel) may be an adjunct to sur­ clinical status and response.
gery and radiation for glioblastoma multi­ Empiric therapy should be given until neu­
forme and high‐grade glioma. tropenia resolves. In neutropenic patients,
continue treatment for at least 7 days after
Dosage: signs and symptoms of infection and neu­
Refer to individual protocol. tropenia have resolved and at least 14 days
Typical dose may be 150–200 mg/m2/dose following a positive culture in patients with
IV every 6 weeks. documented fungal infection.
Bone marrow transplant: 300 mg/m2 gener­
Notes:
ally given as one dose as part of BEAM
(BCNU, etoposide, cytarabine, melphalan) Modify dose in patients with hepatic impair­
conditioning regimen. ment. Drug interactions exist with cyclo­
sporine, tacrolimus, and rifampin and may
Notes: require dose modification.
Do not give to patients with known hyper­
sensitivity to carmustine. Severe, prolonged CISPLATIN
(4–6 weeks) marrow suppression may
Platinol, generic
necessitate change in dosing for subsequent
Antineoplastic, alkylating agent
cycles. Toxicity is cumulative and delayed
Injection: 1 mg/ml; 50, 100, 150 mg vials
pulmonary fibrosis may occur in patients
receiving very high doses (7 701 800 mg/m2). Pregnancy category D
Formulary 423

Indications: severe hearing loss include pediatric age


Treatment of soft tissue sarcoma, osteosar­ (especially <5 years), concurrent or prior
coma, Hodgkin and non‐Hodgkin lym­ cranial radiation, renal impairment, and
phoma, brain tumors, metastatic ovarian concomitant use of other ototoxic drugs.
and testicular tumors, germ cell tumors, and Ototoxicity in the pediatric population is
neuroblastoma. common, occurring in 40–60%, and war­
rants frequent and care audiologic evalua­
Dosage: tions at baseline, prior to each dose, and for
several years after treatment. Sodium thio­
Refer to individual protocol. Verify any dos­
sulfate has been shown to be otoprotective,
ing schedule in which the cisplatin dose
though concerns remain regarding worse
exceeds 120 mg/m2 per course. Doses are
outcomes in high‐risk patients.
lower when used in combination protocols.
All patients should receive hydration prior to
Daily dosing schedule: 15–20 mg/m2/day for
and for 24 hours after administration with a
5 days every 3–4 weeks.
sodium‐chloride‐containing solution (with
Osteosarcoma and neuroblastoma: 60–100 or without mannitol or furosemide), to
mg/m2 once every 3–4 weeks. ensure good urine output and decrease risk
The rate of intravenous infusion is dose‐ of nephrotoxicity. Reduce dose for young
dependent and ranges from a 15–20‐minute children (<6 months) and with renal impair­
infusion to a 6–8‐hour infusion; 24‐hour ment. Electrolytes and magnesium should be
continuous infusions are also used. monitored. Acute leukemia has been
reported as a second malignant neoplasm.
Notes:
Do not give to patients with known hyper­
CLADRIBINE
sensitivity to cisplatin or platinum‐contain­
ing compounds, preexisting renal 2‐chlorodeoxyadenosine, 2‐CdA
impairment, hearing impairment, and mye­ LeustatinTM, generic
losuppression. Increased risk of nephrotox­ Anti‐neoplastic, anti‐metabolite; purine analog
icity when given with other nephrotoxic Injection: single dose 10 mg vials (1 mg/ml)
drugs (aminoglycosides and amphotericin Pregnancy category D
B). Reduces renal elimination of methotrex­
ate. Cisplatin may increase the levels/effects
Indications:
of aminoglycosides, taxane derivatives,
topotecan, and vinorelbine. Effects or levels Cladribine is highly effective in the treat­
of cisplatin may be increased with concomi­ ment of Langerhans cell histiocytosis and
tant administration of loop diuretics. rare histiocytic disorders of the juvenile
Electrolyte abnormalities are common and xanthogranuloma group.
include hypomagnesemia, hypokalemia, 2‐CdA is an active salvage monotherapy for
hyponatremia, and hypophosphatemia. patients with risk‐organ involvement refrac­
Cisplatin is highly emetogenic and com­ tory to initial therapy or patients with recur­
monly has delayed nausea and vomiting. rent, low‐risk Langerhans Cell Histiocytosis
Potential toxicities include peripheral neu­ (LCH) (i.e., patients with non‐risk‐organ
ropathy and hepatic and ocular toxicity. involvement, including multifocal bone dis­
Ototoxicity is common and risk factors for ease). Cladribine appears to be more effective
424 Formulary

in low‐risk patients or patients with multifo­ Notes:


cal bone disease. Patients older than 2 years Cytokine release may develop into a sys­
and those with a longer time between diag­ temic inflammatory response with resultant
nosis and 2‐CdA therapy have better capillary leak syndrome and organ failure. If
responses, presumably because their disease this occurs, discontinue clofarabine and ini­
is less aggressive. tiate therapy with diuretics, corticosteroids,
Combination therapy with 2‐chlorodeoxy­ and albumin. If hypotension resolves with­
adenosine and cytarabine (Ara‐C) has been out pharmacologic intervention, clofarabine
studied in patients with refractory, risk‐ may be resumed. The risk for hepatic toxic­
organ–positive LCH and the combination ity and veno‐occlusive disease is increased
regimen appears to be an effective therapy in patients who have previously undergone
for refractory multisystem LCH. HSCT. Avoid concomitant use of nephro­
2‐CdA may be effective in the treatment of toxic and hepatotoxic drugs.
LCH with CNS involvement.
2‐CdA is also used in adults with hairy cell CYCLOPHOSPHAMIDE
leukemia or multiple sclerosis. Generic
Antineoplastic, alkylating agent
Dosage: Tablets: 25, 50 mg
Injection: 500 mg, 1 g, 2 g vials
Safety and effectiveness in pediatric patients
have not been established. Patients 1–21 years Pregnancy category D
age have received doses ranging from 3 to
10.7 mg/m2/day for 5 consecutive days. See Indications:
protocols for specific dosing regimen.
Treatment of Hodgkin and non‐Hodgkin lym­
phoma, acute and chronic leukemia, retinoblas­
Notes:
toma, mycosis fungoides, and neuroblastoma.
Adverse effects include severe bone marrow Conditioning therapy for bone marrow trans­
suppression and resultant cytopenias, fever, plantation (BMT) and posttransplant after
and neurotoxicity manifested as peripheral haplo‐identical transplantation. Treatment of
neuropathy or pain. Fatal bacterial and fun­ nephrotic syndrome, systemic lupus erythema­
gal infections have followed drug adminis­ tosus, and other rheumatic diseases.
tration. Limited pediatric data are available.
Dosage:
CLOFARABINE Refer to individual protocol.
Clolar Acute lymphoblastic leukemia: 1000–1200
Antineoplastic, antimetabolite mg/m2/dose IV, in Consolidation and
Injection: 1 mg/ml Delayed Intensification phases.
Pregnancy category D Bone marrow transplant conditioning: 50 mg/
kg/day IV for 3–4 days.
Indications: Nephrotic syndrome: 2 mg/kg/day PO for
Treatment of relapsed or refractory ALL, 8–12 weeks; maximum cumulative dose
AML, MDS. 168 mg/kg.
Systemic lupus erythematosus: 500–750 mg/
Dosage: m2 IV monthly; maximum dose 1 g/m2.
Children (≥1 year) to adults: 40 mg/m2/day Juvenile idiopathic arthritis/vasculitis: 10 mg/
for 5 days every 28 days or per protocol. kg IV every 2 weeks.
Formulary 425

Notes: Intravenous:
Do not give to patients with known hyper­ Initial: 5–6 mg/kg IV single dose, administered
sensitivity to cyclophosphamide. Dose may over 2–6 hours, beginning 4–12 hours pre­
need to be adjusted for myelosuppression or transplant. Continue same IV dose posttrans­
impaired renal function. Mesna should be plant until patient able to tolerate oral form.
given with high‐dose therapy (>1 g/m2/day) Conversion from IV to PO dose (1:3 ratio):
to reduce potential of hemorrhagic cystitis. Multiply total daily IV dose by 3 and admin­
Allopurinol may increase the myelotoxicity ister in two divided oral doses per day.
of cyclophosphamide by inhibiting its
metabolism. Ensure aggressive hydration Notes:
with sodium‐chloride‐containing fluids and Do not give to patients with known hyper­
frequent bladder emptying. High cumula­ sensitivity to cyclosporine (castor oil is an
tive doses may cause amenorrhea associated ingredient in the preparation). May cause
with decreased estrogen and elevated gon­ nephrotoxicity, hepatotoxicity, hypomagne­
adotropin levels in females and azoospermia/ semia, hyperkalemia, hyperuricemia, hyper­
infertility in males. Some reversibility of tension, hirsutism, acne, gastrointestinal
infertility may occur after cessation of symptoms, tremor, leukopenia, headache,
therapy. and gingival hyperplasia. Use with caution
with concomitant administration of other
CYCLOSPORINE nephrotoxic drugs (e.g., amphotericin B, ami­
Sandimmune, Neoral, Gengraf, generics noglycosides, tacrolimus, acyclovir, NSAIDs).
Immunosuppressant Requires close monitoring of renal and hepatic
Capsules: 25, 50, 100 mg function and frequent determination of trough
Solution: 100 mg/ml levels (drawn just prior to dose at steady state).
Injection: 50 mg/ml Cyclosporine is a substrate for the cytochrome
Pregnancy category C P450 3A4 oxidase system. Drug interactions
include ketoconazole, itraconazole, flucona­
Indications: zole, erythromycin, and methylprednisolone,
which increase the cyclosporine concen­
Used with corticosteroids to prolong organ
tration by inhibiting hepatic metabolism.
and patient survival in kidney, liver, heart,
Refer to the Physicians’ Desk Reference
and bone marrow transplants; treatment of
(PDR) for more extensive drug interaction
aplastic anemia and other bone marrow fail­
information.
ure syndromes.

Dosage (Sandimmune): CYTARABINE HYDROCHLORIDE

Due to better absorption, lower doses of Ara‐C


Neoral and Gengraf may be required com­ Cytosar‐U, generic
pared to Sandimmune. Antineoplastic, antimetabolite; pyrimidine
antagonist
Oral: Injection: 100 mg/ml; 100 and 500 mg, 1 and
2 g vials
Initial: 15 mg/kg single oral dose, beginning
4–12 hours pretransplant. Pregnancy category D
Maintenance: 15 mg/kg/day PO divided q12–
24h for 1–2 weeks posttransplant, decrease Indications:
by 5% per week to 3–10 mg/kg/day divided Treatment of ALL and AML, refractory non‐
q12–24h. Hodgkin lymphoma, and may be used in
426 Formulary

conditioning regimens for BMT. Treatment administration of IV and IT therapy increases


and prophylaxis of CNS leukemia. risk of spinal cord toxicity.
When prepared for intrathecal use, added
Dosage: precautions should be taken when other
Refer to individual protocol. medications are also being delivered to
Infants <3 years: 3.3 mg/kg/day IV/SC for 4 ensure appropriate labeling and handling
days. such that only medications intended for
Children and adults: administration in the CNS are with the
patient for the procedure.
AML Induction: 100 mg/m2/24 hours for 7
days. Give IV q12h or as continuous
DACARBAZINE
infusion.
DTIC
High‐dose cytarabine: (AML, relapsed or
Antineoplastic
refractory ALL, non‐Hodgkin lymphoma)
Injection: 200 mg/20 ml
3 g/m2/dose q12h for up to 12 doses.
ALL (Delayed Intensification): 75 mg/m2/ Pregnancy category C
dose IV/SC daily for 4 consecutive days,
repeat in 7 days for total of two courses. Indications:
Intrathecal (IT) dosing (CNS treatment and Treatment of Hodgkin lymphoma and met­
prophylaxis): Dosing per age. May be com­ astatic malignant melanoma.
bined with other intrathecal agents such as
hydrocortisone and methotrexate per proto­ Dosage:
col (e.g., intrathecal double/triple). Refer to individual protocol.
Children <1 year: 20 mg. Metastatic malignant melanoma: 2–4.5 mg/
Children 1 to <2 years: 30 mg. kg/day IV for 10 days every 4 weeks or
250 mg/m2 IV for 5 days, every 3 weeks.
Children 2 to <3 years: 50 mg.
Hodgkin lymphoma: 375 mg/m2 IV on days 1
Children ≥3 years and adults: 70 mg.
and 15 of each course, every 4 weeks or
150 mg/m2 IV for 5 days every 4 weeks.
Notes:
Do not give to patients with known hyper­ Notes:
sensitivity to cytarabine. May need to reduce Do not give to patients with known hyper­
dose with myelosuppression or hepatic dys­ sensitivity to dacarbazine. Dosage reduction
function. Causes significant bone marrow may be necessary in patients with renal or
suppression. Particular risk of Strep viridans hepatic insufficiency. Drug extravasation
sepsis. High doses have been associated with may result in tissue damage and severe pain.
gastrointestinal, CNS, pulmonary, and ocular Nausea and vomiting are common; if severe
toxicities, as well as cardiomyopathy. May and protracted, treatment with phenobarbi­
cause nausea, vomiting, mucositis, fever, tal or prochlorperazine may be beneficial.
headache, somnolence, anorexia, alopecia, Hematopoietic toxicity primarily results in
conjunctivitis, ataxia, diarrhea, hepatic dys­ leukopenia and thrombocytopenia.
function, and peripheral neuropathy.
Prophylaxis with dexamethasone ophthalmic
drops may decrease conjunctivitis. Acute DACTINOMYCIN
neurotoxicity has been reported following Actinomycin D
intrathecal administration. Concurrent Cosmegen
Formulary 427

Antineoplastic antibiotic in immunocompromised hosts; treatment


Injection: 0.5 mg (500 mcg) vial of Toxoplasma gondii and Mycobacterium
leprae.
Pregnancy category D
Dosage:
Indications:
Children 1 month to <12 years: 2 mg/kg/day
Treatment of Wilms tumor, rhabdomyosar­
PO or 4 mg/kg/dose PO once weekly (maxi­
coma, Ewing sarcoma, ovarian germ cell tumor,
mum 100 mg/dose daily or 200 mg/dose
and gestational trophoblastic neoplasm.
weekly).
Children >12 years and adults: 100 mg/
Dosage:
day PO or 200 mg PO once weekly.
Refer to individual protocol. Note that medi-
cation orders for dactinomycin may be written
Notes:
in micrograms (mcg) or milligrams (mg).
Children ≥6 months to adult: 15 mcg/kg/ Do not give to patients with known hyper­
day IV or 400–600 mcg/m2/day IV once sensitivity to dapsone. Dapsone is a strong
daily for 5 days; cycles every 3–6 weeks. oxidizing agent and may cause hemolysis in
Higher doses are given in some protocols. susceptible individuals. Screening for glu­
cose‐6‐phosphate dehydrogenase (G6PD)
Notes: deficiency is suggested in high‐risk popula­
Do not give to patients with known hyper­ tions (e.g., Mediterranean) and should not be
sensitivity to dactinomycin. Avoid in infants used if positive. May be safe to use in the
<6 months of age due to increased adverse African‐American variant of G6PD defi­
events. Use with caution in patients with ciency, though some degree of hemolysis may
hepatobiliary dysfunction or who have occur. May also cause marrow suppression or
received radiation (radiation recall effect). methemoglobinemia. Drug interactions
Reduce dosage in patients receiving concur­ occur, primarily with rifampin. May be given
rent radiation. Gastrointestinal and marrow to immunosuppressed patients who cannot
suppressive effects are increased with con­ tolerate or are allergic to cotrimoxazole.
current radiation treatment. Avoid extravasa­
tion. May cause myelosuppression, anorexia, DARBEPOETIN ALFA
vomiting, diarrhea, stomatitis, hepatic toxic­ Aranesp
ity including veno‐occlusive disease, elevated Erythropoiesis stimulating protein
transaminases, hepatomegaly, and hepatitis. Injection: 25, 40, 60, 100, 200, 300 mcg/1 ml
(1 ml); 10 mcg/0.4 ml (0.4 ml)
DAPSONE
Pregnancy category C
Aczone, Diaminodiphenyl sulfone, DDS,
generics Indications:
Antibiotic
Anemia in chronic renal disease, chemo­
Tablet: 25, 100 mg
therapy‐induced anemia.
Oral suspension: 2 mg/ml
Pregnancy category C Dosage:
Renal failure on dialysis: 0.45 mcg/kg/dose IV/
Indications: SC once weekly or 0.75 mcg/kg/dose IV/SC
Prevention and treatment of Pneumocystis every 2 weeks. IV dose is recommended while
jiroveci pneumonia (PCP) (formerly carinii) on hemodialysis (infuse over 1–3 minutes).
428 Formulary

Renal failure not on dialysis: 0.45 mcg/kg/ to prior therapy with imatinib; and adults
dose IV/SC once every 4 weeks. with Ph+ ALL (off label use in pediatrics).
Adjust dosing after 4 weeks based on
response: increase by 25% for hgb rise Dosage:
<1 g/dl (do not adjust more than monthly); Refer to individual protocol.
decrease dose 25% if hgb ≥ 11 g/dl or Children: 60 mg/m2, do not crush or cut tab­
increases by >1 g/dl over a 2‐week period. lets. Take with or without food.
Chemotherapy‐induced anemia: 2.25 mcg/kg/ Adults:
dose SC once weekly.
CML, chronic phase: 100 mg PO QD.
Adjust dosing after 6 weeks: increase dose to
CML, accelerated or blast phase: 140 mg PO
4.5 mcg/kg/dose weekly SC/IV for <1 g/dl
QD.
increase in hgb or if hgb <10 g/dl; decrease
dose 40% if hgb increases >1 g/dl in any 2‐
week period or if hgb >12 g/dl. Discontinue Notes:
therapy if lack of response at 8 weeks or Dasatinib is contraindicated in patients with
chemotherapy completed. known hypersensitivity. It is metabolized via
the CYP450 enzyme. Common adverse
Notes: effects include myelosuppression, fluid
Monitor hemoglobin (hgb), blood pressure, retention, cardiac dysfunction and/or QTc
serum chemistries, and reticulocyte count prolongation, pulmonary arterial hyperten­
on treatment. Dose increases should not be sion, and severe mucocutaneous reactions.
made more frequently than once monthly. Avoid concomitant strong CYP3A4 induc­
Evaluate iron status (ferritin, serum iron, ers including St. John’s Wort and grapefruit
and total iron‐binding capacity [TIBC]); juice. Due to drug interactions avoid simul­
concurrent iron supplementation may be taneous administration of antacids, H2
necessary. Concern remains secondary to antagonists, and proton pump inhibitors.
meta‐analyses in adult oncology patients Severe bleeding has occurred and patients
which have shown increased morbidity with should have frequent monitoring of blood
erythropoietin utilization; should be used counts with platelet and red cell trans­
with caution, and potentially limited to fusions as clinically indicated. Avoid con­
patients aiming to minimize transfusion comitant use of drugs affecting platelet
(i.e., Jehovah’s Witness). function.

DASATINIB DAUNORUBICIN HYDROCHLORIDE


Sprycel Cerubidine
Kinase inhibitor Antineoplastic antibiotic, anthracycline
Tablet: 20, 40, 50, 70, 80, 100, 140 mg Injection: 2 mg/ml; 5, 25, 100 ml vials
Pregnancy category D Pregnancy category D

Indications: Indications:
Treatment of adult and pediatric patients Treatment of ALL, AML, Wilms tumor,
with Philadelphia positive (Ph+) CML in Hodgkin lymphoma, soft tissue and bone
chronic phase; adults with CML in acceler­ sarcomas, primary breast carcinoma, and
ated or blast phase unresponsive or refractory many other carcinomas and sarcomas.
Formulary 429

Dosage: text (see doxorubicin). Secondary AML and


Refer to individual protocol. MDS have been reported, with risk increase
Infants <2 years or BSA <0.6 m2 should have in association with use of radiation, alkyla­
dosing based on body weight: 0.67–1.7 mg/ tors, or topoisomerase II inhibitors. Keep
kg/day IV (dosing/frequency per protocol). drug protected from light.
Children ≥2 years, BSA ≥0.6 m2: 25–60 mg/m2 Extravasation may result in severe local tis­
IV with frequency dependent on protocol. sue necrosis and require intervention (see
dexrazoxane, dimethyl sulfoxide [DMSO]).
Infusion should ideally be via CVC given
A transient red‐orange discoloration of the
significant risk of tissue necrosis with
urine, sweat, saliva, and tears may occur for
extravasation. If administered via peripheral
up to 48 hours after a dose.
vein, care should be taken to ensure good
venous flow through a new IV in a large vein,
DEFEROXAMINE MESYLATE
avoiding joints, and monitoring carefully for
signs/symptoms of extravasation with Desferal, generics
immediate cessation if suspected. Chelating agent; antidote, iron toxicity
Injection: 500 mg
Pregnancy category C
Notes:
Do not give to patients with known hyper­ Indications:
sensitivity to daunorubicin, congestive heart
Treatment of acute iron intoxication and
failure, arrhythmias, or preexisting bone
chronic transfusional iron overload.
marrow suppression. Reduce dosage in
patients with hepatic, biliary, or renal impair­
Dosage:
ment. May cause myelosuppression, nausea,
vomiting, alopecia, stomatitis, and pigmenta­ Acute iron toxicity (age 3 years to adult):
tion of nail beds. Risk of developing irre­ Used as an adjunct to intervention with
versible myocardial toxicity increases rapidly syrup of ipecac, gastric lavage, and support­
at doses over 400 mg/m2. Risk factors that ive care measures.
increase risk of cardiac toxicity include prior 1000 mg IM (preferred route if patient not in
treatment with anthracyclines, concomitant shock) followed by 500 mg IM q4h for two
cardiotoxic drugs, prior or concomitant doses. Dependent on clinical response, sub­
mediastinal/pericardial radiation, and pedi­ sequent doses may be given q4–12 hours,
atric age group (increased in younger 24‐hours dose not to exceed 6000 mg.
patients and female sex). Concomitant usage
For patients with evidence of shock, may
of dexrazoxane should be considered in
give abovementioned doses IV, with IV
young patients with planned doxorubicin
infusion rate not to exceed 15 mg/kg/hour.
equivalent >300 mg/m2 (see doxorubicin for
conversion) and in all patients for doses Chronic iron overload: 25–50 mg/kg/day IV/
>300 mg/m2. Evidence of cardiac impair­ SC over 8–24 hours once daily for 5–7 days per
ment of function (signs, symptoms, week; maximum 2 g/24 hours. Doses should
decreased left ventricular ejection fraction not exceed 1000 mg if the IM route is chosen.
[LVEF]) may occur during treatment or
months to years after completion of therapy. Notes:
Long‐term cardiac monitoring is warranted Avoid in patients with known hypersensitiv­
and dependent on cumulative dose and other ity, severe renal disease, anuria, or primary
risk factors as mentioned in the preceding hemochromatosis. Prolonged use can lead
430 Formulary

to ocular or auditory disturbances including (round to nearest whole tablet). Consider


cataracts, decreased visual acuity, impaired higher doses for ferritin >2500 ng/ml.
peripheral, night, and color vision, and neu­ Consider holding dose or discontinuing for
rotoxicity‐related auditory abnormalities ferritin <500 ng/ml. Maintenance range:
(i.e., high‐frequency hearing loss). Periodic 20–40 mg/kg/day.
hearing and vision exams should be per­ Jadenu:
formed. High doses (>60 mg/kg), especially
Children ≥2 years to adults: 14–28 mg/kg PO
in children ≤3 years, have been associated
daily (calculate dose to the nearest whole
with growth retardation; reduction in dos­
tablet). Adjust dose in 3.5–7 mg/kg incre­
age may increase growth velocity. Vitamin C
ments according to response and goals; if
50 mg <10 years; 100 mg >10 years, 200 mg
not adequately controlled with doses of
adolescent/adult PO is often given to
21 mg/kg (i.e., serum ferritin levels persis­
increase bioavailability of iron to chelate.
tently >2500 ng/ml), doses of up to 28 mg/kg
Discontinue use in febrile patients because may be considered; do not exceed 28 mg/kg.
of increased susceptibility to infection with
Yersinia enterocolitica. Avoid rapid IV Non‐transfusion‐associated iron overload
administration as flushing, urticaria, hypo­ in thalassemia syndromes:
tension, and shock have been reported.
Exjade:
DEFERASIROX Children ≥10 years to adult: 10 mg/kg PO
daily starting dose, titrate by monitoring
Exjade, Jadenu
LIC (liver iron concentration) and ferritin
Chelating agent, iron; antidote, iron toxicity
levels up to 20 mg/kg/dose.
Tablet, for oral suspension: Exjade: 125, 250,
500 mg; Jadenu: 90, 180, 360 mg; Jadenu Jadenu:
sprinkles: 90, 180, 360 mg Children ≥10 years to adult: 7 mg/kg PO daily
(calculate dose to nearest whole tablet); if
Pregnancy category C
LIC >7 mg Fe/g liver dry weight after 6
months, increase dose by 3.5–7 mg/kg up to
Indications:
14 mg/kg/day; do not exceed 14 mg/kg/day.
Treatment of chronic iron overload due to
blood transfusions in patients ≥2 years and
Notes:
for treatment of chronic non‐transfusion
iron overload in thalassemia syndromes in Before initiating treatment, obtain serum
patients ≥10 years. ferritin level, baseline serum creatinine, cre­
atinine clearance (calculated), serum
transaminases, bilirubin, and baseline audi­
Dosage:
tory and ophthalmic examinations. Measure
Transfusion‐associated iron overload: serum ferritin monthly.
Exjade: Consider giving daily dose divided BID to
Children ≥2 years to adults: 20–40 mg/kg PO patients experiencing abdominal discom­
daily (round to nearest whole tablet). fort or nausea and vomiting.
Initiate therapy at 20 mg/kg/day. Adjust Do not chew or swallow whole tablets.
dose every 3–6 months based on serum fer­ Disperse tablets in water, apple juice, or
ritin levels or other measurement of iron orange juice (use 3.5 oz for doses <1 g; 7 oz
overload; increase by 5–10 mg/kg/day for doses ≥1 g); stir to form suspension and
Formulary 431

drink entire contents. Rinse remaining resi­ Dosage:


due in glass and drink. Administer at the Children and adults: 6.25 mg/kg IV every 6
same time every day, at least 30 minutes hours given as a 2‐hour infusion for at least
prior to food ingestion. 21 days, and continued until VOD/SOS res­
Do not give to patients with known hyper­ olution or up to 60 days of treatment.
sensitivity to deferasirox, low platelet counts
(<50 × 109/l), creatinine clearance (eGFR) Notes:
<40 ml/minute/1.73 m2, or serum creatinine In vitro defibrotide sodium has profibrino­
>2× age‐appropriate upper limit of normal. lytic activity and there is no known reversal
Dosing at 50% is suggested for eGFR >40 to agent. The use of defibrotide sodium is con­
<60 ml/minute/1.73 m2. Assess patients for traindicated in patients being treated con­
renal impairment and concurrent nephro­ currently with anticoagulants or fibrinolytic
toxic drugs. May cause intermittent protein­ therapies. For patients requiring an invasive
uria. Monitor serum electrolytes and procedure, discontinue the infusion at least
urinalysis. Use with caution in patients with 2 hours prior and then resume after the pro­
hepatic disease as well as in patients with cedure when it has been determined that the
concomitant anticoagulant, NSAID, or cor­ risk of bleeding is no longer present.
ticosteroid usage. Monitor transaminases Hemorrhage and hypersensitivity reactions
and liver function. Monitor LIC by liver are the major potential adverse reactions.
biopsy, MRI, or superconducting quantum The most common adverse reactions (inci­
interference device (SQUID). May cause dence greater than or equal to 10%) are
ocular or auditory disturbances, skin rash, hypotension, diarrhea, vomiting, nausea,
gastrointestinal pain, headache, diarrhea, and epistaxis.
sore throat, nausea, and vomiting. Monitor
hearing and vision, especially in children. DEFERIPRONE
Interruption or dose modification may be
Ferriprox/Kelfer/L1
necessary for evidence of renal, hepatic, or
Chelation agent, iron
gastrointestinal dysfunction.
Tablet: 500 mg
Safety and efficacy data are not available
when used in combination with other iron Pregnancy category D
chelators. Safety and efficacy have not been
established in children under 2 years of age. Indications:
Treatment of transfusional iron overload
DEFIBROTIDE due to thalassemia syndromes when current
Defitelio (defibrotide sodium) chelation therapy is inadequate.
Injection: 200 mg/2.5 ml vial, (80 mg/ml)
Dosage:
Pregnancy category not assigned (pregnancy
and breastfeeding not recommended) Adults: Initial dose: 25 mg/kg, orally, three
times a day (total daily dose: 75 mg/kg).
Indications: Maximum dose: 33 mg/kg, orally, three
Defibrotide is approved for the treatment of times a day (total daily dose: 99 mg/kg).
adult and pediatric patients with hepatic Round dose to the nearest 250 mg (half tablet).
VOD, also known as SOS, with renal or pul­ Tailor dose to response and therapeutic goals
monary dysfunction following HSCT. (maintenance or reduction of iron burden).
432 Formulary

Notes: Dosage:
Approval based on reduction in serum fer­ Von Willebrand disease, hemophilia A, bleed-
ritin; there are no trials showing a direct ing diathesis:
treatment benefit (i.e., improved symptoms, IV: 0.2–0.4 mcg/kg/dose, dilute in normal
functioning, or survival). saline (10 ml for patients <10 kg and 50 ml for
Safety and efficacy have not been estab­ ≥10 kg), infuse slowly over 15–30 minutes.
lished for transfusional iron overload with or
other chronic anemias. Safety and efficacy Intranasal spray (Stimate): One puff
have not been established in patients (150 mcg) for children under 50 kg and two
younger than 18 years of age. puffs (300 mcg) for children over 50 kg.
There is a FDA black box warning that Peak effect is 1–5 hours with intranasal route,
this drug may cause granulocytosis that 1–3 hours with IV route, and 2–7 hours with
can lead to serious infections and death; PO route. Duration of effect is 5–24 hours.
neutropenia may precede granulocytosis. Tachyphylaxis may occur with repeated dos­
It is recommended to monitor the abso­ ing within 72 hours.
lute neutrophil count (ANC) prior to and
Diabetes insipidus:
every week during therapy. Patients
should be advised to immediately report Children ≤12 years: Start with 0.05 mg/dose
any signs or symptoms suggesting infec­ BID and titrate to effect (i.e., control of
tion such as fever, sore throat, or flu‐like excessive thirst and urination); usual dose
symptoms. range is 0.1–0.8 mg/24 hours.
Children >12 years and adults: Start with
0.05 mg/dose BID and titrate to effect; usual
DESMOPRESSIN ACETATE
dose range is 0.1–1.2 mg/24 hours divided
DDAVP, Stimate, generics BID or TID.
Antihemophilic, hemostatic agent
Children 3 months to 12 years: 5–30 mcg/24 hours
Injection: 4 mcg/ml (1, 10 ml)
intranasally divided BID.
Tablets: 0.1, 0.2 mg
Solution: 1500 mcg/ml, 150 mcg/spray (25 Children >12 years and adults:
sprays, 2.5 ml) (Stimate) 10–40 mcg/24 hours intranasally divided
100 mcg/ml, 10 mcg/spray (50 sprays, 5 ml, daily to TID; titrate dose to effect.
with rhinal tube) (DDAVP) Nocturnal enuresis (≥6 years):
Pregnancy category B 0.2 mg PO at bedtime, titrate to effect (maxi­
mum dose 0.6 mg) or
Indications: 20 mcg intranasal at bedtime; divide and
administer 10 mcg in each nostril.
Intranasal Stimate or IV DDAVP are indi­
cated for maintenance of hemostasis in
Notes:
patients with mild or moderate hemophilia
A during surgery and postoperatively as well Do not give to patients with known hyper­
as treatment of mucosal bleeds in patients sensitivity to desmopressin. Avoid in patients
with von Willebrand disease and mild with severe type I, type IIB, or platelet‐type
hemophilia A. DDAVP is indicated for the von Willebrand disease, hemophilia B, and
treatment of central diabetes insipidus and severe hemophilia A (<1% factor VIII activ­
primary nocturnal enuresis. ity). Patients with moderate hemophilia
Formulary 433

A may not demonstrate an adequate response. Brain‐tumor‐associated cerebral edema:


Use cautiously in patients with a predisposition Loading dose: 1–2 mg/kg IV as a single dose;
to thrombophilia, electrolyte imbalance, and maintenance: 1–2 mg/kg/day IV in four to
hypertensive cardiovascular disease. May cause six divided doses for 1–5 days, or longer;
headache, nausea, emesis, seizures, blood pres­ maximum dose 16 mg/24 hours.
sure changes, hyponatremia, nasal congestion, Spinal cord compression with neurologic
abdominal cramps, and hypertension. Avoid in abnormalities: 2 mg/kg/24 hours IV divided
young children <2 years due to hyponatremia q6h.
and seizures. Not FDA approved in <6 years for
Chemotherapy:
treatment of nocturnal enuresis.
Refer to individual protocol.
A desmopressin challenge to document
responsiveness to these agents is typically Doses range from 6 to 20 mg/m2/day PO/IV
done in patients with bleeding disorders to for 5–7 days (may be longer in induction
document benefit prior to emergent use. therapy for ALL.
Note: intranasal forms come in two concen- Ophthalmic use: Instill one to two drops into
trations, use as specifically indicated. the conjunctival sac bilaterally. May use
q1–2h as needed to prevent and control
DEXAMETHASONE symptoms.
DexPak, TaperPak, Maxidex, generics (pre­
viously available as Decadron and Hexadrol) Notes:
Corticosteroid, anti‐inflammatory, Do not administer during active, untreated
immuno­suppressant infections with viral, fungal, or bacterial
Tablets: 0.25, 0.5, 0.75, 1.5, 2, 4, 6 mg organisms. Prolonged use may cause bone
Solution: 1 mg/ml pain, glucose intolerance, hypertension, fat
Ophthalmic solution: 0.1% (5 ml) redistribution and striae, and avascular
Injection: 4 mg/ml necrosis of bone.
Pregnancy category C
DEXRAZOXANE
Indications: Zinecard, Totec (extravasation kit)
Used systemically for chronic inflammation, Antidote, anthracycline; antidote (anthracy­
allergic, hematologic, neoplastic (i.e., leuke­ cline extravasation)
mia), autoimmune disease, cerebral edema Injection: 250, 500 mg
and increased intracranial pressure, and
septic shock. Utilized as an antiemetic for Pregnancy category D
chemotherapy‐induced nausea and vomit­
ing and as an ophthalmic solution for con­ Indications:
trol of chemical conjunctivitis from Reduction of anthracycline‐induced cardio­
concomitant administrations of agents such toxicity (approved in metastatic breast cancer
as high‐dose cytarabine. for patients who have received a cumulative
dose of 300 mg/m2 of doxorubicin equiva­
Dosage: lents with plan for further dosing). Treatment
Antiemetic: 0.2 mg/kg/dose (5 mg/m2/dose) for local tissue extravasation of anthracy­
IV 30 minutes prior to and every 6 hours clines. Currently FDA approved in adults
after chemotherapy. only, off label use in pediatrics.
434 Formulary

Dosage: DIMERCAPROL
Refer to individual protocol. British anti‐Lewisite (BAL), generics
Children and adults: 10:1 dose ratio with Chelating agent (antidote for gold, mercury,
doxorubicin equivalents (i.e., 300 mg/m2 lead, and arsenic toxicity)
dexrazoxane for 30 mg/m2 doxorubicin). Injection: 100 mg/ml
Complete anthracycline administration Pregnancy category C
within 30 minutes of start of dexrazoxane.
Treatment for extravasation: Adolescents Indications:
≥18 years to adults: 1000 mg/m2/dose within Antidote to gold, mercury, and arsenic poison­
6 hours on days 1 and 2; maximum dose ing; used in conjunction with edetate calciu12
2000 mg; followed by 500 mg/m2/dose on m disodium to treat severe lead poisoning.
days 3; maximum dose 1000 mg. If using ice
to affected area, remove 20 minutes prior to Dosage:
infusion. Infuse in extremity/area other
than the affected area at room temperature For severe lead poisoning (lead level >70 mcg/
in a large vein IV over 1–2 hours. Administer dl) or encephalopathy: 25 mg/kg/day divided
same time every day. Do not mix or admin­ q4h deep IM for a minimum of 72 hours, may
ister with any other drug. give up to 5 days in severely symptomatic
patients. Calcium sodium ethylenediamine­
Infuse over 15 minutes, do not administer tetraacetic acid (EDTA) should be started at a
IV push. separate site with the second dimercaprol
Do not use DMSO in patients receiving dexra- dose. If symptoms of encephalopathy persist,
zoxane for anthracycline‐induced extravasation. a second course of treatment can begin after a
minimum of 2 days of rest following the initial
Notes: 5‐day course. Therapy should continue until
Suggested use in young children at anthracy­ the patient is clinically stable. Once stable, a
cline outset when expected total dose to 10–14‐day period of equilibration should be
exceed 300 mg/m2 doxorubicin equivalents. instated before again measuring the lead level.
Otherwise recommended in patients with a If the level remains ≥70 mcg/dl, another
cumulative doxorubicin equivalent dose of course of double therapy should be initiated.
300 mg/m2 who are continuing to receive Less severe lead poisoning (45–70 mcg/dl):
anthracyclines (or as directed by protocol). Treatment with dimercaprol is not recom­
Dose‐limiting toxicity is myelosuppression, mended in this situation due to toxicity;
which may be additive to chemotherapy. May patients should be treated with succimer or
chelate heavy metals, leading to increases in calcium disodium EDTA instead.
calcium, iron, and triglycerides and decreases
in sodium and zinc. May have antitumor
Notes:
effects. Dose reduction in renal or hepatic
impairment may be needed. Dexrazoxane Do not give to patients with hepatic or renal
has topoisomerase II inhibition activity and insufficiency. Do not use in iron, selenium,
secondary AML and MDS have been or cadmium poisoning. Use with caution in
reported when used in combination with patients with G6PD deficiency (may cause
other anti‐cancer drugs with known links to hemolysis) and peanut sensitivity. Hydrate
second malignant neoplasms. and alkalinize urine to protect the kidneys.
Formulary 435

May cause hypertension, tachycardia, gas­ Indication:


trointestinal disturbance, headache, fever, Part of standard treatment for children
transient neutropenia, and nephrotoxicity. with high‐risk neuroblastoma. Each anti­
Symptoms may be relieved by antihista­ body is made of both mouse and human
mines. Do not use concomitantly with iron. components and targets glycolipid GD2,
Ensure calcium salt is given if edetate calcium expressed on neuroblastoma cells and on
disodium is used in conjunction with BAL. normal cells of neuroectodermal origin,
including the central nervous system and
DIMETHYL SULFOXIDE peripheral nerves. Dinutuximab is indi­
RIMSO‐50, DMSO cated in combination with granulocyte‐
Antidote (anthracycline extravasation) macrophage colony‐stimulating factor
Topical: 50% solution (GM‐CSF), interleukin‐2 (IL‐2), and 13‐
cis‐retinoic acid (RA) after completion of
Pregnancy category C chemotherapy, surgery, autologous trans­
plant, and radiation therapy.
Indications:
FDA indication for treatment of interstitial Dosing:
cystitis. Limited information for use after 17.5 mg/m2/day IV over 10–20 hours for 4
cooling for local control of extravasation of consecutive days for maximum of five
anthracyclines. May also be used for extrava­ cycles:
sation of ifosfamide, cisplatin, fluorouracil, Cycles 1, 3, and 5 (24 days duration): Administer
and carboplatin. on days 4, 5, 6, and 7.
Cycles 2 and 4 (32 days duration): Administer
Dosage: on days 8, 9, 10, and 11.
Apply 1–2 ml topically within 10–25 minutes Initiate at infusion rate of 0.875 mg/m2/hour
to affected area, then every 4–8 hours for 7–14 for 30 minutes; may gradually increase infu­
days, until resolution. Allow to dry and apply sion rate, as tolerated, to maximum of
clean, dry non‐occlusive dressing. Vitamin E 1.75 mg/m2/hour.
or Aloe vera has been used topically following
Prior to each course: Verify that patient has
the first application to alleviate local burning
adequate hematologic, respiratory, hepatic,
or stinging (ensure DMSO has dried).
and renal function.
Notes:
Pretreatment and guidelines for pain
The role of DMSO in the treatment of management:
anthracycline extravasation remains contro-
versial. Do not use concurrently with dexra- IV hydration:
zoxane. Use with local cooling of tissues. administer 0.9% NaCl injection, USP 10 ml/
kg IV over 1 hour.
DINUTUXIMAB
Unituxin Analgesics:
Chimeric monoclonal antibody, Ch14.18 Morphine sulfate (50 mcg/kg) IV immediately
Injection: 17.5 mg/5 ml prior to initiation of dinutuximab; continue as
Pregnancy category drip at infusion rate of 20–50 mcg/kg/hour
436 Formulary

during and for 2 hours following completion causes severe neuropathic pain, and can cause
of dinutuximab. severe sensory neuropathy and severe periph­
Administer additional 25–50 mcg/kg IV eral motor neuropathy.
morphine sulfate doses pro re nata, as neces­ More than 25% of children taking these
sary (PRN) for pain up to once q2h followed drugs experience pain, fever, hives, vomit­
by an increase in morphine sulfate infusion ing, diarrhea, bone marrow suppres­
rate in clinically stable patients (may also sion causing decrease of platelets, red blood
initiate nurse‐controlled analgesia). cells, white blood cells, and albumin, hypo­
May use fentanyl or hydromorphone if mor­ tension, electrolyte imbalance including low
phine sulfate not tolerated. sodium, potassium, and calcium, elevated
transaminases, infusion reactions, and cap­
Antihistamines and antipyretics: illary leak syndrome. Other common
adverse effects include retention or urine for
Administer antihistamine such as diphen­ weeks to months after receiving the drugs,
hydramine (0.5–1 mg/kg; not to exceed protein in urine, blurred vision or dilated
50 mg) IV over 10–15 minutes starting pupils, infections, edema, high blood pres­
20 minutes prior to initiation of dinutuxi­ sure, intractable bleeding, tachycardia, and
mab and as tolerated q4–6h during dinu­ weight gain.
tuximab infusion.
Administer acetaminophen (10–15 mg/kg; DIPHENHYDRAMINE
not to exceed 650 mg) 20 minutes prior to
Benadryl, generics
each dinutuximab infusion and q4–6h PRN
Antihistamine
for fever or pain; administer ibuprofen
Capsules/tablets over the counter (OTC): 25,
(5–10 mg/kg) q6h PRN for control of persis­
50 mg
tent fever or pain.
Chewable tablets: 12.5 mg
Elixir: 12.5 mg/5 ml
Notes:
Injection: 50 mg/ml
Dinutuximab can cause severe side effects,
including severe pain that must be controlled Pregnancy category B
with morphine, and a high risk of infusion
reaction that must be controlled with antihista­ Indications:
mines and anti‐inflammatory drugs. Morphine Treatment of allergic symptoms, anaphy­
is administered prior to, during, and for 2 laxis, medication and transfusion reac­
hours after infusion of dinutuximab to manage tions, chemotherapy and other induced
the severe pain that this drug causes. An anti­ nausea and vomiting, and motion sickness;
histamine and an anti‐inflammatory are also used as an antitussive or for mild sedation.
given before, during, and after to manage Prevents or treats metocloperamide‐
the infusion reaction. Capillary leak is com­ induced and phenothiazine‐induced dys­
mon and the patient should be weighed twice tonic reactions.
daily in addition to daily measures of blood
counts and albumin. Ideally, low blood pres­ Dosage:
sure should be treated with an oncotic load; Antiemetic and antivertigo: 0.5–1 mg/kg/
vasopressor support may be required. dose q6h PO, IM, or IV (maximum dose
The United States’ label for dinutuximab car­ 50 mg and 300 mg/24 hour).
ries a black box warning for life‐­threatening Pruritus: 0.5–1 mg/kg/dose q6h PO, IM, or IV
infusion reactions and neurotoxicity, as it (maximum dose 50 mg and 300 mg/24 hour).
Formulary 437

Notes: ing myelosuppression. Use with caution in


patients who have received very high cumu­
Do not give to patients with known hyper­
lative doses of anthracyclines (i.e., ≥550 mg/
sensitivity to diphenhydramine. Do not use
m2 or ≥400 mg/m2 with concomitant use of
with concurrent monoamine oxidase (MAO)
cyclophosphamide or chest radiation; based
inhibitors, in acute attacks of asthma, or in
on anthracycline dose equivalents). Monitor
patients with gastrointestinal or urinary
cardiac function with LVEF and electrocar­
obstruction. Use with caution in patients
diogram (ECG) after every 75–100 mg/m2 of
with glaucoma, peptic ulcer disease, urinary
anthracycline given. Cardiac toxicity may be
tract obstruction, and hyperthyroidism.
early (acute) or late (delayed months to years)
Avoid alcohol. Note: many preparations con­
and long‐term cardiac monitoring is essen­
tain alcohol. May cause sedation, nausea,
tial. Evidence of early cardiac toxicity may
vomiting, xerostomia, blurred vision, and
include sinus tachycardia and/or ECG
central nervous system effects. May cause
changes with non‐specific ST‐T wave
paradoxical activation in children.
changes. Delayed cardiotoxicity may be man­
ifested by tachyarrhythmias and other abnor­
DOXORUBICIN HYDROCHLORIDE mal rhythms including premature ventricular
Adriamycin, Doxil (liposomal formulation) contractions as well as evidence of bundle
Antineoplastic antibiotic, anthracycline branch block, and/or signs/symptoms of con­
Injection: 10, 50 mg vials (protect from light) gestive heart failure. Monitor cardiac func­
tion during treatment in addition to blood
Pregnancy category D counts, creatinine, and total bilirubin.

Indications: Modify dosage in renal impairment. May


cause severe bone marrow suppression, alo­
Treatment of ALL, AML, Hodgkin lym­ pecia, mucositis, and transient (48 hour)
phoma, lymphoma, Wilms tumor, neuroblas­ red‐orange discoloration of the urine, sweat,
toma, and soft tissue and bone sarcomas. saliva, and tears. Extravasation may result in
severe local tissue necrosis; ensure adminis­
Dosage: tration in CVC or large vein and monitor
Refer to individual protocol. closely. May cause photosensitivity reactions
and patients should be instructed to avoid
20–90 mg/m2 IV in repeated doses (may be
excessive exposure to sunlight and utilize
weekly or monthly or per phase of therapy).
sunblock sun protection factor (SPF) ≥ 15.
Infusions may be over 15 minutes, several
hours, or as a continuous infusion over
24–48 hours. Although controversial, longer DRONABINOL
infusions may be more cardioprotective,
Marinol, tetrahydrocannabinol (THC),
especially when high cumulative doses (i.e.,
generics
>450 mg/m2) are anticipated (though cannot
Cannabinoid
be given with dexrazoxane in such cases).
Capsules: 2.5, 5, and 10 mg
Children <12 kg and infants are dosed based
on weight rather than BSA. Pregnancy category C

Notes: Indications:
Do not give to patients with known hyper­ Treatment of nausea and vomiting associ­
sensitivity to doxorubicin, severe congestive ated with chemotherapy in patients who
heart failure or cardiomyopathy, or preexist­ have failed to respond to conventional
438 Formulary

antiemetic therapy; treatment of anorexia Dosage:


associated with weight loss in patients with For severe lead poisoning (lead level >70 mcg/
human immunodeficiency virus (HIV). dl) and/or encephalopathy: 1000–1500 mg/
Evidence of benefit in chemotherapy‐induced m2/24 hours as an 8–24 hours infusion or
anorexia is mixed; no well‐controlled studies divided q12h, given in combination with
have been performed in children. dimercaprol (BAL). Use the higher dose in
the presence of encephalopathy. Give at a
Dosage: separate site starting with the second dimer-
Antiemetic: 5 mg/m2/dose 1–3 hours PO caprol dose.
prior to chemotherapy, then q2–4h; maxi­ Less severe lead poisoning (20–70 mcg/dl):
mum 6 doses/24 hour, 15 mg/m2/dose in 1000 mg/m2/24 hours IV as an 8–24 hours
adults. Titrate dose to effect. infusion or intermittent dosing q12h for 5
Appetite stimulant: Adult dosing: 2.5 mg PO days. May repeat course as needed after 2–4
BID 1 hour prior to lunch/dinner; if not toler­ days of re‐equilibration without EDTA.
ated, reduce dose to 2.5 mg every night at bed­ Maximum daily dose 75 mg/kg.
time (QHS); maximum dose 20 mg/24 hour.
Notes:
Notes: Do not give to patients with severe renal dis­
Do not give to patients with known hyper­ ease or anuria. Requires inpatient admission
sensitivity to any cannabinoid or sesame oil. with aggressive intravenous hydration and
Do not use in patients with history of sub­ frequent monitoring of serum electrolytes
stance abuse or mental illness. Use with cau­ including calcium and phosphorus, renal
tion in heart disease, seizures, and hepatic function, and urinalyses. Establish urine
disease. A dose‐related “high” (easy laugh­ flow prior to administration and maintain
ing, elation, or heightened awareness) is hydration/urine output throughout course
reported in one‐quarter of patients using of therapy. Dose reduction is recommended
cannabinoids as antiemetics. Other side for mild renal disease. Monitor with contin­
effects include dizziness, anxiety, difficulty uous ECG due to risk for arrhythmias.
concentrating, hypotension, and increased Rapid IV infusion can result in sudden
appetite. Psychological and physiologic increase in intracranial pressure in patients
dependence may occur, but addiction is with cerebral edema. May cause zinc and
uncommon. It is a controlled (Schedule III) copper deficiency. May be administered IM;
substance. give with 0.5% prilocaine. Ensure calcium
salt is given.
EDETATE CALCIUM DISODIUM
Calcium EDTA, CaNa2EDTA, generics ELTROMBOPAG
Calcium disodium versenate Promacta
Heavy metal antagonist; antidote, lead Thrombopoietin receptor agonist
toxicity Tablets: 12.5, 25, 50, 75, 100 mg
Injection: 200 mg/l Oral suspension: 25 mg packet
Pregnancy category B Pregnancy category C

Indications: Indications:
Used as an adjunct in the treatment of acute In children (>1 year) and adults with severe
and chronic lead poisoning. thrombocytopenia and chronic immune
Formulary 439

thrombocytopenia purpura (cITP) who have Indications:


had a suboptimal response to corticosteroids, Prophylaxis and treatment of thromboem­
immunoglobulins, or splenectomy; chronic bolic disorders such as DVT following sur­
hepatitis–C‐associated thrombocytopenia to gery or trauma, due to central line infection
allow initiation/maintenance of treatment with and pulmonary embolus.
interferon; aplastic anemia with insufficient
response to immunosuppressive therapy. Dosage:

Dosage: Treatment of DVT or thromboembolus


(TE):
Chronic ITP:
Infants <2 months: 1.5 mg/kg/dose q12h SC;
Children ≥6 years and adult: 50 mg PO QD higher doses of 1.7–2 mg/kg/dose q12h SC
(maximum 75 mg QD). have been recommended for neonates.
Children 1–5 years: 25 mg PO QD. >2 months through adult: 1 mg/kg/dose q12h
SC; 1.5 mg/kg/dose q24h can be used in
Aplastic anemia: adults. Higher doses may be required in
children to reach therapeutic levels.
50 mg PO QD (maximum dose 150 mg QD);
based on adult data. Titrate dose to achieve peak anti‐factor Xa
levels of 0.5–1.0 U/ml (based on fraction­
Notes: ated anti‐Xa level). Peak anti‐Xa levels
should be drawn 4 hours after administra­
Administer on an empty stomach (1 hour tion of a steady state dose (minimum two to
before or 2 hours after a meal). Take at least 2 three prior doses).
hours before or 4 hours after any medication
See Chapter 10 for further guidelines. Some
or products containing polyvalent cations
centers advocate that TPA be given concom­
including antacids, calcium‐rich foods, and
itantly with enoxaparin in high‐risk situa­
mineral supplements. Do not crush tablets;
tions (i.e., large central embolism).
prepare oral suspension in water only and
Prophylaxis or treatment with enoxaparin
administer within 30 minutes. Dose adjust­
may continue for prolonged periods
ment for hepatic impairment and for some
dependent on the underlying risks for
patients of East‐Asian ancestry. Adjust dose to
thrombophilia and response to therapy. For
maintain platelet count ≥50 × 109/l. Side effects
those patients that will be maintained with
are uncommon and include nausea, vomiting,
oral anticoagulation (e.g., warfarin), 4–5
diarrhea, respiratory tract infections, myalgia,
days of overlap with enoxaparin should be
and increased alanine aminotransferase.
given in order to achieve a therapeutic INR.

ENOXAPARIN DVT/TE prophylaxis:


Lovenox, generics Infant <2 months: 0.75 mg/kg/dose q12h SC.
Anticoagulant, low‐molecular‐weight hepa­ Infant ≥2 months–18 years: 0.5 mg/kg q12h
rin (LMWH) SC or 1 mg/kg/dose q24h, maximum dose
Injection (prefilled syringe): 30 mg/0.3 ml, 30 mg.
40 mg/0.4 ml, 60 mg/0.6 ml, 80 mg/0.8 ml,
Anti‐factor Xa levels do not need to be fol­
100 mg/ml
lowed in patients on prophylactic dosing.
Approximate anti‐factor Xa activity:
Data are limited but 1 mg/kg once daily dos­
100 IU/mg
ing can be considered in patients with compli­
Pregnancy category B ance issues and in those with difficulty giving
440 Formulary

twice daily SC injections. An insuflon catheter Injection (preservative‐free single use vials):
should also be considered.Recommended 2000, 3000, 4000, 10,000, 20,000, 40,000 U/
anti‐Xa fractionated levels (obtained 4 hours ml (1 ml)
after second or third SC dose):
Pregnancy category C
DVT treatment: 0.5–1 units/ml.
DVT prophylaxis: 0.1–0.3 units/ml.
Indications:
Note: LMWH may be given IV at the same
dose as SC. Treatment of anemia associated with
chronic renal failure, anemia related to ther­
Notes: apy with zidovudine (AZT) in HIV‐infected
patients, and anemia of prematurity. Usage
Do not give to patients with known hyper­ in cancer patients remains controversial.
sensitivity to enoxaparin or pork products May be considered as an adjunct for anemia
(derived from porcine intestinal mucosa), treatment in patients with religious beliefs
active major bleeding, prosthetic heart against utilization of blood transfusion (see
valves, acute heparin‐induced (or LMWH‐ Darbepoetin).
induced) thrombocytopenia, and recent
major surgery or cerebral hemorrhage. Use
of the medication should be discussed in Dosage:
patients with religious beliefs which prohibit Anemia in chronic renal failure: Start at 50 U/
the consumption of pork products. Do not kg (dose range 50–250 U/kg) administered
give to patients with drug‐induced throm­ SC/IV three times a week; higher doses may
bocytopenia. Use with caution in patients be needed. Dose is individualized to achieve
with recurrent gastrointestinal ulcers, bleed­ and maintain the lowest hemoglobin suffi­
ing diathesis, and severe renal dysfunction; cient to avoid transfusion, not to exceed
lower initial doses should be given to 11 g/dl.
patients with renal insufficiency and failure. Anemia in AZT‐treated HIV patients: 100 U/
Do not use with concurrent spinal or epi­ kg SC three times a week; dose range
dural anesthesia or lumbar puncture for 50–4000 U/kg two to three times per week;
chemotherapy. May cause fever, confusion, hgb should not exceed 12 g/dl.
edema, nausea, hemorrhage, hypochromic
anemia, thrombocytopenia, and pain/ery­ Anemia in cancer patients: 600 U/kg IV once
thema at the injection site. weekly; dose titrated to effect (maximum
40,000 U). Do not initiate therapy if
In case of overdose, protamine sulfate can be hgb ≥ 10 g/dl; adjust dose to maintain the
given, although the reversal is not complete lowest hgb level needed to avoid transfu­
as with unfractionated heparin (UFH). 1 mg sions. Adult dose 150 U/kg. SC three times a
of protamine sulfate neutralizes approxi­ week or 40,000 U SC once weekly.
mately 0.6–0.7 mg of enoxaparin. Discontinue use after 8 weeks of therapy if
Note that unfractionated anti‐Xa level (used transfusions are still required or chemother­
for monitoring heparin infusions) is not the apy is completed.
same as fractionated anti‐Xa level (used for Anemia of prematurity: 250 U/kg/dose SC
monitoring enoxaparin dosing). three times per week for ten doses; alterna­
tively, 200–400 U/kg/dose IV/SC three to
EPOETIN ALFA five times per week. Administer supplemen­
Recombinant human erythropoietin tal iron 3–6 mg elemental iron/kg/24 hours
Epogen, Procrit, erythropoietin (many other regimens exist).
Formulary 441

Notes: day for three doses q3–4 weeks for three to


four courses.
Do not give to patients with known hyper­
sensitivity to albumin, uncontrolled hyper­
tension, and in newborns with neutropenia. Notes:
Use with caution in patients with porphyria Do not give to patients with known hyper­
or a history of seizures. May cause headache sensitivity to etoposide. Use with caution
and elevated blood pressure. Meta‐analyses and consider dose reduction in patients with
have shown that usage leads to a significant renal or hepatic impairment. May cause
increased risk of death, thrombosis, and facial flushing, fever, fatigue, nausea, vomit­
serious cardiovascular events in cancer ing, bone marrow suppression, alopecia,
patients. and peripheral neuropathy. Associated with
Evaluate iron stores (ferritin and TIBC) second malignant neoplasms, primarily
before therapy initiation. Iron supplementa­ acute leukemia, dose and frequency related.
tion is recommended unless iron stores are Give IV infusions over at least 1 hour due
already in excess. to associated hypotension with rapid infu­
sion. Infusion should be stopped or infu­
ETOPOSIDE sion rate be decreased with hypotension.
VP‐16 After fluid resuscitation, the infusion may
Toposar, Etopophos be restarted at a slower rate if stopped and
Antineoplastic, mitotic inhibitor the hypotension resolved. Patients with
Capsule, softgel: 50 mg hypersensitivity to etoposide can usually be
Injection: 20 mg/ml; 5, 25, 50 ml vials safely treated with etoposide phosphate
(Etopophos; secondary to vehicle used to
Pregnancy category D dissolve etoposide).

Indications:
EUTECTIC MIXTURE OF LIDOCAINE
Treatment of germ cell tumors, lymphoma, AND PRILOCAINE
brain tumors, acute leukemias, neuroblas­ EMLA
toma, rhabdomyosarcoma, histiocytosis, Local anesthetic, topical anesthetic
and conditioning for BMT. Cream: 5, 30 g tubes

Dosage: Pregnancy category B


Refer to individual protocol.
AML: 100–150 mg/m2/day IV for 2–5 days Indications:
in Induction and Intensification courses. Used as a topical anesthetic applied to nor­
Dose per body weight in patients with mal intact skin to provide local anesthesia
BSA < 0.6 m2 (3.3 mg/kg/dose). for minor procedures such as venipuncture,
Brain tumors: 100–150 mg/m2/day IV for placement of peripheral venous access line,
1–3 days; may also be given long‐term PO lumbar puncture, circumcision, and minor
(using injection solution for oral adminis­ dermatologic procedures.
tration); good central nervous system
penetration Dosage:
Neuroblastoma: 100 mg/m2/day IV (dose Apply 2.5 g per site to normal intact skin and
by weight for BSA < 0.6 m2, 3.3 mg/kg/ cover with occlusive dressing for 30–60 minutes
dose) over 1 hour for days 1–5 or alternate prior to procedure.
442 Formulary

Notes: of the infusion. Only administer Injectafer


Do not give to patients with known hyper­ when personnel and therapies are immedi­
sensitivity to lidocaine, prilocaine, or other ately available for the treatment of serious
local anesthetic. Commonly causes blanch­ hypersensitivity reactions.
ing of skin or erythema. May cause mild Safety and efficacy have not been estab­
burning or altered temperature sensation. lished in pediatrics. Recent studies suggest
this is a safe alternative for treatment of
FERRIC CARBOXYMALTOSE hypoferritinemia‐associated fatigue in
Injectafer adolescent and adult women. Other
Iron adverse effects may include hypertension,
Injection: 50 mg elemental Fe/ml; 750 mg/15 ml flushing, injection site discoloration, and
vial transient decrease in phosphorus levels
(<2 mg/dl).
Pregnancy category C
FERRIC GLUCONATE
Indications:
Ferrlecit
Treatment of iron deficiency anemia in
Iron
adults who have intolerance or insufficient
Injection: 62.5 mg elemental iron in 5 ml
response to oral iron; and in patients with
(12.5 mg/ml)
non‐dialysis‐dependent chronic renal
disease. Pregnancy category B

Dosage: Indication:
<50 kg: 15 mg/kg/dose IV for two doses sep­ Ferrlecit is utilized for the repletion of iron
arated by at least 7 days; maximum 740 mg/ stores in adults and children ≥6 years of age
dose and 1500 mg per course; may be undergoing hemodialysis.
repeated if iron deficiency anemia recurs.
≥50 kg: 750 mg IV for two doses, separated Dosage:
by at least 7 days; maximum dose 1500 mg Adults: 10 ml (125 mg of elemental iron,
per course; may be repeated if iron defi­ maximum dose). Ferrlecit may be diluted in
ciency anemia recurs. 100 ml of 0.9% sodium chloride adminis­
tered by IV infusion over 1 hour per dialysis
Notes: session. Ferrlecit may also be administered
Serious hypersensitivity reactions, including undiluted as a slow intravenous injection (at
anaphylactic‐type reactions, some of which a rate of up to 12.5 mg/minute) per dialysis
have been life‐threatening and fatal, have session. For repletion treatment most
been reported in patients receiving patients may require a cumulative dose of
Injectafer. Patients may present with shock, 1000 mg of elemental iron administered
clinically significant hypotension, loss of over eight dialysis sessions.
consciousness, and/or collapse. Monitor Children ≥6 years to adult: 0.12 ml/kg
patients for signs and symptoms of hyper­ Ferrlecit (1.5 mg/kg of elemental iron, maxi­
sensitivity during and after Injectafer mum dose 125 mg) diluted in 25 ml 0.9%
administration for at least 30 minutes and sodium chloride and administered by IV
until clinically stable following completion infusion over 1 hour per dialysis session.
Formulary 443

Notes: Prophylaxis:
The most commonly reported adverse reac­ Premature infant: 2 mg/kg/day, maximum
tions (≥10%) in adult patients are nausea, dose 15 mg/24 hour.
vomiting and/or diarrhea, injection site Term infant: 1–2 mg/kg/day, maximum dose
reaction, hypotension, cramps, hyperten­ 15 mg/24 hour.
sion, dizziness, abnormal erythrocytes (e.g., Child 2–12 years: 2 mg/kg/day, maximum
changes in morphology, color, or number dose 30 mg/24 hour.
of red blood cells), dyspnea, chest pain, leg
Adolescent to adult: 60 mg PO QD.
cramps, and pain. In patients 6–15 years of
age, the most common adverse reactions
(≥10%) were hypotension, headache, hyper­ Notes:
tension, tachycardia, and vomiting. Do not give to patients with known hyper­
Ferrlecit may cause clinically significant hypo­ sensitivity to iron salts, hemochromatosis,
tension. Hypotension associated with light­ and transfusional iron overload. Absorption
headedness, malaise, fatigue, weakness, or of iron is decreased when given with tetra­
severe pain in the chest, back, flanks, cycline, antacids, or milk. Less gastrointesti­
or groin has been reported. These hypoten­ nal irritation when given with food.
sive reactions may or may not be associated Concurrent administration of 200 mg or
with signs and symptoms of hypersensitivity more of vitamin C per 30 mg elemental iron
reactions and usually resolve within 1–2 hours. increases absorption of oral iron; iron
replacement products should be given with
FERROUS GLUCONATE orange juice. May cause constipation, dark
Ferate, generics stools, nausea, and epigastric pain. Recent
Iron (12% elemental Fe) studies suggest once daily dosing (or even
Tablets (OTC): 240 mg (27 mg Fe), 325 mg alternate day dosing) may optimize iron
(36 mg Fe) absorption in iron deficiency states.

Pregnancy category A
FERROUS SULFATE
Indications: Fer‐In‐Sol, FeroSul, Slow FE, generics
Prevention and treatment of iron deficiency Iron (20% elemental Fe)
anemia. Tablet OTC: 325 mg (65 mg Fe)
Drops (Fer‐In‐Sol) OTC: 75 mg (15 mg
Fe)/1 ml
Dosage:
Elixir (FeroSul and generics)
Dose is expressed in terms of elemental iron. OTC: 220 mg (44 mg Fe)/5 ml
Extended release tabs (Slow FE, generics)
Iron deficiency anemia: OTC: 140 mg (45 mg Fe), 160 mg (50 mg Fe),
Premature infant: 2–4 mg/kg/day PO 324 mg (65 mg Fe), 325 mg (65 mg Fe)
divided QD BID; maximum dose
15 mg/24 hour. Pregnancy category A
Child: 3–6 mg/kg/day PO in 1–3 divided
doses. Indications:
Adult: 60–100 mg PO in 1–2 divided doses; Prevention and treatment of iron deficiency
up to 240 mg/day. anemia.
444 Formulary

Dosage: Dosage:
See Ferrous Gluconate. Neonatal sepsis with neutropenia: 5–10 mcg/
kg IV/SC daily for 3–5 days.
Notes: Chemotherapy‐induced neutropenia:
See Ferrous Gluconate. Children and adults: 5–10 mcg/kg IV/SC
once daily until ANC is greater than
FERROUS FUMARATE 2–10 × 109/l (per protocol; given beyond
Ferrets, generics nadir period 7–10 days after chemotherapy
Iron salt (33% elemental Fe) administration).
Tablet OTC: 90 mg (29.5 mg Fe), 324 mg Peripheral blood progenitor cell mobilization:
(106 mg Fe), 325 mg (106 mg Fe), 456 mg 10 mcg/kg SC daily for 4 days before the first
(150 mg Fe) leukapheresis procedure and continued
until the last leukapheresis.
Pregnancy category A
Congenital neutropenia: 2.5–6 mcg/kg/day
Indications: SC; titrate dose to desired ANC to prevent
infection.
Prevention and treatment of iron deficiency
anemia. Idiopathic or cyclic neutropenia: 5 mcg/kg SC
once daily; titrate dose to desired ANC to
prevent infection.
Dosage:
Elevation of ANC is usually within 24 hours,
See Ferrous Gluconate.
though it may be delayed in severe myelo­
suppression. Transient increase in ANC may
Notes:
occur when granulocyte colony‐stimulating
See Ferrous Gluconate. factor (G‐CSF) is begun shortly after com­
pletion of chemotherapy; avoid premature
FILGRASTIM discontinuation.
Neupogen, Granix, Zarxio, G‐CSF SC route of administration is preferred due
Blood formation, colony‐stimulating factor to prolonged serum levels over IV route. If
Injection: 300 mcg/ml (1, 1.6 ml) used IV and the G‐CSF concentration is
Pregnancy category C >15 mcg/ml, add 2 mg albumin/1 ml of IV
fluid to prevent drug absorption in the IV
Indications: administration set.

Used to decrease the period of neutropenia


and the associated risk of infection in Notes:
patients with malignancies receiving myelo­
suppressive chemotherapy associated with a Avoid in patients with hypersensitivity to E.
significant incidence (i.e., >20%) of severe coli‐derived proteins or G‐CSF. May cause
neutropenia. Has been used in zidovudine‐ bone pain and increases in uric acid and lac­
associated neutropenia in HIV‐infected tate dehydrogenase. Do not administer
patients and in patients with nonchemo­ 24 hours before or after administration of
therapy‐induced neutropenia (acquired and chemotherapy due to potential for pro­
congenital). Should be considered in longed myelosuppression from increased
patients with complicated infection with chemosensitivity of myeloid progenitors
underlying neutropenia. stimulated by G‐CSF.
Formulary 445

FLUCONAZOLE Notes:
Diflucan, generics Do not give to patients with known hyper­
Antifungal sensitivity to fluconazole or other azoles.
Tablet: 50, 100, 150, 200 mg May cause nausea, headache, rash, vomit­
Injection: 2 mg/ml ing, abdominal pain, hepatitis, cholestasis,
Suspension: 10, 40 mg/ml and diarrhea. Antagonism may occur if
amphotericin B and fluconazole are used
Pregnancy category C (vaginal candidiasis)/D
concurrently. Many drug interactions exist;
for all other indications
contraindicated concurrently with other
medications that prolong QT interval and
Indications: are metabolized via the CYP450 3A4
Prophylaxis and treatment of susceptible enzyme. May increase effects, toxicity, and/
fungal infections, including oropharyngeal, or levels of cyclosporine, midazolam, tac­
esophageal, and vaginal candidiasis, and rolimus, and many other drugs. Rifampin
systemic fungal infections with Candida sp. increases fluconazole metabolism. Consult
as well as treatment and suppression of the PDR or a pharmacist when ordering
cryptococcal meningitis. Species of Candida fluconazole in a patient receiving many
with decreased in vitro susceptibility to flu­ medications.
conazole are being isolated. Fluconazole is
more active against candidal species such as FLUDARABINE
Candida albicans than species such as Fludara
Candida parapsilosis, Candida glabrata, and Antineoplastic, antimetabolite; purine analog
Candida tropicalis. Injection: 50 mg vial

Dosage: Pregnancy category D


Oropharyngeal candidiasis: 6 mg/kg load­
Indications:
ing dose PO/IV followed by 3 mg/kg/day
(dependent on severity of infection) for Treatment of leukemias including ALL,
2–4 weeks (dependent on clinical response AML, CLL, and non‐Hodgkin lymphoma.
and immune status of the patient). Conditioning regimen for HSCT.
Neonates under 14 days of age are dosed
every 24–72 hours. Maximum dose 400 mg/ Dosage:
day. 25 mg/m2 IV over 30 minutes daily for 5
Esophageal candidiasis: 12 mg/kg loading consecutive days. Dosage may be decreased
dose PO/IV, followed by 6 mg/kg/day for or delayed based on evidence of hemato­
2–4 weeks; maximum dose 400 mg/day. logic or nonhematologic toxicity.
Invasive systemic candidiasis and cryptococ- Conditioning regimen pretransplant: 40 mg/
cal meningitis: 12 mg/kg loading dose PO/ m2 IV daily for 4 days. For patients <10 kg,
IV, followed by 6–12 mg/kg/day for use weight‐based dosing: 1.33 mg/kg/dose.
2–4 weeks or longer. Maximum dose 12 mg/
kg/day or 800 mg/day in adults. Notes:
Prophylaxis in immunocompromised hosts/ Common adverse effects include marked
bone marrow transplant prophylaxis: myelosuppression and lymphopenia, increas­
3–6 mg/kg/dose PO/IV; maximum dose ing the risk of opportunistic infections.
400 mg/day. Edema, skin rash, and peripheral neuropathy
446 Formulary

are reported. Has been associated with Patients weighing 50–100 kg – 7.5 mg SC
autoimmune hemolytic anemia and transfu­ once daily.
sion‐associated graft‐versus‐host disease. Patients weighing >100 kg – 10 mg SC once
Pretreatment may result in difficulty collect­ daily.
ing peripheral blood stem cells for transplant.
Notes:
FONDAPARINUX
Fondaparinux cannot be given intramuscu­
Arixtra larly. Safety and efficacy in pediatric patients
Anticoagulants, hematologic; Factor Xa have not been established. Because risk for
inhibitors bleeding during treatment is increased in
Injection: (Single‐dose, prefilled syringes) adults who weigh <50 kg, bleeding may be an
2.5, 5, 7.5, or 10 mg important safety concern in pediatric
Pregnancy category B patients. Fondaparinux does not require rou­
tine monitoring with coagulation studies or
Indications: drug levels, though can be initially assessed
with a fondaparinux level 3 hours after the
Prophylaxis of DVT in patients undergoing second or subsequent dose. Renal function
hip fracture surgery (including extended should be evaluated as renal dosing may be
prophylaxis), hip replacement surgery, knee indicated. Pediatric trials are ongoing, and
replacement surgery, or abdominal surgery. weight‐based dosing (i.e., 0.1 mg/kg) in
Treatment of DVT or acute pulmonary patients 1–18 years of age has been reported.
embolism (PE) when administered in con­ A specific fondaparinux reversal agent is in
junction with warfarin. development, though protamine would par­
tially reverse the medication in an emergent
Dosage: situation. Given the longer half‐life com­
pared with enoxaparin, fondaparinux must
Fondaparinux is given subcutaneously once be held a minimum of 48 hours prior to lum­
daily. The dose in individuals with normal bar puncture and longer with more invasive
renal function is based on body weight and procedures.
the indication for anticoagulant use (e.g.,
prophylaxis for venous thromboembolism
[VTE] versus therapy). Fondaparinux is not FOLIC ACID
currently indicated in the pediatric popula­ Folate
tion and guidelines exist for treatment based Folvite, generics
on a weight of 50 kg or greater. Blood formation, water‐soluble vitamin
VTE prophylaxis: 2.5 mg SC once daily in Tablet OTC: 0.4, 0.8, 1 mg
patients weighing ≥50 kg. In the periopera­ Solution: 50 mcg/ml
tive setting, the first dose is given 6–8 hours Injection: 5 mg/ml
postoperatively (after skin closure), as done
Pregnancy category A/C
in all of the major clinical trials.
Superficial vein thrombosis (treatment): 2.5 mg Indications:
SC once daily in patients weighing ≥50 kg.
Treatment of megaloblastic anemia resulting
VTE or pulmonary embolus (treatment): from folate deficiency; supplementation for
Patients weighing <50 kg – 5 mg SC once patients with chronic hemolytic anemia (e.g.,
daily. sickle cell disease, hereditary spherocytosis).
Formulary 447

Dosage: hydration. May adversely affect tooth and


Infants to children <12 months: 15 mcg/kg bone growth in children, safety and effi­
daily PO; maximum 50 mcg/day. cacy not fully evaluated. May cause elec­
Children ≥1–11 years: Initial dose 1 mg/day trolyte imbalances and symptoms of
PO; maintenance dose 0.1–0.4 mg/day PO. hypocalcemia. May cause seizures; risk
Children >11 years and adults: Initial dose factors include renal impairment, low
1 mg/day PO; maintenance dose 0.4–0.8 serum calcium, and underlying central
mg/day PO. nervous system condition. Foscarnet is a
vesicant; only infuse into veins with ade­
Notes: quate blood flow.
May mask hematologic effects of vitamin B12
deficiency but will not prevent progression GANCICLOVIR
of neurologic abnormalities. Cytovene, Zirgan, generics
Antiviral
FOSCARNET Injection: 500 mg (can be prepared into oral
Foscavir, generics suspension)
Antiviral Pregnancy category C
Injection: 24 mg/ml
Pregnancy category C Indications:
Treatment of CMV retinitis, pneumonitis,
Indications: encephalitis, and gastrointestinal infection in
Alternative to ganciclovir for treatment of immunocompromised patients. Prevention of
cytomegalovirus (CMV) infection; treat­ symptomatic CMV disease in transplant
ment of acyclovir‐resistant HSV infections patients with reactivation of latent disease.
in immunocompromised hosts. Has antiviral activity against HSV‐1 and
HSV‐2.
Dosage:
Dosage:
Children and adults:
CMV retinitis: 180 mg/kg/day IV divided Congenital CMV infections, neonates and
q8–12h for 14–21 days (with or without gan­ infants: 12 mg/kg/day slow IV infusion
ciclovir) followed by daily IV maintenance divided q12h for 6 weeks.
of 90–120 mg/kg/day. CMV retinitis, children >3 years to adults:
10 mg/kg/day IV divided q12h for
Acyclovir‐resistant HSV infection: 40 mg/kg/ 14–21 days followed by maintenance 5 mg/
dose IV q8–12h up to 3 weeks or until kg/day IV as a single daily dose for 7 days/
lesions heal; repeat treatment may lead to week or 6 mg/kg/day IV for 5 days/week.
resistance. Prevention of CMV in transplant recipients,
Varicella zoster unresponsive to acyclovir: children and adults: 10 mg/kg/day IV
40 mg/kg/dose q8–12h for 7–10 days (ado­ divided q12h for 1–2 weeks followed by
lescent give 90 mg/kg/dose q12h). maintenance 5 mg/kg/day IV once daily for
7 days/week or 6 mg/kg/day IV for 5 days/
Notes: week until 100–120 days posttransplant.
May cause renal impairment, adjust dose Minimum dilution is 10 mg/ml and should
for renal dysfunction. Assure adequate be infused ≥1 hour.
448 Formulary

Notes: Injection: 5 mg
Do not give to patients with known hyper­ Pregnancy category D
sensitivity to ganciclovir, acyclovir, or any
component. Patients must use appropriate Indications:
contraception due to teratogenic effects for
Treatment of AML (newly diagnosed adults
at least 90 days after therapy completion.
and refractory/relapsed pediatric AML).
Immunosuppressive drugs may increase
risk of cytopenias. Concurrent ampho­
Dosage:
tericin B, tacrolimus, or cyclosporine
increase risk of nephrotoxicity. Use with Refer to individual protocol; used as single
caution in patients with renal impairment. agent or in combination treatment.
Children with BSA <0.6 m2: 0.1 mg/kg/dose.
GEMCITABINE Children with BSA ≥0.6 m2: 3 mg/m2/dose,
as a single dose or multidosing.
Gemzar
Antineoplastic; pyrimidine antagonist Premedicate children with acetaminophen
Injection: 200 mg, 1 g 15 mg/kg (maximum 650 mg) and diphen­
hydramine 1 mg/kg (maximum 50 mg).
Pregnancy category D Consider addition of methylprednisolone
1 mg/kg PO or IV; additional doses of aceta­
Indications: minophen and diphenhydramine may be
Treatment of ovarian, breast, non‐small cell administered every 4 hours after the initial
lung, pancreatic cancer, and other solid pretreatment dose. Give methylpredniso­
tumors. Recently evaluated in refractory/ lone or an equivalent corticosteroid for any
relapsed pediatric ALL. sign of an infusion reaction such as fever,
chills, hypotension, or dyspnea during the
Dosage: infusion or within 4 hours afterward.
Refer to individual protocol.
Notes:
Pediatric ALL: 10 mg/m2/minute IV over
360 minutes weekly for 3 weeks, followed by Do not give to patients with known hyper­
1 week of rest. sensitivity to gemtuzumab. Severe hyper­
sensitivity reactions and infusion‐related
Notes: reactions may occur including pulmonary
edema, acute respiratory distress syndrome,
Contraindicated in patients with known
and anaphylaxis. Administer by slow infu­
hypersensitivity to gemcitabine. Common
sion over 2 hours. Severe hepatic toxicity
adverse effects include myelosuppression,
including veno‐occlusive disease (SOS) has
nausea and vomiting, hepatic transaminitis,
been reported. Use with caution in patients
and peripheral edema. Toxicities requiring
with renal or hepatic impairment or pulmo­
cessation of treatment include: severe pul­
nary disease. May cause prolonged QTc.
monary toxicity, capillary leak syndrome,
hemolytic uremic syndrome or severe renal
GLUCARPIDASE
toxicity, and posterior reversible encepha­
lopathy syndrome (PRES). Can rarely cause Voraxaze
radiation recall. Antidote, recombinant glutamate carboxy­
peptidase (carboxypeptidase G2)
GEMTUZUMAB OZOGAMICIN Solution: 1000 unit vial, reconstitute with
1 ml NS
Mylotarg
Antineoplastic, monoclonal antibody Pregnancy category C
Formulary 449

Indications: Solution: 0.2 mg/ml, 50 mcg/ml


Injection: 0.1 mg/ml, 1 mg/ml (prefilled
Adjunct to leucovorin rescue for the treat­
syringe for SC extended release [Sustol]
ment of toxic plasma methotrexate concen­
10 mg/0.4 ml)
trations (>0.454 mcg/ml [1 μmol/l]) in
patients with delayed methotrexate clear­ Pregnancy category B
ance due to renal impairment.
Indications:
Dosage: Prevention and treatment of nausea and
Children and adults: Single 50‐unit/kg dose vomiting associated with moderately eme­
IV, administer over 5 minutes. togenic chemotherapy. Has also been uti­
lized in prevention of postoperative and
Notes: radiation‐induced nausea and vomiting.
Provides an alternative, nonrenal route for
methotrexate elimination (in patients with Dosage:
delayed methotrexate clearance due to renal Chemotherapy‐induced nausea and
impairment) by converting circulating vomiting:
methotrexate to inactive metabolites that IV:
are primarily eliminated hepatically.
Children ≥2 years to adults: 10–20 mcg/kg/
Administration of glucarpidase 2 hours
dose IV 15–60 minutes before chemother­
before leucovorin reduces peak concentra­
apy; may be repeated two to three times fol­
tions and AUC of leucovorin and of its
lowing chemotherapy over 24 hours;
active metabolite 5‐methyl‐tetrahydrofolate.
maximum dose 3 mg/dose or 9 mg/24 hours.
Do not administer leucovorin within 2
Alternatively, a single dose of 40 mcg/kg
hours before or after glucarpidase
15–60 minutes before chemotherapy has
Plasma methotrexate concentrations meas­ been used.
ured by immunoassay within 48 hours follow­
PO:
ing glucarpidase administration are
unreliable. During first 48 hours after glucarp­ Infant, child, adolescent: 40 mcg/kg/dose
idase administration, use chromatographic PO/IV BID, initiate first dose 1 hour prior to
method to obtain reliable measurements of chemotherapy.
plasma methotrexate concentrations. Adults: 2 mg/24 hours PO divided q12–24h;
Common adverse effects include paresthe­ initiate first dose 1 hour before
sia, flushing, nausea and/or vomiting, hypo­ chemotherapy.
tension, and headache. The drug is SC (Sustol): 10 mg SC at least 30 minutes
distributed primarily in the intravascular prior to first dose in combination with dexa­
compartment and does not cross the blood– methasone; do not administer more than
brain barrier. every 7 days.
Transdermal patch (Sancuso): Adult: Apply 1
GRANISETRON patch 24–48 hours prior to chemotherapy,
Sancuso, Sustol, generics (previously availa­ may be worn for up to 7 days.
ble as Kytril) PONV prevention: Children ≥4 years to
Antiemetic agent, serotonin (5‐HT3) adults: 20–40 mcg/kg IV dosed prior to
antagonist anesthesia or immediately before anesthesia
Tablet: 1 mg reversal; maximum dose 1 mg once.
450 Formulary

Radiation‐induced nausea and vomiting >16 years: 15 U/kg/hour (maximum rate


prevention: 1650 U/hour).
Adults: 2 mg PO daily administered 1 hour Adjust dose per one of the following labora­
prior to radiation. tory goals:
UFH anti‐Xa level: 0.3–0.7 units/ml.
Notes:
or
Inducers or inhibitors of the cytochrome
Activated partial thromboplastin time
P450 drug‐metabolizing enzymes may
(APTT) level (reagent specific to reflect anti‐
increase or decrease, respectively, the
Xa level of 0.3–0.7 units/ml): 50–80 seconds.
drug’s clearance. Use with caution in liver
disease. May cause hypertension, hypoten­ or
sion, arrhythmias, agitation, and insom­ If anti‐Xa levels not available, use APTT
nia. Safety and efficacy in pediatric 1.5–2.5 times the control value.
patients for the prevention of PONV has Lab samples should be collected 4–6 hours
not been established. after initiation or dose/infusion rate change;
do not collect from a heparinized line or the
HEPARIN SODIUM same extremity as the site of heparin infusion.
Generics Note that measurement of unfractionated
Anticoagulant anti‐Xa will provide the most accurate level.
Injection: many vial sizes, porcine‐based ori­ Heparin flush:
gin; preservative free
Peripheral IV: 1–2 ml of 10 U/ml solution q4h.
Lock flush solution: 1, 10, 100 U/ml (por­
cine‐based, some products may be preserva­ Central lines: 2–3 ml of 100 U/ml solution
tive free or contain benzyl alcohol) q24h and after access. Dosing often estab­
Injection for IV infusion: Porcine‐based, lished by institutional policy.
D5W: 40 U/ml, 50 U/ml, 100 U/ml; 0.9% Arterial lines and TPN lines (central lines):
NaCl: 2 U/ml; 0.45% NaCl: 50 U/ml, 100 U/ Add heparin to make a final concentration
ml, contains EDTA of 0.5–1 U/ml.
120 U = approximately 1 mg
Notes:
Pregnancy category C
Do not give to patients with known hypersen­
Indications: sitivity to heparin or pork products, severe
Prophylaxis and treatment of thromboem­ thrombocytopenia, suspected intracranial
bolic disorders; thrombus prophylaxis for hemorrhage, shock, severe hypotension, and
central venous access devices. uncontrolled bleeding (unless secondary to
disseminated intravascular coagulation).
Dosage: Avoid IM injections and medications affecting
platelet function. Use actual body weight when
Anticoagulation in infants and children:
dosing. Should discuss usage in patients with
Initial: 75 U/kg IV bolus over 10 minutes in
religious beliefs which prohibit the consump­
infants to <16 years; 70 U/kg in >16 years
tion of pork products. Use preservative free
(maximum dose 7700 U).
heparin in neonates and consider more dilute
Maintenance IV continuous infusion: heparin.
<1 year: 28 U/kg/hour. Unfractionated anti‐Xa level is not the same
≥1–16 years: 20 U/kg/hour (maximum rate as fractionated anti‐Xa level (used for moni­
1650 U/hour). toring enoxaparin dosing).
Formulary 451

Reversal agent: Protamine sulfate 1 mg per Indications:


100 U heparin infused in previous 4 hours.
Treatment of inflammatory dermatoses and
adrenal insufficiency; also used as a chemother­
HYALURONIDASE apeutic agent for intrathecal administration.
Amphadase, Hydase (bovine sources, may
contain edetate disodium and thimerosal) Dosage:
Hylenex (recombinant human source) Physiologic replacement: 12–18 mg/m2/day
Vitrase (ovine source, preservative free) PO divided q6–8h.
Antidote (extravasation) Stress dosing (consider for >2 weeks on gluco-
Injection: 150 U/ml (Vitrase 200 U/ml) corticoid therapy or patients in shock): Solu‐
Pregnancy category C Cortef, 25–100 mg/m2/day PO/IV.
Intrathecal chemotherapy with methotrexate
Indications: and/or cytarabine: Dose per protocol, range
15–30 mg/dose.
Used to influence the dispersion and absorp­
tion of other drugs and increase rate of
Notes:
absorption of parenteral fluids given by
hypodermoclysis (subcutaneously); treat­ Do not give to patients with known hyper­
ment of IV extravasations. sensitivity to hydrocortisone, polymyxin B
sulfate, or neomycin sulfate. Avoid in
Dosage: patients with infections from herpes sim­
plex, vaccinia, and varicella.
Infants and children: Dilute 150‐unit vial
(1 ml) in NS to volume of 10 ml (15 U/ml). HYDROMORPHONE
Give 1 ml (15 U) by administering five sepa­ HYDROCHLORIDE
rate injections of 0.2 ml (3 U) at borders of
extravasation site SC or intradermal using a Dilaudid, Dilaudid‐HP, Exalgo, generics
25‐ or 26‐gauge needle. Administer as early Narcotic, analgesic
as possible after extravasation (minutes to Tablets: 2, 4, 8 mg
1 hour). Tablets (extended release, Exalgo, generics):
8, 12, 16, 32 mg
Notes: Solution: 1 mg/ml
Suppository: 3 mg
Do not give to patients with known hyper­ Injection: 1, 2, 4, 10 mg/ml
sensitivity to hyaluronidase or respective
sources (bovine, ovine). Do not inject in or Pregnancy category C/D (prolonged use,
around infected, inflamed, or cancerous high doses at term)
lesions; do not use with dopamine or alpha‐
agonist extravasation. May cause urticaria. Indications:
Management of moderate to severe pain.
HYDROCORTISONE
Solu‐Cortef, Cortef, generics Dosage:
Corticosteroid Children <50 kg (not for use in neonates):
Tablet (Cortef, generics): 5, 10, and 20 mg PO: 0.03–0.1 mg/kg/dose q3–4h PRN (max­
Suspension: 2.5 mg/ml imum 5 mg/dose).
Injection, as sodium succinate (Solu‐Cortef): IV: 0.015 mg/kg/dose q3–6h PRN.
100, 250, 500, 1000 mg vial
Adolescents–adults ≥50 kg (doses not weight
Pregnancy category C based):
452 Formulary

PO: 1–2 mg (up to 4 mg/dose in adults) increase dose in increments of 5 mg/kg/day


q4–6h PRN. every 12 weeks to a maximum of 35 mg/kg/
IV: 0.2–0.6 mg/dose q2–4h PRN (up to day. Discontinue for bone marrow toxicity:
1 mg/dose in adults). ANC <2 × 109/l, platelets <80 × 109/l, reticu­
locyte count <8 × 109/l, or hemoglobin
IM/SC: 0.8–1 mg/dose q4–6h PRN.
<9 g/dl. Restart therapy after recovery at
PR: 3 mg q4–8h PRN. 2.5 mg/kg/day less than dose that produced
toxicity.
Notes:
Do not give to patients with known hyper­ Notes:
sensitivity to hydromorphone. Dose reduc­ Do not give to patients with known hyper­
tion recommended in renal insufficiency or sensitivity to hydroxyurea or those with
severe hepatic impairment. Avoid use in severe anemia or severe bone marrow sup­
neonates because of potential central nerv­ pression. Use with caution in renal impair­
ous system effects. Use with caution in ment. Common adverse reactions are
infants and young children. Causes less pru­ hematologic and gastrointestinal.
ritus than morphine. Approximate 6:1 con­
version from morphine (i.e., 6 mg morphine IDARUBICIN
is equivalent to 1 mg dilaudid).
Idamycin, generic
Antineoplastic antibiotic, anthracycline
HYDROXYUREA
Injection: 1 mg/ml; 5, 10, 20 ml vials
Hydrea, Droxia
Antineoplastic Pregnancy category D
Capsule: 500 mg (Hydrea); 200, 300, 400 mg
(Droxia) Indications:
Treatment of AML and relapsed ALL.
Pregnancy category D
Dosage:
Indications:
Refer to individual protocol.
Treatment of malignancies including CML,
APL induction: 12 mg/m2/day IV over
desmoid tumors; adjunct in the manage­
15–20 minutes for 3–4 consecutive days;
ment of sickle cell anemia to reduce pain
often given concurrently with cytarabine.
events, acute chest syndrome, hospitaliza­
tions, transfusion needs, and mortality;
Notes:
hyperleukocytosis due to blast crisis in CML
and in relapsed acute leukemias; hypereo­ Do not give to patients with known hypersen­
sinophilic syndrome. sitivity to idarubicin, severe congestive heart
failure, or cardiomyopathy. Avoid in preexist­
Dosage: ing bone marrow suppression unless the
potential benefit warrants the risk. May need to
Refer to individual protocol. reduce dosage with impaired renal or hepatic
CML, hyperleukocytosis: Initial dose 10–20 function. Irreversible myocardial toxicity may
mg/kg PO once daily; adjust dose according to occur as the cumulative dosage approaches
hematologic response and symptoms. 550 mg/m2 doxorubicin equivalents (or
Sickle cell anemia: Initial dose 15 mg/kg 400 mg/m2 with chest irradiation or concomi­
(range 10–20 mg/kg/day) once daily; tant cyclophosphamide administration).
Formulary 453

This may be an acute or late effect and cardiac Tablet: 100, 400 mg
monitoring with echocardiogram, LVEF deter­
Pregnancy category D
mination, and ECG is required during and
after completion of therapy. Secondary leuke­
Indications:
mia has been reported.
Treatment of newly diagnosed adult and
IFOSFAMIDE pediatric Philadelphia chromosome positive
Ifex (Ph+) CML in chronic or accelerated phase;
Antineoplastic, alkylating agent Ph+ CML in blast phase adult Ph+ ALL
Injection: 1 g vial (relapsed or refractory); pediatric Ph+ ALL;
aggressive systemic mastocytosis without
Pregnancy category D KIT mutation; KIT positive (CD117) unre­
sectable gastrointestinal stromal tumors
Indications: (GIST); hypereosinophilic syndrome or
Used in combination with other antineo­ chronic eosinophilic leukemia with specific
plastics in the treatment of Hodgkin and mutations; and myelodysplastic/myelopro­
non‐Hodgkin lymphoma, ALL, osteosar­ liferative disease associated with platelet‐
coma, rhabdomyosarcoma, Ewing sarcoma, derived growth factor receptor (PDGFR)
and advanced Wilms tumor. gene rearrangements.

Dosage: Dosage:
Refer to individual protocol. Refer to individual protocol.
Usual dose is 700–1800 mg/m2/day IV for 5 Children ≥2 years:
consecutive days every 3–4 weeks.
Ph+ CML, chronic phase, new diagnosis:
340 mg/m2/day PO q24h.
Notes:
Ph+ ALL: 340 mg/m2/day PO q24h postin­
Do not give to patients with known hyper­
duction and through maintenance.
sensitivity to ifosfamide. Avoid in severe
bone marrow suppression. Use with caution If daily dose exceeds 400–600 mg, give doses
in impaired renal function; hydrate the divided q12h. Do not crush tablets, but may
patient prior to administration and ensure dissolve in water or apple juice if patient
good urine flow (i.e., urine specific gravity cannot swallow tablets.
≤1.010). Ifosfamide may lead to syndrome of May need to adjust dose with hepatic or
inappropriate antidiuretic hormone (SIADH) hematologic toxicity.
secretion. Mesna is used for uroprotection The optimal duration of therapy for CML or
with higher doses to decrease risk of hemor­ ALL is not determined.
rhagic cystitis. Urinalysis should be moni­
tored closely for specific gravity and heme.
May cause central nervous system toxicity Notes:
including hallucination, somnolence, confu­ Do not give to patients with known hyper­
sion, coma, or encephalopathy. sensitivity to imatinib. May cause fluid
retention, weight gain, edema, pleural effu­
IMATINIB MESYLATE sion, pericardial effusion, and pulmonary
Gleevec edema. Use with caution in patients where
Antineoplastic, tyrosine kinase inhibitor (TKI) fluid retention may be poorly tolerated such
454 Formulary

as congestive heart failure or left ventricular Notes:


dysfunction. May cause Stevens–Johnson
Avoid in patients with hypersensitivity to
syndrome, hepatotoxicity, hemorrhage, and
immune globulin or blood products and in
hematologic toxicity. Use with caution in
those with immunoglobulin A (IgA) defi­
patients with preexisting hepatic or renal
ciency; consult pharmacist for IgA‐depleted
impairment. Imatinib is metabolized via the
products. Give slow intravenous infusion,
CYP3A4 enzyme system, use with caution
over 4–8 hours, and consider premedication
in patients receiving concurrent therapy
with acetaminophen and diphenhydramine
that may be an inducer or inhibitor and
to avoid infusional toxicity (nausea, vomit­
affect drug concentrations. Avoid warfarin.
ing, flushing, fever, chills, severe headaches).
May cause photosensitivity, pruritus, rash,
If toxicity develops, stop infusion and treat
gastrointestinal disturbance, bone or joint
symptomatically, then resume at slower rate
pain, and blurred vision.
of infusion. Future doses should be given at
slower rate with premedications before and
IMMUNE GLOBULIN
every 4 hours during infusion. May cause
Flebogamma 5% (50 mg/ml) aseptic meningitis or acute renal failure.
Gamunex‐C 10% (100 mg/ml)
Gammagard 10% (100 mg/ml) INOTUZUMAB OZOGAMICIN
Octagam 5% (50 mg/ml)
Besponsa
Carimmune NF: 1, 3, 6, 12 g for reconstitution
Antibody–drug conjugate
Hizentra (SC): 20% (200 mg/ml) (check
Injection: 0.9 mg in single dose vial
package insert for dose conversion)
Pregnancy category not assigned, can cause
Pregnancy category C
fetal harm
Indications:
Indication:
Treatment of immunodeficiency states (e.g., Approved in adults for the treatment of
HIV, agammaglobulinemia), secondary relapsed or refractory B‐cell precursor ALL.
immunodeficiencies (e.g., bone marrow
transplant), immune thrombocytopenic Dosage:
purpura (ITP), Kawasaki disease, and lym­
phoproliferative disorders. Adults:
For the first cycle, the recommended dose of
Dosage: inotuzumab ozogamicin for all patients is
Immunodeficiency/HIV infection/post bone 1.8 mg/m2 per cycle, administered as three
marrow transplant (replacement dose): 400– divided doses on day 1 (0.8 mg/m2), day 8
500 mg/kg/dose IV q3–4 weeks; maintain (0.5 mg/m2), and day 15 (0.5 mg/m2). The
trough IgG level ≥400–500 mg/dl recommended dosing for subsequent cycles
depends on response to treatment.
ITP: 800–1000 mg/kg IV for 1–2 consecu­
tive days pending response; may be given Pediatric:
repeatedly q3–6 weeks based on clinical Refer to protocol.
need.
Kawasaki disease: 2 g/kg IV as a single dose; Notes:
consider second dose for persistent The medication consists of the humanized mon­
symptoms. oclonal antibody against CD22 (inotuzumab),
Formulary 455

linked to a cytotoxic agent from the class of cali­ Children: refractory solid tumor or brain
cheamicins called ozogamicin. tumor: 50 mg/m2/day IV for 5 days, repeat
The drug prolongs the QT interval in some cycle q3 weeks.
people, so it should be used with caution in
people with heart arrhythmias. Notes:
The United States’ label for the use of inotu­ Do not give to patients with known hyper­
zumab ozagamicin carries an FDA black box sensitivity to irinotecan. Avoid concomitant
warning concerning the risk of liver toxicity, administration with St. John’s Wort or keto­
in particular hepatic VOD/SOS, which has conazole and in patients with severe bone
been fatal in some people. The risk of this is marrow failure. A new cycle of irinotecan
higher in adults who take the drug before should not begin until serious treatment‐
having HSCT and more people die who have induced toxicity has recovered: ANC
HSCT following treatment with this drug ≥1.0 × 109/l and platelet count >100 × 109/l. If
than people who have HSCT taking other the patient has not recovered following a 2‐
chemotherapies. The risk increases as more week rest, consider discontinuing. May
rounds of treatment with inotuzumab ozo­ cause severe, dose‐limiting, and potentially
gamicin are administered. There is unclear fatal diarrhea. Early diarrhea (during or
risk for VOD/SOS in pediatric patients. shortly after infusion) may be accompanied
The most common serious adverse reac­ by symptoms of rhinitis, increased saliva­
tions are infection, neutropenia with fever, tion, flushing, miosis, lacrimation, diapho­
hemorrhage, stomach pain, fever, VOD/ resis, and abdominal cramping. Atropine
SOS, and fatigue. 0.01 mg/kg IV (maximum dose 0.4 mg) may
be used to prevent or treat cholinergic
Between 10 and 20% of people also report
symptoms and early diarrhea.
loss of appetite, vomiting, diarrhea, mouth
sores, constipation, chills, and injection site Late diarrhea occurs >24 hours after therapy
reactions. and can be prolonged leading to life‐threat­
ening dehydration and electrolyte imbalance.
IRINOTECAN Treat promptly with loperamide until a nor­
mal pattern of bowel movements returns.
Camptosar, generic
Antibiotic support (cefixime or cefpodoxime
Antineoplastic, topoisomerase inhibitor
starting several days prior to treatment) can
Injection: 20 mg/ml; 2, 5 ml vial
be utilized as a prophylactic and treatment
Pregnancy category D measure by preventing gastrointestinal bacte­
rial conversion of irinotecan to SN‐38, the
Indications: metabolite which causes diarrhea.
Treatment of metastatic colon carcinoma, neu­ If the patient develops persistent diarrhea,
roblastoma, hepatoblastoma, brain tumors, ileus, fever, or severe neutropenia, interrupt
Ewing sarcoma, and rhabdomyosarcoma. or reduce subsequent doses. If grade 3 (seven
to nine stools/day, incontinence, and/or
Dosage: severe cramping) or grade 4 (≥10 stools/day,
Referral to individual protocol. grossly bloody stool, and/or need for paren­
Children: refractory solid tumor, low dose, teral support) diarrhea occurs, interrupt
protracted: 20 mg/m2/day IV daily for 2 con­ treatment and reduce subsequent dosing.
secutive weeks, followed by a week of rest; Moderately emetogenic, therefore premedication
repeat q3 weeks. with an anti‐emetic regimen is recommended.
456 Formulary

May cause severe myelosuppression; halt for neu­ Children >20 kg to adult: 25 mg (0.5 ml).
tropenic fever. Use with caution in patients with Total replacement dose of iron dextran for iron
renal or hepatic impairment. deficiency anemia:

IRON COMPLEX, POLYSACCHARIDE (ml) = 0.0442 × LBW (kg) ×


(Hgbn − Hgbo) + [0.26 × ABW up to
Myferon 150, PIC 200, Poly‐Iron 150,
maximum 14 ml];
NovaFerrum, NovaFerrum Pediatric drops,
many brands (previously available as Niferex) ABW = actual body weight; LBW = lean
Capsule (OTC): 50 mg (NovaFerrum 50), body weight.
150 mg (Myferon 150, Poly‐Iron 150, oth­
Males: 50 kg + 2.3 kg for every inch over 5 ft.
ers), 200 mg (PIC 200); 150 mg strength may
in height.
contain 50 mg vitamin C
Oral liquid: (NovaFerrum 125, OTC) Females: 45.5 kg + 2.3 kg for every inch over
125 mg/5 ml 5 ft. in height.
Oral drops: (NovaFerrum Pediatric drops Hgbn = desired hemoglobin (g/dl) = 12 if
OTC) 15 mg/ml <15 kg or 14.8 if >15 kg.
Pregnancy category A Hgbo = measured hemoglobin (g/dl).
Total replacement dose of iron dextran for
Indications: acute blood loss:
Treatment of iron deficiency anemia. (Assumes 1 ml of normocytic, normochro­
mic red cells = 1 mg elemental iron).
Dosage: Replacement iron (mg) = 0.02 × blood loss
See Ferrous Gluconate. (ml) × hematocrit expressed as a decimal
fraction.
Notes: Maximum daily dose, IM/IV:
See Ferrous Gluconate. Infants <5 kg: 25 mg (0.5 ml).
Children 5–10 kg: 50 mg (1 ml).
IRON DEXTRAN COMPLEX
Children >10 kg to adults: 100 mg (2 ml).
Dexferrum, INFeD
Iron salt Parenteral administration:
Injection: 50 mg elemental iron/ml (2 ml) IM: use Z‐track technique (deep into upper
outer quadrant of buttock); test dose at same
Pregnancy category C
site using the same method.
Indications: IV: infuse test dose over at least 5 minutes
(Dexferrum) or 30 seconds (INFeD). May ini­
Treatment of microcytic hypochromic ane­
tiate treatment 1 hour after test dose and com­
mia resulting from iron deficiency in
plete replacement doses in two to three daily
patients in whom oral administration is not
infusions if replacement exceeds daily maxi­
feasible or is ineffective. Approved in chil­
mum dose. Dilute replacement dose in normal
dren ≥4 months.
saline (50–100 ml) to maximum concentra­
tion of 50 mg/ml and infuse over 2–6 hours at
Dosage (IV or IM): a maximum rate of 50 mg/minute. Avoid dilu­
Begin with a test dose 1 hour prior to start­ tion in dextrose due to an increased incidence
ing iron dextran therapy: of local pain and phlebitis.
Infants <10 kg: 10 mg (0.2 ml). Monitor vital signs and for symptoms of
Children 10–20 kg: 15 mg (0.3 ml). anaphylaxis during the IV infusion.
Formulary 457

Notes: Dosage:
Do not give to patients with known hyper­ Test dose (optional): Infuse 25% of the first
sensitivity to iron dextran (anaphylaxis may day’s dose up to a maximum of 25 mg undi­
occur), in anemia not associated with iron luted over 30 minutes. A 10 mg test dose can
deficiency, with hemochromatosis, or with be given in dialysis patients.
hemolytic anemia. Use with caution in
IV administration is undiluted over
patients with histories of significant aller­
2–5 minutes. May infuse 100 mg with a
gies, asthma, serious hepatic impairment,
maximum of 100 ml NS over 15 minutes. IM
preexisting cardiac disease, and rheumatoid
administration is not possible with this
arthritis (may cause an exacerbation of
product.
arthritis). Discontinue oral iron prior to ini­
tiating parenteral iron. Sweating, urticaria, Children: end stage renal disease on
arthralgia, fever, chills, dizziness, headache, hemodialysis:
and nausea may be delayed 24–48 hours 1 mg (elemental iron)/kg/dialysis treatment
after large doses of IV administered drug or (repletion).
3–4 days following IM administration. The 0.3 mg (elemental iron)/kg/dialysis treat­
IV route is preferred for patients with ment (maintenance), administer at last hour
chronic renal disease and cancer‐related of dialysis treatment at a frequency of three
anemia (adults). times a week.
Agents for treatment of acute anaphylaxis should Children: iron deficiency anemia, refractory
be readily available. Adverse events are much to PO therapy:
more associated with the high‐molecular‐
Calculation of iron deficit: 0.6 × body weight
weight formulation (Dexferrum) than with
(kg) × [100 − (measured hgb divided by
the low‐molecular‐weight formulation
desired hgb × 100]. Replacement dose is
(INFeD). The low‐molecular‐weight formu-
administered by giving an initial dose of
lation is recommended.
5–7 mg/kg (maximum dose 300 mg/24 hour)
Total‐dose infusions have been used safely q3–7 days until total iron replacement is
and are the preferred method of administra­ achieved.
tion, though not currently approved in the
Adults: hemodialysis‐dependent: 100 mg one
United States. Must wait 14 days after a dose
to three times/week during dialysis, total of
for reequilibration if retesting iron stores.
10 doses (1000 mg); may continue to admin­
ister at lowest dose possible to maintain tar­
IRON SUCROSE get hemoglobin and iron storage parameters.
Adults: non‐dialysis‐dependent chronic renal
Venofer
failure: 200 mg on 5 different days over a 2‐
Iron salt
week period (total dose 1000 mg).
Injection: 20 mg elemental iron/ml (2.5, 5,
10 ml)
Pregnancy category B Notes:
Do not give to patients with known hyper­
Indications: sensitivity to iron formulations, anemia not
Treatment of microcytic, hypochromic associated with iron deficiency, hemochro­
anemia resulting from iron deficiency in matosis, hemolytic anemia, or iron over­
chronic kidney disease patients, either dial­ load. Use with caution in patients with
ysis‐dependent or non‐dialysis‐dependent, history of significant allergies, asthma,
who may or may not be receiving hepatic impairment, or rheumatoid
erythropoietin. arthritis.
458 Formulary

ISOTRETINOIN mouth, dry nares and bleeding, headaches


13‐cis‐retinoic acid; Accutane, Amnesteem, (including potential development of pseudo­
Absorica, Claravis, Myorisan, Zenatane tumor cerebri), joint or muscle pain, hair
Anti‐neoplastic, retinoid loss, sunburn, constipation, and mood shifts.
Capsule: 10, 20, 30, and 40 mg soft gelatin
capsules KETOROLAC
Generic (previously available as Toradol)
Pregnancy category X
NSAID
Tablet: 10 mg
Indications:
Injection: 15, 30 mg/ml
Isotretinoin is a form of concentrated vita­
min A that is a standard part of neuroblas­ Pregnancy category C/D if used in third
toma treatment in combination with other trimester
immunotherapy drugs after completion of
chemotherapy, surgery, autologous trans­ Indications:
plant, and radiation therapy. Short‐term management of pain; systemic
treatment not to exceed 5 days regardless of
Dosage: route of administration.
160 mg/m2/day, divided into two daily doses,
for 14 consecutive days within each of six Dosage:
consecutive 28‐day cycles (see protocol). Children ≥2 years: 0.5 mg/kg/dose IM/IV
Isotretinoin can be given by mouth whole, q6–8h (maximum dose 30 mg q6h or 120 mg
buried in food whole, or dissolved and q24h).
mixed with food. Caregivers should wear Adult: 30 mg IV/IM q6h (maximum dose
disposable gloves to avoid touching the liq­ 120 mg/24 hour).
uid inside the capsule. Children >16 years (>50 kg) and adults:
10 mg PO q6h PRN (maximum dose
If the medicine is removed from inside the
40 mg/24 hour).
capsule and then mixed with food, it must
be given within 1 hour. Isotretinoin is
Notes:
extremely sensitive to light and air.
Isotretinoin is better absorbed by the body Do not give to patients with known hyper­
when taken with foods high in fat like whole sensitivity to ketorolac or other NSAIDs,
(4%) milk, ice cream, pudding, peanut but­ patients with active peptic ulcer disease, gas­
ter, or whipped cream. It does not mix well trointestinal bleeding or perforation, renal
with liquids made with water or juice. dysfunction, bleeding diathesis, thrombocy­
Parents may try any favorite food to help the topenia, or cerebrovascular bleeding. Use
child take the medicine. with caution in patients with congestive
heart failure, hypertension, or decreased
Notes: renal or hepatic function. May cause
impaired platelet function, nausea, dyspep­
Because of the dangers of birth defects with
sia, drowsiness, and interstitial nephritis.
isotretinoin, the patient, physician, and
pharmacist must register with the iPLEDGE™
program (www.ipledgeprogram.com). LEUCOVORIN CALCIUM
Adverse effects include severe dry peeling Generic
skin, dry eyes, itching, red/chapped lips, dry Antidote, methotrexate; folic acid derivative
Formulary 459

Tablets: 5, 10, 15, 25 mg administer intrathecal or intraventricular


Injection: 50, 100, 200, 350 mg vials (may be harmful or fatal).
Pregnancy category C
LOMUSTINE
Indications: CCNU
CeeNU, Gleostine, generic
Reduction of toxic effects (leucovorin res­ Antineoplastic, alkylating agent
cue) of high‐dose methotrexate, to counter­ Capsules: 10, 40, 100 mg
act effects of impaired methotrexate
elimination, or as an antidote for folic acid Pregnancy category D
antagonist overdose. Indicated for the treat­
ment of folate‐deficient megaloblastic ane­ Indications:
mia of infancy, sprue, or pregnancy; Treatment of primary or metastatic brain
treatment of nutritional deficiencies when tumors and Hodgkin lymphoma.
oral folate therapy is not possible.
Dosage:
Dosage: Refer to individual protocol.
Refer to individual protocol. Usual dose is 75–130 mg/m2 PO as a single
Rescue dose (following administration of dose every 6 weeks; subsequent doses
high‐dose methotrexate: 15 mg/m2 IV to adjusted per platelet and leukocyte counts.
start, then 15 mg/m2 q6h; dependent on
protocol, leucovorin may be increased to Notes:
100–1000 mg/m2 based on methotrexate Do not give to patients with known hyper­
level/clearance. sensitivity to lomustine. May need to adjust
Folate‐deficient megaloblastic anemia: 1 mg/day dose due to prolonged myelosuppression.
IM/IV. Delayed pulmonary fibrosis may occur with
high cumulative doses (i.e., ≥1 g/m2).
Megaloblastic anemia secondary to congeni-
tal deficiency of dihydrofolate reductase:
3–6 mg/day IM. MECHLORETHAMINE
Folic acid antagonist (e.g., pyrimethamine, Mustargen
trimethoprim [TMP]) overdose: 5–15 mg/ Antineoplastic, alkylating agent
day PO for 3 days or until the blood counts Injection: 10 mg
are normal or 5 mg every 3 days; doses of Pregnancy category D
6 mg/day are needed for patients with plate­
let counts <100 × 109/l. Indications:
Following intrathecal (IT) methotrexate Treatment of Hodgkin and non‐Hodgkin
(Down syndrome): 5 mg/m2/dose PO/IV at lymphoma and brain tumors; sclerosing
48 hours and 60 hours post IT 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 Children: MOPP regimen (Mustargen
ondary to vitamin B12 deficiency. Do not [mechlorethamine], Oncovin [vincristine],
460 Formulary

procarbazine, prednisone): 6 mg/m2 IV on secondary leukemia. May lead to amenor­


days 1 and 6 of a 28‐day cycle. rhea. Causes bone marrow suppression; use
Brain tumors: MOPP regimen: 3 mg/m2 IV with caution in those with prior myelosup­
on days 1 and 8 of a 28‐day cycle. pressive chemotherapy or radiation therapy.
Intracavitary (adults): 10–30 mg or 0.2– Use with caution and consider dose reduc­
0.4 mg/kg. tion in patients with renal impairment. May
increase the effects/levels of cyclosporine
Notes: and carmustine.
Do not give to patients with known hyper­
sensitivity to mechlorethamine, preexisting MERCAPTOPURINE
profound myelosuppression, active infec­ 6‐MP, 6‐mercaptopurine
tion, or pregnancy. Extravasation may result Purinethol
in severe tissue damage; treat promptly with Antineoplastic, antimetabolite; purine
cold compresses (6–12 hours) and sodium antagonist
thiosulfate. Mechlorethamine is highly Tablet: 50 mg
toxic, handle with care; consider benefits of
Pregnancy category D
treatment carefully.

MELPHALAN Indications:

Alkeran Used in conjunction with methotrexate for


Antineoplastic, alkylating agent maintenance therapy in ALL; combination
Tablet: 2 mg regimen in acute myelogenous and CMLs;
Injection: 50 mg non‐Hodgkin lymphoma.

Pregnancy category D Dosage:


Refer to individual protocol.
Indications: 50–100 mg/m2 PO once daily. Many proto­
Treatment of multiple myeloma, epithelial cols have dosing charts by BSA, including
carcinoma of the ovary; palliation in refrac­ recommended dose modifications for toxic­
tory cancers; conditioning prior to HSCT. ity or thiopurine methyltransferase
(TPMT)/NUDT15 metabolization status
Dosage: (based on genotyping).
Refer to individual protocol.
HSCT conditioning: 70–100 mg/m2 IV on Notes:
days 7 and 6 prior to HSCT; or 140–220 mg/ Do not give to patients with known hyper­
m2 IV single dose prior to HSCT; or 50 mg/ sensitivity to mercaptopurine, severe liver
m2/day × 4 days; or 70 mg/m2/day × 3 days. disease, or severe bone marrow suppression.
Children: 4–20 mg/m2/day PO days 1–21. Use with caution and adjust dose in patients
with renal or hepatic dysfunction. Patients
Notes: who receive allopurinol concurrently should
Do not give to patients with known hyper­ have their mercaptopurine dose reduced by
sensitivity or to those whose disease was 33–50%.
resistant to prior therapy. Long‐term oral 6‐MP and 6‐thioguanine (6‐TG) are methyl­
therapy and high cumulative doses (i.e., ated by TPMT to an inactive metabolite.
>600 mg) can increase the incidence of TPMT activity varies significantly among
Formulary 461

patients. Approximately 1 in 300 patients is 15 minutes before alkylator then q3h for three
completely deficient in the TPMT enzyme to six doses. Total daily Mesna dose ranges
(homozygous) and 10% have intermediate from 60 to 160% of the daily alkylator dose.
activity (heterozygous). Patients who have IV continuous infusion of Mesna is given at
low TPMT are most susceptible to toxicity doses equivalent to 60–100% of the ifosfa­
(primarily manifested as severe/prolonged mide or cyclophosphamide dose.
myelosuppression) due to higher concentra­
Mesna is given by IV infusion over 15–30 min­
tions of thioguanine nucleotides (TGNs).
utes or by continuous IV infusion, or per
Homozygous patients may tolerate less than
protocol.
10% of 6‐MP protocol dosing and approxi­
mately 35% of heterozygotes will require
Notes:
lower doses of 6‐MP to avoid dose limiting
toxicity with resultant interruption in dosing. Do not give to patients with known hyper­
Germline mutations resulting in the NUDT15 sensitivity to Mesna or thiol compounds.
polymorphism may also lead to intolerance of May cause false positive urinary ketone
protocol dosing. Many protocols recommend measurements.
TPMT genotyping to predict potential intol­
erance. For patients with high transaminases, METHADONE HYDROCHLORIDE
concern regarding compliance or elevated Dolophine, Methadose, generics
6MP dosing without resulting decrement in Antidote, analgesic
ANC, thiopurine metabolites can provide Tablet: 5, 10 mg
helpful information in regard to 6‐TGN and Solution: 5 mg/5 ml, 10 mg/5 ml
6‐MMPN metabolization and assist in guid­ Concentrated solution: 10 mg/ml
ing treatment decisions. Injection: 10 mg/ml (20 ml)
Pregnancy category C
MESNA
Mesnex Indications:
Prophylaxis, cyclophosphamide, or ifosfa­ Management of severe pain; used in nar­
mide‐induced hemorrhagic cystitis. cotic detoxification maintenance programs
Tablet: 400 mg and for the treatment of iatrogenic narcotic
Injection: 100 mg/ml dependency.
Pregnancy category B Dosage: Analgesia

Indications: Children: 0.7 mg/kg/24 hours divided q4–6h


PO/SC/IM/IV PRN (maximum dose 10 mg/
Detoxifying agent used to inhibit hemor­ dose).
rhagic cystitis induced by ifosfamide and
cyclophosphamide. Adults:
2.5–10 mg/dose q3–4 hours PO/IM/IV/SC
Dosage: PRN.
Refer to individual protocol. Detoxification: see PDR: 15–40 mg/day PO.
Mesna dose is 20% of alkylator dose IV
15 minutes before or combined with alkyla­ Notes:
tor, followed by repeat doses 4 and 8h later; Do not give to patients with known hypersen­
for high‐dose alkylator therapy, give dose sitivity to methadone. Use with caution in
462 Formulary

patients with respiratory disease as respiratory 100 mg/m2 IV bolus every 10 days (Interim
depression lasts longer than analgesic effects. Maintenance), with dose escalation up to
May cause cardiac arrhythmias (must follow 450 mg/m2 per protocol.
for QTc prolongation), sedation, increased Doses >500 mg/m2 require leucovorin
intracranial pressure, hypotension, and brady­ rescue.
cardia, in addition to respiratory depression.
Osteosarcoma/solid tumors:
Prolonged half‐life; average 19 hours in chil­
dren and 35 hours in adults. Peak effect is much Children <12 years: 12 g/m2 IV over 4 hours
more rapid with IV dosing. Repeated use can (dose range 12–18 g) with leucovorin rescue.
result in cumulative effects necessitating Children ≥12 years: 8 g/m2 IV over 4 hours
adjustment to the dose and frequency of (maximum dose 18 g) with leucovorin
administration. Duration of action PO is rescue.
6–8 hours initially and then 22–48 hours after Non‐Hodgkin lymphoma: 200–500 mg/m2
repeated dosing. Methadone is a substrate for IV; repeat every 4 weeks, as per protocol.
CYP450 3A4, 2D6, and 1A2, and inhibitor of
CNS leukemia (and prophylaxis):
206. Conversion from other narcotics to meth­
adone can be quite challenging; discussion Dosed by age:
with a provider with experience in this area is Children <1 year: 6 mg.
warranted (see Chapter 28). Children 1 to <2 years: 8 mg.
Children 2 to <3 years: 10 mg.
METHOTREXATE
Children 3 to <9 years: 12 mg.
Rheumatrex, Trexall, generic
≥9 years: 15 mg.
Antineoplastic, antimetabolite, antirheu­
matic; folic acid antagonist Use preservative‐free formulation for IT
Tablet (Trexall): 2.5, 5, 7.5, 10, 15 mg administration. Dilute in 0.9% NaCl or
Injection: 1 g vial Elliotts B solution for a total volume of
5–10 ml.
Pregnancy category X
Intrathecal methotrexate may be combined
with other agents for IT administration (i.e.,
Indications:
cytarabine, hydrocortisone).
Treatment of ALL (including CNS leuke­
mia), trophoblastic neoplasms, osteosar­
Notes:
coma, non‐Hodgkin lymphoma, rheumatoid
arthritis, dermatomyositis, and psoriasis. Do not give to patients with known hyper­
sensitivity to methotrexate, severe renal or
Dosage: hepatic impairment, or preexisting pro­
Refer to individual protocol. found bone marrow suppression. High‐dose
Acute lymphoblastic leukemia (refer to proto- methotrexate (>1 g/m2) should not be
col, phase for specific dosing, modifications, administered to patients with a creatinine
and frequency): clearance of less than 50–75% of normal.
Patients should receive alkaline fluids to
Starting doses:
maintain urine pH of 7 or higher while
20 mg/m2 PO once per week, may be held in receiving high‐dose methotrexate. Follow
weeks intrathecal methotrexate is given; serum levels per protocol and administer
1000–5000 mg/m2 bolus dosing or by continu­ leucovorin rescue per protocol. Methotrexate
ous infusion IV over 6–42 hours (dependent has been associated with acute and severe
on phase of therapy); chronic hepatotoxicity, severe bone marrow
Formulary 463

suppression, and renal failure with delayed deficiency; treatment and prevention of
clearance, high‐dose administration, con­ ifosfamide‐induced encephalopathy.
current nephrotoxic drugs, or inadequate
hydration. See treatment with Glucarpidase Dosage:
in the event of acute renal failure and Children and adults: Methemoglobinemia:
delayed clearance resulting in toxic levels. 1–2 mg/kg (25–50 mg/m2) IV as a single
Intrathecal and parenteral administration of dose over 5 minutes; may be repeated after 1
methotrexate have been associated with hour if needed.
acute neurotoxicity. Severe dermatologic
reactions and radiation dermatitis have
Notes:
been reported. May accumulate in fluid col­
lections (pleural effusions, ascites) increas­ Use with caution in patients with G6PD
ing local toxicity. deficiency or renal insufficiency. May cause
nausea, vomiting, dizziness, headache,
Ensure no TMP–sulfamethoxazole (SMX),
abdominal pain, diaphoresis, phototoxicity,
penicillin, NSAIDs, or PPIs are given until the
and skin staining. May cause transient blue‐
methotrexate level is <0.1 μm. Urinary acidi-
green coloration of urine and stool. At high
fiers may cause precipitation of methotrexate
doses, may cause methemoglobinemia.
in the urinary tract.
When IT and parenteral dosing of metho­
METHYLPREDNISOLONE
trexate is scheduled for the same day, deliver
the IT therapy within 6 hours of the begin­ Medrol, Medrol Dosepak, Depo‐Medrol,
ning of the IV infusion. Solu‐Medrol, generics
Patients with Down syndrome may have Corticosteroid
protocol‐specific adjustments or limitations Tablets: 2, 4, 8, 16, 32 mg
in dosing. Current ALL trials do not include Tablets (dose pack, generics): 4 mg
HD MTX infusions in interim maintenance Injection, sodium succinate (Solu‐Medrol,
and leucovorin (5 mg/m2/dose at 48 and generics): 40, 125, 500, 1000, 2000 mg (IV/
60 hours post IT MTX) is recommended IM)
during all phases except maintenance. Injection (acetate) (Depo‐Medrol, generics):
20, 40, 80 mg/ml (IM; not for IV use)
Intrathecal drugs should be prepared and
administered separately from other IV Pregnancy category C
chemotherapeutic drugs to avoid inappro­
priate administration. Indications:

METHYLENE BLUE Anti‐inflammatory or immunosuppressive


agent used to treat a variety of diseases of
ProvayBlue, generics
hematologic, allergic, inflammatory, neo­
Antidote, drug‐induced methemoglobinemia
plastic, and autoimmune origin.
Tablet (Urolene Blue): 65 mg
Injection: 10 mg/ml
Dosage:
Pregnancy category X Anti‐inflammatory/immunosuppressive: 0.5–
1.7 mg/kg/24 hours PO/IM/IV divided
Indications: q6–12h.
Antidote for cyanide poisoning and Chemotherapy (IV Solu‐Medrol, generics):
drug‐induced methemoglobinemia; treat­ Refer to individual protocols for dosing (in
ment of NADPH‐methemoglobin reductase lieu of prednisone; convert dose for steroid
464 Formulary

potency, 80% of prednisone dose; note some Must be given with diphenhydramine to
protocols recommend 1:1 conversion). reduce incidence of extrapyramidal symp­
toms. Consensus guidelines now advocate
Notes: for initial 1 mg/kg dose (with diphenhy­
dramine) followed by 0.0375 mg/kg dose
Do not give to patients with hypersensitivity
q6h subsequently (does not have to be given
to methylprednisolone. Do not administer
with diphenhydramine).
with live‐virus vaccines or during active
infection with varicella or herpes zoster.
Notes:
Avoid use in patients with systemic fungal
infections. May cause hypertension, glucose Do not give to patients with known hyper­
intolerance, gastrointestinal bleeding, osteo­ sensitivity to metoclopramide, gastrointesti­
porosis, pseudotumor cerebri, Cushing’s nal obstruction, pheochromocytoma, or
syndrome, adrenal axis suppression, and history of seizure disorder. Sedation, head­
acne. May increase levels of cyclosporine ache, anxiety, depression, leukopenia, and
and tacrolimus. diarrhea may occur. Reduce dose in renal
impairment.
METOCLOPRAMIDE
Reglan, Metozolv, generics MICAFUNGIN
Antiemetic Mycamine
Tablets: 5, 10 mg Antifungal, echinocandin
Tablets (orally disintegrating [ODT], Injection: 50, 100 mg
Metozolv, and generics): 5, 10 mg
Syrup: 5 mg/5 ml Pregnancy category C
Injection: 5 mg/ml
Indications:
Pregnancy category B
Treatment of patients with candidemia,
Indications: esophageal candidiasis, disseminated can­
didiasis; prophylaxis of Candida infections
Treatment of gastroesophageal reflux (GER) in patients undergoing HSCT. Not effective
and prevention of nausea and vomiting against cryptococcus, fusariosis, and zygo­
associated with chemotherapy. mycosis. Fungistatic again Aspergillus.

Dosage: Dosage:
GER or gastrointestinal dysmotility: Give Limited data in neonates and infants.
30 minutes before meals and at bedtime. Guidelines are per pharmacokinetic studies,
Infants and children: 0.1–0.2 mg/kg/dose up short duration trials, and case reports.
to QID PO/IV/IM (maximum dose 0.8 mg/
Prophylaxis of Candida infections in HSCT
kg/24 hours or 10 mg/dose).
recipients:
Adults: 10–15 mg/dose on awakening,
before each meal (QAC), and at bedtime Children and adolescents (<50 kg): 1.5 mg/
(QHS) PO/IV/IM kg/day IV once daily (maximum dose
Antiemetic: All ages. 50 mg/dose).
1‐mg/kg/dose q6h PO/IV/IM. Give first Adolescents/adults (≥50 kg): 50 mg IV once
dose 30 minutes prior to emetogenic drug. daily.
Formulary 465

Disseminated candidiasis: Dosage:


Neonates <1 kg: 10–15 mg/kg IV once daily. Refer to individual protocol.
Neonates ≥1 kg: 7–10 mg/kg IV once daily; Leukemia:
10–12 mg/kg IV once daily for HIV infected/
Children ≥2 years, BSA < 0.6 m2: 0.4 mg/kg
exposed neonates.
IV daily for 35 days.
Children and adolescents (<50 kg): 3–4
Children >2 years and adults: 12 mg/m2 IV
mg/kg IV once daily (maximum dose
over 10 minutes daily for 2–3 days.
200 mg/dose).
Acute leukemia in relapse: 8–12 mg/m2 IV
Adolescents/adults (>50 kg): 100–150 mg IV
daily for 5 days;
once daily.
AML: 10 mg/m2 IV daily for 3–5 days.
Aspergillosis, esophageal candidiasis:
Solid tumors:
Children and adolescents (<50 kg): 3–4 mg/
kg IV once daily (doses up to 7.5 mg/kg have Children: 18–20 mg/m2 IV q3–4 weeks or
been tolerated in invasive aspergillosis). 5–8 mg/m2 IV weekly.
Adolescents/adults (≥50 kg): 150 mg IV once Adults: 12–14 mg/m2 IV q3–4 weeks (maxi­
daily (mean duration of therapy in esopha­ mum total 80–120 mg/m2).
geal candidiasis 15 days, range 10–30 days).
Notes:
Notes: Do not give to patients with known hyper­
Do not give to patients with known hyper­ sensitivity to mitoxantrone. May cause
sensitivity to micafungin or other echino­ severe myelosuppression; use with caution
candins. Use with caution in patients with in patients with preexisting myelosuppres­
renal or hepatic impairment and in patients sion. Irreversible myocardial toxicity may
receiving concomitant hepatotoxic drugs; occur as the cumulative dosage approaches
monitor for evidence of worsening. May 550 mg/m2 doxorubicin equivalents (or
cause electrolyte disturbances, cytopenias, 400 mg/m2 with chest irradiation or con­
skin rash, central nervous system, and car­ comitant cyclophosphamide administra­
diovascular effects. Micafungin is a CYP450 tion). This may be an acute or late effect and
3A isoenzyme substrate and weak inhibitor, cardiac monitoring with ECHO, measure­
may increase effects/toxicity of nifedipine ment of LVEF, and ECG is highly recom­
and sirolimus. mended during therapy as well as months to
years following cessation of therapy.
MITOXANTRONE Extravasation may lead to severe tissue
damage. Dosage may need to be reduced in
Novantrone, generic
patients with impaired hepatobiliary func­
Antineoplastic, anthracenedione
tion, preexisting bone marrow suppression,
Injection: 2 mg/ml
or previous treatment with cardiotoxic
Pregnancy category D drugs or chest radiation.

Indications: MORPHINE SULFATE


Treatment of AML and multiple sclerosis; Roxanol, Oramorph SR, MS Contin, Avinza,
active in pediatric sarcoma, Hodgkin and Kadian, generics
non‐Hodgkin lymphoma, ALL, and MDS. Narcotic, analgesic
466 Formulary

Tablets: 15, 30 mg PO: 10–30 mg PO q4h PRN (immediate


Controlled‐release tablets (MS Contin, release) or 15–30 mg q8–12h PRN (con­
Oramorph SR): 15, 30, 60 mg (100 and trolled release).
200 mg for opioid‐tolerant patients) IM/IV/SC: 2–15/dose q2–6h PRN.
Extended‐release capsules (Avinza): 30 mg
Patient‐controlled analgesia (PCA) IV dosing
(60, 90, 120 mg for opioid tolerant patients)
guidelines:
Sustained‐release pellets in capsules (Kadian,
generics): 10, 20, 30, 50, 60 mg (100, 200 mg PCA (on demand) dose: 0.01–0.04 mg/kg.
for opioid tolerant patients) Suggested lock out (between PCA doses):
Solution: 10, 20, 100 mg/5 ml 10–20 minutes.
Suppository: 5, 10, 20, 30 mg Continuous IV dose:
Injection: 0.5, 1, 2, 3, 4, 5, 8, 10, 15, 25,
Neonate: 0.01–0.02 mg/kg/hour.
50 mg/ml
Infant/child: 0.01–0.07 mg/kg/hour.
Pregnancy category C/D (prolonged use or
Adult: 0.8–10 mg/hour (higher doses with
in higher doses at term)
tolerance).
Indications: Bolus (loading): 0.03–0.1 mg/kg.
The 1 hour maximum is 0.1–0.15 mg/kg
Relief of moderate to severe pain, acute and
(6–8 mg/hour in tolerant adults), and typi­
chronic, after non‐narcotic analgesics have
cally equals the continuous dose plus two to
failed; preanesthetic medication; relief of
three PCA bolus doses.
dyspnea from acute left ventricular failure
and pulmonary edema.
Notes:
Dosage: Do not give to patients with severe renal or
liver insufficiency. May cause severe pruri­
Dose should be titrated to effect.
tus, urinary retention, respiratory depres­
Neonates: Analgesia: 0.05–0.2 mg/kg/dose sion, central nervous system depression,
IM/slow IV/SC q4h. nausea, constipation, and ileus. Prolonged
Opiate withdrawal: 0.08–0.2 mg/kg/dose PO use can result in physical and psychological
q3–4h PRN. dependence. Consider nighttime low con­
Infant 1–6 months: tinuous dose infusion of 0.01–0.02 mg/kg/
hour without a change in lock out so as to
PO: 0.08–0.1 mg/kg/dose q3–4h PRN. allow sleep. Avoid narcotization with seda­
IV: 0.025–0.03 mg/kg/dose q2–4h PRN. tion and respiratory depression. Monitor
Infant > 6 months and children: vitals and encourage use of incentive
PO: 0.2–0.5 mg/kg/dose (initial maximum spirometry and ambulation with repetitive
dose 15–20 mg/dose) q4–6h PRN (immediate dosing or PCA use. Naloxone may be used
release) or 0.3–0.6 mg/kg/dose q12h PRN to reverse effects of respiratory depression.
(controlled release). Low‐dose naloxone has been used to coun­
ter effects of pruritus and urinary retention.
IM/IV/SC: 0.1–0.2 mg/kg/dose q2–4h PRN
(maximum initial dose infant 2 mg/dose;
1–6 years 4 mg/dose; 7–12 years 8 mg/dose; MYCOPHENOLATE MOFETIL (MMF)
>12 years 10 mg/dose) CellCept, Myfortic, generic (capsule, tablet)
Adults: Immunosuppressive agent
Formulary 467

Capsule, as mofetil or CellCept: 250 mg with the other oral dosage forms on a mg to
Tablet, as mofetil or CellCept: 500 mg mg basis. Do not give to patients with known
Tablet, delayed release, as mycophenolic acid hypersensitivity to MMF. Common side
Myfortic: 180, 360 mg (not recommended effects include headache, hypertension, vom­
for children whose total body surface area iting, diarrhea, abdominal pain, bone marrow
[TBSA] is <1.19 m2) suppression, fever, and opportunistic infec­
Injection, as mofetil or CellCept: 500 mg tions. Risk appears to be associated with the
Powder for oral suspension, CellCept: 200 mg/ml intensity and duration of immunosuppres­
sion; may result in an increased incidence of
Pregnancy category D
lymphoma or other malignancies especially in
combination with other immunosuppres­
Indications: sants. Patients should avoid excessive expo­
sure to sunlight and should use sun protection
Used as an immunosuppressant drug fre­
factor. Use with caution and adjust dose in
quently in combination with other immuno­
patients with renal impairment.
suppressants (e.g., cyclosporine, corticosteroids)
for the prophylaxis of organ rejection (renal,
hepatic, cardiac, and BMT), chronic GVHD, NALOXONE
myasthenia gravis, proliferative lupus nephritis,
Narcan, Evzio, generics
and relapsing nephrotic syndrome.
Narcotic antagonist
Injection: 0.4 mg/ml
Dosage: Injection in syringe: 2 mg/2 ml
Limited data are available for pediatric dos­ Autoinjector (Evzio): 0.4 mg/0.4 ml
ing. Pharmacokinetic studies have indicated Nasal liquid (Narcan): 4 mg/0.1 ml
that doses based on BSA result in a more
Pregnancy category C
appropriate AUC than doses based on body
weight.
Indications:
Children and adolescents: 600 mg/m2/dose PO/
Used to reverse central nervous system and
IV BID (maximum dose 2000 mg/24 hours) or
respiratory depression in suspected narcotic
alternatively dose by BSA if ≥1.25 m2 as
overdose; treatment of coma of unknown
follows:
etiology.
1.25–1.5 m2: 750 mg PO BID.
>1.5 m2: 1 g PO BID. Dosage:
Adult (in combination with corticosteroids Treatment of opiate intoxication: May be
and cyclosporine per specific transplant pro- given via endotracheal tube (ETT), use two
tocol): 2000–3000 mg/24 hours PO/IV to ten times IV dose).
divided BID or Neonates, children <5 years or <20 kg: 0.1 mg/
Delayed release tabs (Myfortic): 720– kg/dose; repeat q2–3 minutes PRN IV/IM/SC.
1080 mg PO BID. Children >5 years or ≥20 kg: 2 mg/dose; if no
response, repeat q2–3 minutes PRN IV/IM/SC.
Notes:
Adults: 0.4–2.0 mg/dose q2–3 minutes PRN
Check specific transplant protocol for dosing. IV/IM/SC. Use in 0.1–0.2 mg increments in
Due to differences in bioavailability, the opioid‐dependent patients. If no response and
delayed release tablets are not interchangeable cumulative dose >10 mg, reevaluate diagnosis.
468 Formulary

IV continuous infusion (children and adults): Notes:


0.005 mg/kg loading dose, then 0.0025– Do not give to patients with known hyper­
0.16 mg/kg/hour. Taper gradually to avoid sensitivity to nelarabine. May cause severe
relapse. neurotoxicity, including severe somnolence,
PCA side effect reversal (pruritus and/or seizures, confusion, ataxia, and peripheral
urinary retention with morphine): neuropathy. Observe closely for neurotoxic­
IV continuous infusion: Begin with 1 mcg/ ity. Adverse effects associated with demyeli­
kg/hour, titrate up or down to 0.25–2 mcg/ nation or similar to Guillain–Barré syndrome
kg/hour to abate opioid‐related side effects; (ascending peripheral neuropathies) have
taper infusion gradually over 2–4 hours been reported. Neurotoxicity is dose‐limited
when discontinuing. and may not reverse completely. Risk of neu­
rotoxicity may increase in patients with con­
Notes: current intrathecal chemotherapy or history
of cranial irradiation. May cause bone mar­
Do not give to patients with known hyper­
row suppression. Use with caution in patients
sensitivity to naloxone. Short duration of
with renal or hepatic impairment. Monitor
action may necessitate repeated doses. The
blood counts, electrolytes, renal function,
dose for pediatric postoperative narcotic
and transaminases frequently.
reversal is one‐tenth of the dose for opiate
intoxication. Endotracheal administration
NILOTINIB
can be done safely by diluting in 1–2 ml of
normal saline. Will produce narcotic with­ Tasigna
drawal in patients with dependence. Use TKI of BCR–ABL
with caution in patients with chronic heart Capsules: 50, 150, 200 mg
disease. Abrupt reversal of narcotic depend­ Pregnancy category D
ency may result in nausea, vomiting, dia­
phoresis, tachycardia, hypertension, and Indications:
tremulousness.
Treatment of adult CML newly diagnosed
NELARABINE chronic phase, or chronic and accelerated
phases not responsive to imatinib; pediatric
Arranon
CML in chronic phase, newly diagnosed or
Antineoplastic, antimetabolite; purine
resistant/intolerant of prior TKI therapy.
nucleoside analog
Injection: 5 mg/ml; 250 mg vial
Dosage:
Pregnancy category D Pediatric dose: 230 mg/m2 PO twice daily,
rounded to the nearest 50 mg dose (to a
Indications: maximum single dose of 400 mg).
Treatment of relapsed or refractory T‐cell Adult dose: 400 mg PO twice daily.
ALL and T‐cell lymphoblastic lymphoma.
Notes:
Dosage:
Patients must have typical BCR–ABL tran­
Refer to individual protocol.
scripts. Contraindicated in patients with
Children: 650 mg/m2 IV daily, for 5 consecu­ hypokalemia, hypomagnesemia, or long
tive days; repeat per protocol. QTc syndrome. Most common adverse
Adults: 1500 mg/m2/dose IV on days 1, 3, events include nausea, vomiting, headache,
and 5; repeat per protocol. fatigue, pruritus, rash, arthralgia, pyrexia,
Formulary 469

night sweats, and diarrhea. Serious adverse Notes:


effects include myelosuppression, electro­
Do not give to patients with known hyper­
lyte abnormalities (hypophosphatemia,
sensitivity to ondansetron. Ondansetron is a
hypokalemia, hyponatremia, hypocalce­
substrate for the cytochrome CYP450
mia), pancreatitis and elevated serum lipase,
enzyme, so inducers or inhibitors of this sys­
hemorrhage, fluid retention, cardiovascular
tem may affect the elimination of ondanse­
and arterial occlusive events, and growth
tron. May need to adjust dose and interval
and developmental retardation in children.
for severe hepatic impairment. Side effects
are usually mild, with headache, sedation,
ONDANSETRON constipation, and dry mouth being the most
Zofran, Zofran ODT, Zuplenz, generics common. May also cause bronchospasm,
Antiemetic, serotonin 5‐HT3 receptor tachycardia, hypokalemia, seizures, light­
antagonist headedness, diarrhea, transient increases in
Tablets: 4, 8, 24 mg bilirubin or transaminases, and transient
Tablets (ODT): 4, 8 mg blindness (minutes to 48 hours). ECG
Solution: 4 mg/5 ml changes (QTc prolongation) have been
Injection: 2 mg/ml reported. Give 1–2 hours prior to radiation;
Pregnancy category B may need around the clock dosing for
abdominal radiation.
Indications:
OSELTAMIVIR
Prevention of nausea and vomiting associ­
ated with initial and repeat courses of eme­ Tamiflu, generics
togenic cancer chemotherapy or radiation, Antiviral
prevention of PONV, treatment of emesis Capsules: 30, 45, 75 mg
induced by acute gastroenteritis. Oral suspension: 6 mg/ml
Pregnancy category C
Dosage:
Prevention of chemotherapy or radiation‐ Indications:
induced nausea and vomiting:
Treatment of influenza A and B strains (ini­
IV: 0.15 mg/kg/dose 30 minutes prior to the
tiate therapy within 2 days of onset of
start of emetogenic chemotherapy, with sub­
symptoms).
sequent doses administered q8h until chem­
otherapy is complete. Maximum single dose
Dosage:
is 16 mg, though is generally maxed at 8 mg
with q8h dosing. Children ≥1–12 years:
PO/IV: Dose based on BSA or age: <15 kg: 30 mg PO BID for 5 days.
<0.3 m2: 1 mg TID PRN. >15–23 kg: 45 mg PO BID for 5 days.
0.3–0.6 m : 2 mg TID PRN.
2 >23–40 kg: 60 mg PO BID for 5 days.
0.6–1 m2: 3 mg TID PRN. >40 kg: 75 mg PO BID for 5 days.
>1 m : 4–8 mg TID PRN.
2 Children >13 years to adult: 75 mg PO BID
for 5 days.
Age:
<4 years: Dose based on BSA. Notes:
4–11 years: 4 mg TID PRN. The most common adverse symptoms are
>11 years to adult: 8 mg TID. nausea and vomiting. Reduce dosage in
470 Formulary

renal impairment. Not for use in children derivatives to limit myelosuppression and
<1 years due to animal studies suggesting enhance efficacy.
increased CNS penetrance and fatalities.
PALONOSETRON
OXALIPLATIN
Aloxi
Eloxatin, generic
Anti‐emetic, serotonin 5‐HT3 receptor
Antineoplastic, alkylating agent
antagonist
Injection: 5 mg/ml
Tablets: 0.5 mg capsule
Pregnancy category D Injection: 5 mL vial (0.25 mg)
Pregnancy category B
Indications:
Treatment of colon carcinoma, relapsed or Indications:
refractory solid tumors, brain tumors, and
Prevention of CINV in patients receiving ini­
non‐Hodgkin lymphoma.
tial and repeat courses of moderate to highly
emetogenic regimens, especially if previous
Dosage:
history or high risk of delayed nausea.
Refer to individual protocol.
Children: ≤1 year: 4.3 mg/kg IV over 2 hours Dosage:
q3 weeks. Ages 1 month to <17 years: 20 μg/kg IV 30 min­
Children: >1 year: 130 mg/m2 IV over 2 hours utes prior to chemotherapy.
q3 weeks. Adult dosing: 0.5 mg capsule PO 1 hour prior
to chemotherapy.
Notes:
Do not give to patients with known hyper­ Notes:
sensitivity to oxaliplatin, in pregnancy, or in Contraindicated in known hypersensitivity.
those with grade 3 or 4 neuropathy (usually May cause headache or constipation. Use with
due to prior exposure). Anaphylaxis may caution in combination with other serotonin
occur within minutes of administration; receptor antagonists due to risk for serotonin
appropriate supportive and resuscitative syndrome. Given the extended half‐life, gen­
medications and equipment should be avail­ erally given as a single dose prior to chemo­
able. Two different types of neuropathy may therapy initiation, though may be repeated
occur: (i) an acute (within 2 days) reversible after 72 hours. Should be considered in
(resolves within 14 days) sensory neuropa­ patients with significant, refractory delayed
thy with peripheral symptoms that are often nausea/vomiting (particularly from platinum
exacerbated by cold (may include pharyngo­ chemotherapy) in lieu of ondansetron or
laryngeal dysesthesia), and (ii) persistent granisetron and especially if unable to receive
(over 14 days) sensory neuropathy that pre­ decadron (i.e., brain tumors) or aprepitant
sents with paresthesias, dysesthesias, (i.e., potential drug interactions).
hypoesthesias, and impaired proprioception
that interferes with activities of daily living. PAZOPANIB
Symptoms may improve with discontinuing
Votrient
treatment. May cause pulmonary fibrosis or
Antineoplastic, kinase inhibitor
hepatotoxicity. When administered as
Tablet: 200 mg
sequential infusions, taxane derivatives
should be administered before platinum Pregnancy category D
Formulary 471

Indications: Treatment of PCP: 4 mg/kg/dose IM/IV


Treatment of advanced soft tissue sarcomas, daily for 14–21 days (preferably IV).
rhabdomyosarcoma, and renal cell carcinoma.
Notes:
Dosage: Do not give to patients with known hyper­
sensitivity to pentamidine isethionate.
Refer to individual protocol. Safety and effi­
Adjust dose in renal impairment. Use with
cacy in pediatrics have not been established.
caution in patients with diabetes mellitus,
Adult: 800 mg po QD renal or hepatic dysfunction, hypertension,
or hypotension. Additive toxicity may occur
Notes: with aminoglycosides, amphotericin B, cis­
Most common adverse reactions are nausea, platin, and vancomycin. Can see Jarisch–
vomiting, diarrhea, hair color changes, Herxheimer‐like reaction (fever, chills,
fatigue, decreased appetite, decreased headache, myalgia). Inhalation therapy may
weight, hypertension, tumor pain, headache, cause irritation of the airway, bronchos­
myalgia, dysgeusia, gastrointestinal pain, pasm, cough, oxygen desaturation, dyspnea,
and dyspnea. Use with caution with other and loss of appetite. May cause hypoglyce­
medications, CYP34A inducers and inhibi­ mia, hyperglycemia, hypotension (with
tors. Avoid proton pump inhibitors and H2 infusion <2 hours), nausea, vomiting, fever,
antagonists; separate antacids and pazopanib mild hepatotoxicity, pancreatitis, hypocalce­
dosing by several hours. mia, megaloblastic anemia, granulocytope­
nia, and nephrotoxicity. Unclear efficacy of
PENTAMIDINE ISETHIONATE IV/IM route.
Pentam 300, NebuPent
Antibiotic, antiprotozoal POSACONAZOLE
Injection: 300 mg vial (Pentam 300) Noxafil
Inhalation: 300 mg vial (NebuPent) Antifungal, azole
Injection: 300 mg/16.7 ml
Pregnancy category C
Tablet (delayed release): 100 mg
Suspension: 40 mg/ml
Indications:
Treatment of PCP in patients who cannot Pregnancy category C
tolerate or who fail to respond to TMP–
SMX, prevention of PCP infection in immu­ Indications:
nocompromised hosts, treatment of African Prophylaxis of invasive Aspergillus and
trypanosomiasis, and treatment of visceral Candida infections in high‐risk, severely
and cutaneous leishmaniasis caused by immunocompromised patients (e.g., HSCT
Leishmania donovani. recipient, GVHD, hematologic malignancy
with prolonged chemotherapy‐induced
Dosage: neutropenia); treatment of oropharyngeal
Prophylaxis: 4 mg/kg/24 hours IM/IV (IV candidiasis (including those refractory to
preferred route, over 1–2 hours) q2–4 weeks; echinocandins and/or fluconazole).
maximum single dose 300 mg.
Dosage:
Inhalation (≥5 years), use with Respigard II
nebulizer: 300 mg in 6 ml water via inhala­ Injection form has not been approved for
tion qmonth. <18 years of age. Oral solution and tablets
472 Formulary

are not interchangeable due to differences immunosuppressant effects in autoimmune


in dosing. diseases such as immune thrombocytopenia
Prophylaxis of invasive Aspergillus and purpura; chemotherapy for ALL and
Candida infections: Hodgkin and non‐Hodgkin lymphoma.
Children ≥13 years and adults:
Dosage:
Oral solution: 200 mg PO TID.
Dose is dependent on condition being
DR tablet: 300 mg PO BID on day 1, fol­ treated and response of patient. Consider
lowed by 300 mg PO QD. alternate day dosing for long‐term therapy.
≥18 years: Option to use injection form: Discontinuation of long‐term therapy
300 mg IV BID on day 1, followed by 300 mg requires gradual tapering.
IV QD. Anti‐inflammatory or immunosuppressant
Length of treatment is dependent on recov­ (includes ITP, aplastic anemia): 0.5–2 mg/
ery of immune suppression. kg/24 hours PO divided one to three times
Treatment of oropharyngeal candidiasis: daily.
Initial: 100 mg BID (oral solution) on day 1, Chemotherapy: 40–180 mg/m2/24 hours, as
then 100 mg daily for 13 days. per protocol.
Refractory: 400 mg BID (oral solution).
Notes:
Notes: Avoid use in patients with life‐threatening
infections (except septic shock or tubercu­
Do not give to patients with known hyper­ lous meningitis), systemic fungal infections,
sensitivity to posaconazole and avoid con­ varicella, and in those with hypersensitivity
current administration with ergot alkaloids. to prednisone. Use with caution in patients
Cross‐sensitivity reactions with other azoles with hypothyroidism, cirrhosis, hyperten­
have not yet been studied but may exist. May sion, congestive heart failure, ulcerative
cause hepatotoxicity, and frequent monitor­ ­colitis, gastrointestinal bleeding, and throm­
ing is recommended. May alter drug levels boembolic disorders. May cause adrenal axis
of cyclosporine (also tacrolimus and siroli­ suppression, hypertension, hyperglycemia,
mus) resulting in nephrotoxicity, leukoen­ irritability, gastritis, skin atrophy, osteoporo­
cephalopathy, and death. Concomitant sis, cataracts, fluid retention, mood lability,
utilization with vincristine may lead to nausea, diarrhea, bone pain, acne, and weight
increased neurotoxicity. gain. Consider use of antacid in long‐term
therapy. Avascular necrosis and growth retar­
PREDNISONE dation are seen in patients with long‐term or
Deltasone, Rayos, generics repeated high‐dose therapy. Ensure patients
Corticosteroid are aware of risk of varicella infection in non‐
Tablets: 1, 2.5, 5, 10, 20, 50 mg immune states.
Solution: 1, 5 mg/ml
Pregnancy category C/D PROCARBAZINE HYDROCHLORIDE
Matulane
Indications: Antineoplastic, alkylating agent
Capsule: 50 mg
Management of adrenocortical insuffi­
ciency; used for anti‐inflammatory or Pregnancy category D
Formulary 473

Indications: lymphoma, or a select group of patients with


Treatment of advanced Hodgkin lymphoma solid tumors at risk for tumor lysis syndrome
and brain tumors. with initiation of chemotherapy. Indicated
only for short‐term use; maximum of 5 days.
Dosage:
Dosage:
Refer to individual protocol.
50–100 mg/m2 PO daily for 10–14 days. 0.05–0.2 mg/kg/dose IV over 30 minutes
(round down to nearest 1.5 mg vial). Patients
Notes: usually respond to one dose, but may repeat
q24h PRN up to four additional doses.
Avoid in patients with known hypersensitiv­
Give prior to chemotherapy if expected
ity to procarbazine or with preexisting bone
massive tumor lysis and/or renal involve­
marrow aplasia. Delay treatment for 1 month
ment/dysfunction (e.g., Burkitt lymphoma
or longer following radiation due to potenti­
with bulky disease), especially if evidence of
ating effect on marrow suppression. May
tumor lysis or renal compromise.
cause nausea, vomiting, dry mouth, consti­
pation, headache, dizziness, and hair loss.
Notes:
Azoospermia and infertility have been
reported when procarbazine has been given Do not give to patients with known hyper­
in combination with other antineoplastics. sensitivity to rasburicase. Contraindicated in
Use with caution and reduce dose in patients patients with G6PD deficiency (may cause
with renal or hepatic impairment or marrow hemolysis) or severe asthma.
suppression. May potentiate central nervous Methemoglobinemia has been reported with
system depression when used with pheno­ use. May cause vomiting, nausea, fever,
thiazine derivatives, barbiturates, narcotics, headache, abdominal pain, constipation,
alcohol, tricyclic antidepressants, and diarrhea, and rash. When measuring serum
methyldopa. Drug (e.g., monoamine oxidase uric acid levels, ensure tubes are prechilled,
inhibitors) and food interactions are com­ contain heparin, are placed in an ice water
mon. Avoid food with high tyramine content bath, and analyzed within 4 hours. Collection
(i.e., aged cheese or meats, yogurt, tea, dark at room temperature may lead to spuriously
beer, coffee, cola drinks, wine, soybean prod­ low uric acid levels due to continued degra­
ucts, peanuts, avocadoes, bananas) because dation by rasburicase in the sample.
hypertensive crisis, tremor, excitation, car­
diac palpitations, and angina may occur. Rho(D) IMMUNE GLOBULIN
WinRho SDF, Rhophylac
RASBURICASE Immune globulin
Elitek Injection: 1500, 2500, 5000, 15 000 IU
Recombinant urate oxidase, uric acid lower­ (1 mcg = 5 IU)
ing agent, antigout agent Prefilled injection (Rhophylac): 1500 IU (2 ml)
Injection: 1.5, 7.5 mg vials
Pregnancy category C
Pregnancy category C
Indications:
Indications: Treatment of ITP in non‐splenectomized
Initial management or prevention of hyper­ Rho(D)‐positive patients; suppression of Rho(D)
uricemia in high‐risk patients with leukemia, isoimmunization in an Rho(D)‐negative
474 Formulary

individual exposed to Rho(D)‐positive blood or RITUXIMAB


during delivery (or pregnancy) of an Rho(D)‐ Rituxan
positive infant (if father known to be Rho(D)‐ Antineoplastic, monoclonal antibody
positive or status unknown). Injection: 10 mg/ml

Dosage: Pregnancy category C


ITP:
Indications:
Hemoglobin ≥10 g/dl: 50 mcg/kg IV (250 IU/
kg), administer dose over 3–5 minutes. Treatment of relapsed or refractory low‐
grade or follicular CD20 positive, B‐cell non‐
Hemoglobin <10 g/dl: 25–40 mcg/kg IV
Hodgkin lymphoma; first‐line therapy for
(125–200 IU/kg).
diffuse large B‐cell non‐Hodgkin lymphoma
New onset ITP: 75 mcg/kg (375 IU/kg) as a in combination with other chemotherapeu­
single dose has been used safely and effica­ tics; systemic autoimmune disorders includ­
ciously in pediatric patients with platelets ing autoimmune hemolytic anemia and
≤20 × 109/l. Subsequent dosing may be immune thrombocytopenia purpura; post­
adjusted (125–3000 IU/kg) dependent on transplant lymphoproliferative disorder;
response. rheumatoid arthritis; refractory GVHD;
Patients who do not respond to an initial Epstein Barr Virus (EBV) reactivation;
dose can be re‐dosed, and if hemoglobin refractory systemic lupus erythematosus.
>10 g/dl can increase to 250–300 IU/kg).
Dose frequency is dependent on duration of Dosage:
response, clinical symptoms, and adverse Children and adults: 375 mg/m2 IV qweek
effects; may be given q2–4 weeks. for 4 weeks (range 3–6 weeks).
Courses may be repeated for treatment of
Notes: malignancies or recurrent symptoms with
Avoid in patients with known hypersensitiv­ autoimmune disorders.
ity to immunoglobulins or thimerosal and
in patients with IgA deficiency. Not recom­ Notes:
mended for use in patients with hemoglobin Refer to individual protocol for treatment
<8 g/dl. Adverse events associated with doses and frequency. Rare cases of progres­
administration in ITP include headache, sive multifocal leukoencephalopathy due to
chills, fever, and reduction in hemoglobin Creutzfeldt–Jakob virus have been reported.
(resulting from the destruction of Rho(D)‐ Infusional toxicity is common and primarily
positive red cells). May interfere with avoided with a slow infusion and premedi­
immune‐response to live‐virus vaccines. cation. Most commonly, infusional toxicity
Causes acute hemolysis with an average is seen with the first infusion and reactions
hemoglobin decrease of 1.5–2 g/dl at 1 week. may include development of hives, bron­
Do not use in patients with active bleeding. chospasm, hypoxia, hypotension, pulmo­
Current FDA guidelines strongly recom­ nary infiltrates, and respiratory distress
mend that patients be monitored in a health syndrome. Some reactions have been fatal.
care center for 8 hours after a dose of Rho(D) Close monitoring is required for all infu­
immune globulin, with urinalyses at base­ sions and appropriate resuscitative medica­
line and 2, 4, and 8 hours after administra­ tions and equipment must be close by. In the
tion due to risk of severe hemolysis. case of mild reactions, temporary cessation
Formulary 475

of the infusion is indicated with acetami­ Dosage:


nophen, diphenhydramine, and fluids as Safety and efficacy have not yet been estab­
indicated. Hydrocortisone may also be indi­ lished in pediatrics, but clinical trials are
cated. If symptoms resolve, reinitiate the ongoing. In patients ≥18 years of age, the fol­
infusion at a 50% reduced rate and monitor lowing doses are recommended:
closely. Subsequent infusions should run
DVT or PE treatment: 15 mg PO q12h for
slowly, and the patient should be premedi­
21 days, then 20 mg PO daily.
cated. Rituximab should be discontinued
altogether following a life‐threatening reac­ Reduction of recurrence risk of DVT/PE in
tion. Use with caution in patients with pre­ at‐risk patients following ≥6 months initial
existing pulmonary or cardiac conditions treatment with anticoagulation: 10 mg PO
and in those at risk for development of daily.
tumor lysis syndrome. Causes immunosup­
pression. Quantitative immunoglobulins Notes:
and specific antibody response should be This is the first available direct oral antico­
checked prior to and following treatment agulant (DOAC) inhibiting factor Xa. The
with rituximab to determine if immunoglob­ maximum inhibition of factor Xa is 4 hours
ulin infusions might be needed posttreat­ following a dose, with effects lasting
ment until recovery of immune function. 8–24 hours and necessitating only once daily
Reactivation of hepatitis B has been noted administration. Inhibition of factor Xa
(mainly in adult patients); thus, immune sta­ interrupts the intrinsic and extrinsic coagu­
tus to hepatitis B virus should be docu­ lation cascades, inhibiting both thrombin
mented. Although specific to B‐cells, PCP formation and development of thrombi.
infection after rituximab has been reported; Rivaroxaban does not inhibit thrombin
therefore, patients should receive PCP (activated factor II) and has no effect on
prophylaxis for 6 months or until recovery of platelet function. Dose adjustments and
immune function is documented. routine coagulation monitoring are not
needed, nor are dietary restrictions needed.
RIVAROXABAN Adverse effects include bleeding, including
Xarelto severe intestinal bleeding. A reversal agent is
Anticoagulant available (Andexanet alfa/AndexXa), though
Tablet: 2.5, 10, 15, 20 mg bleeding may still be difficult to control.
Contraindicated in patients with severe liver
Pregnancy category C or end‐stage renal disease.

Indications: ROMIPLOSTIM
Indicated for treatment of DVT and PE; Nplate
reduction in risk of recurrence and/or PE in Thrombopoietin receptor agonist
patients at continued risk for recurrence Injection: 250, 500 mcg vial
after completion of initial treatment lasting
≤6 months; reduction of stroke risk and sys­ Pregnancy category C
temic embolism in patients with non‐valvu­
lar atrial fibrillation; prophylaxis of DVT Indications:
which may lead to PE in patients undergo­ Treatment of thrombocytopenia in patients
ing knee or hip surgery. with chronic immune thrombocytopenia
476 Formulary

who have had an insufficient response to Colony‐stimulating factor


corticosteroids, immunoglobulins, or sple­ Injection: 250, 500 mcg
nectomy. It should be used only in patients
Pregnancy category C
with ITP whose degree of thrombocytope­
nia and clinical condition increase the risk
Indications:
for bleeding, and not to normalize a platelet
count. Used to stimulate Ch14.18 antibody in high‐
risk neuroblastoma patients.
Dosage:
Dosage:
Safety and efficacy have not been estab­
250 mcg/m2 daily SC/IV (SC preferred
lished in pediatrics. Dosing is for ≥18 years.
route) before, during, and after Ch14.18
Initial dose: 1 mcg/kg SC based on ABW.
antibody infusion (see protocol for details).
Obtain weekly CBCs during dose adjust­ May be given differently in relapse setting.
ment until stable platelet count ≥50 × 109/l is
achieved for ≥4 weeks without further Notes:
changes in dose.
May lead to excessive immature myeloid cells
If platelet count is <50 × 109/l, increase by
in the peripheral blood or bone marrow
1 mcg/kg.
(>10%) as well as fever and bone pain. Avoid
If platelet count is >200 × 109/l for 2 consec­
in patients with a history of ITP, known hyper­
utive weeks, reduce by 1 mcg/kg.
sensitivity to GM‐CSFs, or yeast‐derived
products. Use with caution in patients with
If platelet count is >400 × 109/l, DO NOT
autoimmune or chronic inflammatory condi­
DOSE. Assess platelet count weekly. After
tions, hypertension, cardiovascular disease,
platelet count is <200 × 109/l, resume at
pulmonary disease, or renal or hepatic impair­
dose decreased by 1 mcg/kg.
ment. May need to be held with excessively
If platelet count is 50–200 × 109/l, maintain high white blood cell count (i.e., 50 × 109/l).
dose and assess CBCs weekly until stable
platelet count ≥ 50 × 109/l is achieved for SIROLIMUS
≥4 weeks without further changes in dose.
Rapamune, generics
After achieving stable platelet count and sta­ Immunosuppressant agent
ble dose, obtain monthly CBCs. Solution: 1 mg/ml
Tablet: 0.5, 1, 2 mg
Notes:
Pregnancy category C
Most common adverse reactions are head­
ache, arthralgia, dizziness, insomnia, myal­
Indications:
gia, extremity pain, abdominal pain,
shoulder pain, dyspepsia, and paresthesia. GVHD prophylaxis in HSCT recipients;
Administration may increase the risk for prophylaxis and treatment of rejection in
development or progression of reticulin renal transplant patients; primary immuno­
fiber formation within the bone marrow, suppression in other organ transplant.
which may improve upon discontinuation.
Dosage:
SARGRAMOSTIM Refer to individual protocol.
GM‐CSF Children ≥13 years of age and <40 kg:
Leukine Loading dose 3 mg/m2/dose PO on day 1
Formulary 477

immediately after transplant, followed by Dosage:


maintenance 1 mg/m2/24 h divided q12– Chemotherapy infiltration: Children and
24h; adjust dose to achieve target sirolimus Adults:
trough blood concentration. Mechlorethamine: Use 2 ml for each mg
Children >40 kg to adults: 6 mg PO once infiltrated.
immediately following transplant, followed
by 2 mg PO once daily. Notes:
Do not give to patients with known hyper­
Notes: sensitivity to sodium thiosulfate. Inject
slowly, over at least 10 minutes; rapid
Do not give to patients with known hyper­
administration may cause hypotension.
sensitivity to sirolimus. Refer to individual
protocol for optimal dosing and target
SUCCIMER
trough concentrations. Monitor whole
blood trough levels (just prior to dose at Chemet (2,3‐dimercaptosuccinic acid;
steady state) especially with concurrent use DMSA)
of CYP450 3A4 and/or P‐gp inducers and Antidote (lead toxicity), chelating agent
inhibitors. Steady state is usually achieved Capsule: 100 mg
at 5–7 days. Immunosuppression may result Pregnancy category C
in increased susceptibility to infection. May
increase risk for development of lymphoma Indications:
or other malignancy. Avoid excessive sun
exposure and use sun protection factor. Treatment of lead poisoning in asympto­
Severe hypersensitivity and skin reactions matic children with blood levels between 45
have been reported. Monitor renal and and 69.9 mcg/dl. Can be considered at lower
hepatic function; dose reduction may be lead levels (20–44.9 mcg/dl) if aggressive
necessary. Use with caution in the periop­ environmental interventions have not
erative period due to an increased risk of impacted level, although not noted to
surgical complications from wound dehis­ improve neurocognitive outcome.
cence and impaired wound and tissue heal­
ing. Prophylaxis against P. jiroveci (carinii) Dosage:
and CMV is recommended. Can lead to Lead chelation, children: 10 mg/kg/dose
hypertension, electrolyte wasting, renal (350 mg/m2/dose) PO q8h for 5 days, fol­
insufficiency, and transplant‐associated lowed by 10 mg/kg/dose PO q12h for
microangiopathy. 14 days. Repeat courses separated by a mini-
mum of 2 weeks may be necessary to treat
rebound lead concentrations (resulting
SODIUM THIOSULFATE
from mobilization of lead from bone stores).
Versiclear
Antidote, extravasation of mechlore­ Notes:
thamine, cisplatin, or cyanide; antifungal
Do not give to patients with known hyper­
agent (topical)
sensitivity to succimer. Use with caution in
Injection: 100, 250 mg/ml
impaired hepatic or renal function. May see
gastrointestinal symptoms, increased
Indications: transaminases, rash, headache, and dizziness.
Treatment of extravasations of selected Do not administer with other chelators. Treat
chemotherapeutic agents. iron deficiency and eliminate environmental
478 Formulary

exposure to lead. Monitor lead levels. May Notes:


sprinkle contents of the capsules on food if
Do not give to patients with known hypersen­
unable to swallow.
sitivity to tacrolimus, castor oil, or any compo­
nent. Refer to specific transplant protocol for
TACROLIMUS
dosing guidelines and therapeutic monitoring.
Prograf, Astagraf XL, Envarsus XR, FK506, Monitor trough levels just prior to steady state
generics (generally achieved after 2–5 days of dosing).
Immunosuppressant agent Oral dosing should be given 1 hour prior or 2
Capsule (Prograf, generics): 0.5, 1, 5 mg hours after a meal. Immunosuppression may
increase risk of infection or development of
Extended release capsule (Astagraf XL): 0.5,
lymphoma or other malignancies. Risk is
1, 5 mg
related to intensity and duration of use. May
Extended release tablet (Envarsus XR): 0.75, cause nephrotoxicity and neurotoxicity. Do
1, 4 mg not administer concurrently with cyclo­
Injection (Prograf): 5 mg/ml sporine. Monitor electrolytes, magnesium
wasting is common. Monitor tacrolimus drug
Indications: levels and adjust dosages per protocol.
Hypertension may lead to PRES; therefore,
Prevention of organ rejection in solid organ blood pressure must be followed closely and
transplant patients; prevention or treatment treated promptly if high. Transplant‐associ­
of GVHD in allogeneic HSCT. ated microangiopathy is also possible.

Dosage: TEMOZOLOMIDE
Children: Younger children usually require Temodar
higher dosing on a mg/kg basis than older Antineoplastic, alkylating agent
children. Capsule: 5, 20, 100, 140, 180, 250 mg
Solid organ transplant: Injection: 100 mg
Children: Initial dose 0.15–0.2 mg/ Pregnancy category D
kg/24 hours PO divided q12h or IV continu­
ous infusion 0.03–0.05 mg/kg/24 hours. Indications:
Titrate to therapeutic level.
Adult: Initial dose 0.075–0.2 mg/kg/24 hours Treatment of refractory anaplastic astrocy­
divided q12h or 0.01–0.05 mg/kg/24 hours toma; treatment of newly diagnosed glioblas­
by IV continuous infusion. Titrate to thera­ toma multiforme; active against recurrent
peutic level. glioblastoma multiforme, metastatic mela­
noma, brain stem glioma, ependymoma,
Prevention of GVHD: Initial 0.03 mg/ medulloblastoma, primitive neuroectoder­
kg/24 hours (based on LBW) IV continuous mal tumor (PNET), neuroblastoma, Ewing
infusion. Begin 24 hours prior to stem cell sarcoma, and anaplastic oligodendroglioma.
infusion and continue until oral medication
can be tolerated. May occasionally be given
Dosage:
as a q12h IV infusion.
Conversion from IV to PO dose (1:4 ratio): Refer to individual protocol.
Multiply total daily IV dose by 4 and admin­ Children: 100–200 mg/m2 IV/PO once daily
ister in two divided oral doses per day. for 5 days every 28 days.
Formulary 479

Metronomic dosing: 75 mg/m2/day 5 days/ hepatic impairment. May cause nausea, vom­
week, 21–42 day cycles. iting, anorexia, stomatitis, diarrhea, myelo­
May also be administered concurrently with suppression, and veno‐occlusive disease
radiation therapy. (sinusoidal obstructive syndrome). Increases
busulfan toxicity.
Notes:
Do not give to patients with known hyper­ THIOTEPA
sensitivity to temozolomide, dacarbazine, or Thioplex, generic
any component. Thrombocytopenia and Antineoplastic, alkylating agent
neutropenia are dose‐limiting toxicities and Powder for injection: 15, mg
may occur late in the treatment cycle and take
14 days to resolve. Monitor blood counts and Pregnancy category D
ensure platelet count ≥100 × 109/l and ANC
≥1.5 × 109/l prior to initiation of each cycle of Indications:
therapy. Rare cases of aplastic anemia, myelo­
dysplasia, and secondary malignancies have Treatment of superficial tumors of the blad­
been reported. Prophylaxis against P. jiroveci der, brain tumors, and other CNS neoplasms
is required, especially with concurrent (including intrathecal administration); con­
administration of irradiation. trol of pleural, pericardial, or peritoneal
effusions caused by metastatic tumors; also
used in high‐dose regimens with autologous
THIOGUANINE
HSCT.
6‐TG
Antineoplastic, antimetabolite; purine Dosage:
antagonist
Tablet (scored): 40 mg Refer to individual protocol.
Children, HSCT: 300 mg/m2/dose over
Pregnancy category D 3 hours IV; repeat q24h for three doses.
Maximum tolerated dose over 3 days is 900–
Indications: 1125 mg/m2.
Induction and consolidation phases in Intrathecal: 5–11.5 mg/m2 per dose weekly
AML; delayed intensification phase in ALL. for 2–7 doses.

Dosage: Notes:
Refer to individual protocol. Do not give to patients with known hyper­
sensitivity to thiotepa or severe myelosup­
Infants and children <3 years: 3.3 mg/kg/day
pression. Reduce dose in patients with renal,
PO divided q12h for 4 days.
hepatic, or bone marrow dysfunction. May
Children ≥3 years and adults: 50–200 mg/m2/ cause central nervous system changes, skin
day PO divided q12–24 hours for 4–14 days hyperpigmentation, nausea, vomiting,
or per protocol. hematuria, and elevation of liver transami­
nases and bilirubin. Due to skin excretion
Notes: and risk for irritation, frequent bathing is
Do not give to patients with known hyper­ required until 48 hours after completion of
sensitivity to thioguanine. Use with caution thiotepa when given as part of a transplant
and reduce dosage in patients with renal or preparative regimen.
480 Formulary

THROMBIN, TOPICAL reactions. Topical use only. Not for adminis­


Evithrom, Recothrom, Thrombi‐Gel, tration systemically or directly into sites of
Thrombi‐Pad, Thrombin‐JMI (epistaxis kit, brisk arterial bleeding. Avoid in patients with
spray kit) factor V deficiency due to cross‐reactivity.
Hemostatic agent
TOPOTECAN
Powder (recombinant, Recothrom): 1000 IU
Powder, topical (bovine, Thrombin‐JMI): Hycamtin
Available in epistaxis kit 5000 IU, spray Antineoplastic, camptothecin, topoisomer­
20,000 IU ase inhibitor
Solution, topical (human origin, Evithrom): Capsule: 0.25, 1 mg
800–1200 IU/ml Injection: 4 mg
Sponge, topical (bovine, Thrombi‐Gel): Pregnancy category D
1000 IU, 20,000 IU
Pad, topical (bovine, Thrombi‐Pad): 200 IU Indications:
Pregnancy category C Treatment of pediatric solid tumors, includ­
ing sarcoma and neuroblastoma.
Indications:
Evithrom, Recothrom, Thrombin‐JMI: As an Dosage:
aid to hemostasis whenever oozing blood, Refer to individual protocol.
and minor bleeding from capillaries and
Safety and efficacy have not been estab­
small venules is accessible and control of
lished in pediatrics.
bleeding by standard surgical techniques is
ineffective or impractical. Children:
Thrombi‐Gel, Thrombi‐Pad: As a trauma Pediatric solid tumors: 1 mg/m2/day (range
dressing for temporary control of moder­ 0.75–0.9 mg/m2/day) for 3 days as a continu­
ately to severely bleeding wounds and for ous IV infusion; repeat q3 weeks.
the control of surface bleeding from vascu­ Single agent therapy for refractory solid
lar access sites and percutaneous catheters tumors or hematologic malignancy: 2.4 mg/
and tubes. m2/day PO once daily for 5 days of a 21‐day
course.
Dosage: Combination therapy for solid tumors:
Apply powder directly to the site of bleeding 0.75 mg/m2/dose PO once daily for 5 days
or on oozing surface, or use 1000–2000 IU/ every 21 days in combination with
ml of solution where bleeding is profuse. cyclophosphamide.
May be diluted with NS to other concentra­
tions as needed. Use 100 IU/ml for bleeding Notes:
from skin or mucosal surfaces. May utilize Do not give to patients with known hyper­
epistaxis/spray kit for mucosal/nosebleeds. sensitivity to topotecan. Bone marrow toxic­
ity is dose‐limiting, primarily neutropenia.
Notes: Has been associated with neutropenic coli­
Avoid in patients with known hypersensitiv­ tis; should be a strong consideration in
ity to thrombin. Some forms are of human or patients with neutropenia, fever, and
bovine origin. May cause fever or allergic abdominal pain. Severe diarrhea has been
Formulary 481

reported, may require dose adjustment. Use caution in patients with cardiovascular or
with caution in patients with renal or hepatic cerebrovascular disease. Dose modification
impairment. Extravasation can cause tissue is required in patients with renal impair­
injury. ment. May cause hypotension, thromboem­
bolic complications, headache, and visual
TRANEXAMIC ACID abnormalities (seen in animals). Do not
Lysteda, generic administer concomitantly with factor IX
Antifibrinolytic, antihemophilic, hemo­ and PCC or hormonal contraception due to
static agent increased risk of thrombosis. Use with cau­
Tablet (Lysteda): 650 mg tion in patients with upper urinary tract
Injection: 100 mg/ml bleeding due to potential for clot formation
and ureteral obstruction. Use with extreme
Pregnancy category B caution in patients with disseminated intra­
vascular coagulation.
Indications:
Short‐term use (up to 5 days) to treat men­ TRETINOIN
orrhagia primarily associated with von Vesanoid, all trans‐retinoic acid (ATRA),
Willebrand disease. Has been used to treat Retin‐A, generic
or prevent mucosal bleeding (including Antineoplastic, retinoid
tooth extraction) in patients with mild Capsule: 10 mg
hemophilia or von Willebrand disease
(maximum 2–8 days). May be used to treat Pregnancy category D
recurrent epistaxis, to prevent hemorrhage
Indications: Tretinoin is used to induce
following trauma, delivery, and postpartum
remission in people with APL who have
blood loss, and to decrease perioperative
the PML/RARα gene mutation. The topi-
blood loss and need for transfusion in
cal form is used to treat acne.
patients undergoing repair of congenital
heart disease, craniosynostosis, or scoliosis
surgery. Dosage:
See specific protocol for dosing, doses may
Dosage: differ when combined with other medica­
tions including arsenic trioxide.
Children and adults:
Tooth extraction in patients with hemophilia: APL Induction/Consolidation:
10 mg/kg immediately before oral surgery
Children ≥1 year and adolescents: 45 mg/
IV, then 10 mg/kg/dose IV/PO q6–8h; may
m2/24 hours PO in two divided doses after
be used for 2–8 days.
meals until complete remission is docu­
Menorrhagia: 1300 mg PO TID for up to 5 mented. Therapy should be discontinued
days during monthly menstruation. 30 days after remission or after 90 days of
treatment, whichever occurs first.
Notes: Adults: 45 mg/m2/24 hours PO in two
Do not use in patients with known sub‐ equally divided doses until complete remis­
arachnoid hemorrhage, active intravascular sion is documented. Therapy should be dis­
clotting process, history of seizures, or continued 30 days after remission or after
acquired defective color vision. Use with 90 days of treatment, whichever occurs first.
482 Formulary

APL Maintenance: Antibiotic, sulfonamide derivative


Children, adolescents, and adults: 45 mg/m / 2 Tablet, single strength: 80 mg TMP and
day PO daily or divided q12h alone or with 400 mg SMX Tablet, double strength: 160 mg
regimens containing 6‐MP and methotrex­ TMP and 800 mg SMX
ate for 1–2 years following induction and Suspension: 40 mg TMP/200 mg SMX per
consolidation therapy has been studied in 5 ml
randomized clinical trials. Injection: 16 mg TMP/80 mg SMX per ml
Pregnancy category D
Notes:
The oral form of the drug has boxed warn­ Indications:
ings concerning the risks of retinoic acid syn­ Oral treatment of urinary tract infection
drome (RA‐APL) and leukocytosis. Other (UTI) and otitis media; prophylaxis for
significant side effects include a risk of throm­ PCP; IV treatment of documented or sus­
bosis, benign intracranial hypertension in pected PCP infection in immunocompro­
children, hypercholesterolemia and/or hyper­ mised patients.
triglyceridemia, and liver damage.
Side effects include malaise, shivering, hem­ Dosage:
orrhage, infections, peripheral edema, pain, Dosage recommendations are based on the
chest discomfort, edema, disseminated TMP component. May be given PO or IV.
intravascular coagulation, weight increase,
injection site reactions, anorexia, weight Minor/moderate infection:
decrease, and myalgia.
Children >2 months: 8–12 mg TMP/
Respiratory side effects usually signify RA‐ kg/24 hours PO divided BID (maximum
APL, and include upper respiratory tract dis­ 160 mg/dose).
orders, dyspnea, respiratory insufficiency, Adults (>40 kg): 160 mg TMP/dose BID.
pleural effusion, pneumonia, rales, and expir­
atory wheezing. This is seen most commonly Severe infection/PCP:
when given in combination with arsenic tri­
oxide (see ATO, differentiation syndrome). Children and adults: 20 mg TMP/kg/24 hours
Immediate intervention is required with dex­ divided q6–8h PO/IV.
amethasone 10 mg q12h × 3 days or until
symptoms resolve. UTI or otitis media prophylaxis:
Also reported are earache or a feeling of full­ Children: 2–4 mg TMP/kg PO once daily.
ness in the ears, abdominal pain, diarrhea,
constipation, dyspepsia, and swollen belly. Prophylaxis for PCP:
Cardiovascular side effects include arrhyth­ Children >1 month: 5 mg TMP/kg/24 hours
mias, flushing, hypotension, hyperten­ PO divided q12h 2–3 consecutive days a
sion, phlebitis, and rarely cardiac failure or week (maximum dose 320 mg
arrest. Nervous system side effects include TMP/24 hours).
dizziness, paresthesias, anxiety, insomnia, Adolescent and adults: 160 mg PO twice
depression, and confusion. daily for 2–3 consecutive days/week.

TRIMETHOPRIM/ Notes:
SULFAMETHOXAZOLE Do not give to patients with known hyper­
Bactrim, Septra, Co‐trimoxazole, Sulfatrim, sensitivity to sulfa drugs or any component,
TMP–SMX, generics porphyria, or megaloblastic anemia due to
Formulary 483

folate deficiency. Do not use in infants Herpes labialis (cold sores): >12 years: 2 g q12h
under 2 months of age. Use with caution in for 1 day; initiated at earliest symptoms.
patients with G6PD deficiency and renal or
hepatic impairment. Serious adverse reac­ Immunocompromised children at risk for
tions include Stevens–Johnson syndrome, HSV or VZV infection with normal renal
toxic epidermal necrolysis, hepatic necrosis, function: (limited data) 15–30 mg/kg/dose
agranulocytosis, aplastic anemia, and other PO TID (maximum 2 g/dose).
blood dyscrasias. Discontinue with rash.
May need to be held temporarily in oncol­ Notes:
ogy patients who are on TMP–SMX for PCP Do not give to patients with known hyper­
prophylaxis and subsequently develop neu­ sensitivity to valacyclovir or acyclovir. Use
tropenia. Numerous drug interactions are with caution in patients with renal impair­
reported. TMP–SMX decreases the clear­ ment or those receiving concomitant nephro­
ance of warfarin and methotrexate, toxic drugs. Adjust dose in patients with
decreases serum cyclosporine concentra­ renal impairment. Monitor renal function.
tions, and increases the effect of sulfonylu­
reas, phenytoin, and thiopental. Do not give VALGANCICLOVIR
to patients within 24 hours of receiving high‐
Valcyte
dose methotrexate and until level is <0.1 μm
Trade and other names
due to competitive excretion and increased
Antiviral
risk of methotrexate toxicity due to delayed
Tablets: 450 mg
clearance.
Oral solution: 50 mg/ml
Pregnancy category D
VALACYCLOVIR
Valtrex, generics Indications:
Antiviral
Valganciclovir is indicated for the preven­
Tablets/caplet: 500 mg, 1 g
tion of CMV disease in kidney transplant
Oral suspension: 50 mg/ml
patients (4 months to 16 years of age) and
Pregnancy category B heart transplant patients (1 month to
16 years of age) at high risk for infection.
Indications:
Dosage:
Treatment of herpes zoster and HSV in
immunocompromised patients; treatment of For pediatric kidney transplant patients
varicella zoster virus (VZV) in immunocom­ 4 months to 16 years of age, the recom­
petent children (ages 2–18 years); treatment mended once daily PO mg dose
of HSV labialis in adolescents and adults. (7 × BSA × CrCl) should start within 10 days
of posttransplantation until 200 days
posttransplantation.
Dosage: For pediatric heart transplant patients
1 month to 16 years of age, the recommended
Varicella, immunocompetent: once daily PO mg dose (7 × BSA × CrCl)
2 to <18 years: 20 mg/kg/dose PO TID for should start within 10 days of transplanta­
5 days (maximum 1 g TID); initiate within tion until 100 days posttransplantation.
24 hours of onset of rash. The recommended once daily dosage is
Adults: 1 g/dose PO TID for 7 days within based on BSA and creatinine clearance
48–72 hours of onset of rash. (CrCl) derived from a modified Schwartz
484 Formulary

formula, and is calculated using the equa­ or if neutrophil counts are <1000/μl at the
tion provided in the following text: beginning of treatment.
Pediatric dose (mg) = 7 × BSA × CrCl. If the
calculated Schwartz creatinine clearance VINBLASTINE SULFATE
exceeds 150 ml/minute/1.73 m2, then a max­ Velban, generic
imum value of 150 ml/minute/1.73 m2 Antineoplastic, mitotic inhibitor, vinca
should be used in the equation. alkaloid
Injection: 1 mg/ml; 10 mg vial
Notes:
Pregnancy category D
Take tablets or oral solution with food.
Valganciclovir is frequently used in the set­ Indications:
ting of severe immune suppression associ­
ated with BMT for the prevention of CMV Treatment of Hodgkin and non‐Hodgkin
infection. Use with caution in renal impair­ lymphoma, pediatric brain tumors (glio­
ment, and dosing should be adjusted for mas), Langerhans cell histiocytosis, chorio­
changes in creatinine (see package insert for carcinoma, advanced testicular germ cell
dose calculation). Side effects may include: tumors, maintenance therapy in intermedi­
diarrhea, pyrexia, fatigue, nausea, tremor, ate risk rhabdomyosarcoma.
neutropenia, anemia, leukopenia, thrombo­
cytopenia, headache, insomnia, UTI, and Dosage:
vomiting.
Severe leukopenia, neutropenia, anemia, Refer to individual protocol.
thrombocytopenia, pancytopenia, and bone Hodgkin lymphoma: 2.5–6 mg/m2/dose IV
marrow failure including aplastic anemia once every 1–2 weeks for three to six cycles;
have been reported in patients treated with maximum weekly dose 12.5 mg/m2.
valganciclovir or ganciclovir. Valganciclovir Langerhans cell histiocytosis: 6 mg/m2/dose
should be avoided if the ANC is <500/μl, the IV once every 1–3 weeks.
platelet count is <25 × 109/l, or the hemo­
Brain tumors (low‐grade gliomas): 6 mg/m2/
globin is <8 g/dl. Use with caution in patients
dose IV weekly or biweekly.
with pre‐existing cytopenias and in patients
receiving myelosuppressive drugs or irradi­
Notes:
ation. Cytopenias may occur at any time
during treatment and may worsen with con­ Do not give to patients with known hyper­
tinued dosing. Cell counts usually begin to sensitivity to vinblastine or with severe leu­
recover 3–7 days after drug discontinuation. kopenia. Dose modification may be needed
In patients with severe leukopenia, neutro­ in patients with hepatic impairment or neu­
penia, anemia, and/or thrombocytopenia, rotoxicity. Extravasation may cause local
treatment with hematopoietic growth fac­ severe tissue damage. May cause peripheral
tors may be considered. neuropathy, myelosuppression, jaw pain,
Frequent monitoring of CBC with differen­ myalgia, paresthesia, constipation, abdomi­
tial as well as renal function should be per­ nal pain, ileus, and mild alopecia.
formed frequently, especially in infants, The metabolism of vinca alkaloids has been
patients with renal impairment, those in shown to be mediated by hepatic CYP450
whom ganciclovir or other nucleoside ana­ isoenzymes in the CYP3A subfamily. This
logs have previously resulted in leukopenia, metabolic pathway may be impaired in
Formulary 485

patients with hepatic dysfunction or who may cause local severe tissue damage. May
are taking concomitant potent inhibitors of cause peripheral neuropathy, paresthesias,
these isoenzymes. ileus, jaw pain, cranial nerve paralysis (pto­
Intrathecal administration is fatal. Vinblastine sis), vocal cord paralysis, hyponatremia,
should never be taken into the procedure room SIADH secretion, alopecia, and constipation.
for a patient undergoing a lumbar puncture The metabolism of vinca alkaloids has been
for instillation of intrathecal chemotherapy. shown to be mediated by hepatic CYP450
isoenzymes in the CYP3A subfamily. This
metabolic pathway may be impaired in
VINCRISTINE SULFATE
patients with hepatic dysfunction or in
Vincasar, Oncovin, generic those taking concomitant potent inhibitors
Antineoplastic, mitotic inhibitor, vinca of these isoenzymes.
alkaloid
Intrathecal administration is fatal. Vincristine
Injection: 1 mg/ml; 1, 2 mg vials
should never be taken into the procedure room
Pregnancy category D for a patient undergoing a lumbar puncture
for instillation of intrathecal chemotherapy.
Indications:
Treatment of ALL, Hodgkin and non‐Hodgkin VINORELBINE
lymphoma, neuroblastoma, brain tumors, Navelbine, generic
Wilms tumor, and rhabdomyosarcoma. Antineoplastic, mitotic inhibitor, vinca
alkaloid
Dosage: Injection: 10 mg/ml; 50 mg/5 ml vials
Refer to individual protocol.
Pregnancy category D
Children ≤10 kg or BSA <1 m2: 0.05 mg/kg/
dose IV once weekly; maximum single dose
2 mg. Indications:
Children >10 kg or BSA ≥1 m2: 1–2 mg/m2 Indicated for the treatment of non‐small cell
IV; may repeat weekly for 310 weeks. lung cancer and used off label for rhabdo­
Maximum single dose is 2 mg (some proto­ myosarcoma and other cancers.
cols allow for higher dosing [i.e., 2.8 mg] in
Hodgkin lymphoma). Dosage:
Neuroblastoma: IV continuous infusion Refer to individual protocol.
with doxorubicin: 1 mg/m2/day for 72 hours. 30 mg/m2 IV over 6–10 minutes weekly, or
per protocol.
Notes:
Notes:
Do not give to patients with known hyper­
sensitivity to vincristine. Avoid in patients Dose modifications may need to be made
with the demyelinating form of Charcot– for neutropenia and/or hyperbilirubinemia.
Marie–Tooth disease. Asparaginase may Side effects include peripheral neuropathy,
decrease clearance of vincristine. Dose modi­ cytopenias, immune suppression, constipa­
fication may be required in patients with tion, nausea, vomiting, and fatigue.
impaired hepatic function, preexisting neu­ Extravasation can cause severe local tissue
romuscular disease, or severe side effects of damage. Less common adverse effects
treatment with vincristine. Extravasation include hair loss and allergic reaction.
486 Formulary

The metabolism of vinca alkaloids has been Infants and children: 2.5–5 mg/24 hours PO
shown to be mediated by hepatic CYP450 iso­ or 1–2 mg/dose IV/IM/SC.
enzymes in the CYP3A subfamily. This meta­ Adolescents and adults: 2.5–25 mg/24 hours
bolic pathway may be impaired in patients with PO or 2.5–10 mg/dose IV/IM//SC once.
hepatic dysfunction or in those taking concom­
itant potent inhibitors of these isoenzymes.
Minor bleeding (any elevated INR):
<40 kg: 0.03 mg/kg PO once or 0.5–2.5 mg
VITAMIN K1/PHYTONADIONE IV once.
≥40 kg: 1–2.5 mg PO once or 0.5–2.5 mg IV
Mephyton, generics once.
Vitamin, water soluble
Tablet: 5 mg Repeat dosing q12–24h if bleeding persists
Suspension: 1 mg/ml and INR not corrected.
Injection: 2, 10 mg/ml
Significant bleeding (any elevated INR):
Pregnancy category C
5–10 mg IV.
Consider additional measures including
Indications: fresh frozen plasma (FFP) (10–15 ml/kg).
Prevention and treatment of hypoprothrom­ Monitor INR q4–6h, repeat dosing if full
binemia caused by anticoagulants or drug‐ correction not achieved at 12–24 hours and
induced vitamin K deficiency; hemorrhagic bleeding continues.
disease of the newborn.
Notes:
Dosage: Monitor INR until <4. For INR >10 monitor
Hemorrhagic disease of the newborn: q12h and repeat dose q12–24h until INR <4.
Prophylaxis: 0.5–1 mg IM within 1 hour of Do not give to patients with known hypersen­
birth (for preterm infants <1 kg 0.3–0.5 mg/kg). sitivity to phytonadione. Antagonizes action
Treatment: 1–2 mg/24 hours IV/IM/SC. of warfarin. Protect product from light.
Parenteral dosing may cause flushing, dizzi­
Oral anticoagulant (warfarin) overdose with- ness, cardiac or respiratory arrest, hypoten­
out significant bleeding, INR >4 <10: sion, or anaphylaxis. High doses (10–20 mg)
in neonates may cause hyperbilirubinemia
Infants and children (<40 kg): 0.03 mg/kg PO
and severe hemolytic anemia. Monitor pro­
once.
thrombin time (PT), APTT, INR; blood coag­
Adolescents and adults (≥40 kg): 1–2.5 mg
ulation factors may increase within 6–12 hours
PO once.
after oral dosing and within 1–2 hours after
parenteral dosing. Parenteral IM or IV dosing
Oral anticoagulant (warfarin) overdose with-
is indicated when the oral route is not feasible
out significant bleeding, INR ≥10:
in emergency situations.
Infants and children (<40 kg): 0.06 mg/kg PO
once, may repeat q12–24h.
Adolescents and adults (>40 kg): 5–10 mg PO VORICONAZOLE
once, may repeat q12–24h. Vfend, generics
Antifungal agent
Vitamin K deficiency (due to drugs, malab-
Injection: 200 mg
sorption, or decreased synthesis of vitamin K
by the liver): Oral suspension: 200 mg/5 ml
Formulary 487

Tablet: 50, 200 mg with rifampin, CYP3A4 substrates, and ergot


Pregnancy category D alkaloids. Voriconazole is metabolized by
CYP450 enzymes and many drug interac­
Indications: tions exist. May cause severe hepatic toxicity,
visual changes, cardiac arrhythmias, audi­
Treatment of invasive aspergillosis, especially
tory hallucinations, and photosensitivity.
in immunocompromised patients; treatment
May cause fetal harm; must ensure contra­
of candidemia in nonneutropenic patients;
ception to avoid pregnancy. Use with caution
invasive Candida infections including esoph­
in neonates; oral suspension contains
ageal; treatment of serious fungal infections
sodium benzoate, a metabolite of benzyl
caused by molds including Scedosporium or
alcohol, which may cause a potentially fatal
Fusarium species in patients intolerant or
toxicity. Use with caution in patients with
refractory to conventional antifungal therapy.
renal or hepatic toxicity; dose adjustment
may be necessary. Considerable interpatient
Dosage: variability; monitor trough levels and adjust
Invasive aspergillosis or candidiasis or other dosing as necessary. Limited data for use in
rare molds (Scedosporium or Fusarium): children <2 years. Due to risk of concomitant
Children 2–11 years: 7–9 mg/kg/dose PO/IV neurotoxicity with vincristine, should be
q12h (maximum initial dose 350 mg). held around vincristine doses (as feasible).
Children ≥12 years and adolescents (≥50 kg):
6 mg/kg/dose IV q12h × two doses, followed WARFARIN SODIUM
by 4 mg/kg/dose IV q12h. Coumadin, Jantoven, generics
Convert to oral therapy when significant Anticoagulant
clinical improvement after 1 week of IV Tablets: 1, 2, 2.5, 3, 4, 5, 6, 7.5, 10 mg
therapy at 9 mg/kg/dose PO q12h (maxi­ Pregnancy category D/X
mum dose 350 mg) or if ≥50 kg administer
200 mg PO q12h. Indications:
Esophageal candidiasis: Prophylaxis and treatment of venous throm­
Children 2–11 years: 4 mg/kg/dose q12h IV boembolic disorders; prevention of arterial
or 9 mg/kg/dose PO q12h (maximum dose thromboembolism in patients with pros­
350 mg q12h). thetic heart valves or atrial fibrillation; pre­
Children >12 years and adolescents (≥50 kg): vention of death, VTE, and recurrent
3 mg/kg/dose IV q12h or 200 mg PO q12h. myocardial infarction (MI) after acute MI.
Prophylaxis (pediatric acute leukemia):
Dosage:
6 mg/kg/dose PO q12h × two doses, fol­
lowed by 4 mg/kg/dose PO q12h. Infants and children (to achieve/maintain
INR between 2 and 3):
Notes: Initial dose: 0.2 mg/kg PO for 1–2 days; max­
imum dose 10 mg.
Do not give to patients with known hypersen­
sitivity to voriconazole. Use with caution in Maintenance dose: 0.1 mg/kg/24 hours PO
patients with known hypersensitivity to daily; range 0.05–0.34 mg/kg/24 hours (con­
azoles; cross‐reactivity studies have not been sult published charts based on INR).
done. Tablets may contain lactose and should Adults: 5–10 mg PO daily × 2–5 days, adjust
be used with caution in patients with lactose for desired INR; maintenance dose range
intolerance. Avoid concurrent administration 2–10 mg/day PO.
488 Formulary

Onset of action is within 36–72 hours and review all medications prior to initiation of
peak effects occur within 5–7 days. Monitor therapy. The INR is the recommended test
INR after 5–7 days of new dosage; high‐risk to monitor anticoagulant effect. The abso­
patients may require more frequent lute INR desired is dependent on the indica­
monitoring. tion and has been extrapolated from adults.
Usual duration of therapy for first venous An INR of two to three has been recom­
thrombotic event is 3 months (depending mended for prophylaxis and treatment of
on elimination of procoagulant factor and DVT, pulmonary emboli, and bioprosthetic
based on resolution of clot). heart valves. Younger children may require
higher dosing. Certain foods and medica­
tions may alter levels and should be reviewed
Notes:
in detail with the patient. Antidote is vitamin
Do not give to patients with known hyper­ K and PCC (fresh frozen plasma if PCC una-
sensitivity to warfarin, severe liver or kidney vailable). Due to time to effect, must bridge
disease, uncontrolled bleeding, gastrointes­ with anti‐Xa (i.e., enoxaparin) for 3 days.
tinal ulcers, status–post neurosurgical pro­
cedures, and malignant hypertension.
Concomitant use with vitamin K may References
decrease anticoagulant effect. Concomitant
use with aspirin, nonsteroidal anti‐inflam­ Hughes, H.K. and Kahl, L.K. (2018). The Johns
Hopkins Hospital: The Harriet Lane Handbook,
matory drugs, or indomethacin may
A Manual for Pediatric House Officers, 21e.
increase warfarin’s anticoagulant effect and Philadelphia, PA: Elsevier‐Mosby.
cause severe gastrointestinal irritation. May (2019). Physicians’ Desk Reference, 73e. PDR
cause fever, skin lesions, necrosis (especially Network.
in protein C deficiency), hemorrhage, hem­ Taketomo, C.K., Hodding, J.H., and Kraus, D.M.
optysis, anorexia, nausea, vomiting, and (2016). Pediatric & Neonatal Dosage
diarrhea. Many drug interactions exist; Handbook, 23e. Lexi‐Comp.
Index

abnormal hemoglobins, 27, 53 risk group classification, 201–203


absolute neutrophil count (ANC), 119, 333, 334, treatment, 203–204
394, 401, 432 twins and siblings, 200
absolute phagocyte count (APC), 334 acute promyelocytic leukemia (APL), 148, 180,
acetaminophen, 6, 32, 73, 76, 141, 448, 454, 475 199
acquired von Willebrand disease, 101, 166, 240 acyclovir, 315, 409–410
acute anemia adjuvant medications, 33, 356
PRBCs, 181 adrenocortical carcinoma (ACC)
splenic sequestration, 40–41 clinical presentation, 281
acute chest syndrome (ACS), 35–38, 355 diagnosis and staging, 281
acute leukemia (AL) incidence, 281
etiology of, 191 treatment, 282
molecular characteristics, 192 afibrinogenemia, 88, 103
prevalence, 191 allergic transfusion reactions, 65, 73
acute lymphoblastic leukemia (ALL), 158, allopurinol, 180, 183, 188, 410–411, 460
333 all‐trans retinoic acid (ATRA), 201, 481
clinical manifestations, 192–194 alpha2‐antiplasmin deficiency, 104
complications of therapy, 198 alpha‐thalassemias, 27
diagnostic evaluation, 194–195 alpha‐globin production, 51
echocardiogram and electrocardiogram, 195 diagnosis, 52
novel agents, 199 diagnosis of, 51–52
relapse, 198–199 genetic counseling and testing, 53
risk group classification, 196 Hemoglobin H, 52
T‐cell acute lymphoblastic leukemia (T‐ALL), mean corpuscular volume (MCV), 52
191 silent carrier state, 52
treatment, 196–198 alveolar soft part sarcoma, 253
acute myelogenous leukemia (AML), 308, 333 alveolar variant (ARMS), 256
acquired conditions, 200 Amicar, 100, 142
clinical presentation, 200–201 amifostine, 411
complications of therapy, 204–205 aminocaproic acid, 411–412
diagnostic evaluation, 201 amphotericin B, 412–413
etiology of, 199 anaerobic therapy, 338
post‐test, 207–209 analgesics, 33–35
relapse, 205 anaplastic lymphoma kinase (ALK), 235

Handbook of Pediatric Hematology and Oncology – Children’s Hospital & Research Center Oakland,
Third Edition. Caroline A. Hastings, Joseph C. Torkildson, and Anurag K. Agrawal.
© 2021 John Wiley & Sons Ltd. Published 2021 by John Wiley & Sons Ltd.
490 Index

anemia, 181 hemoglobin E, 54


Coombs, 10 infants, 54
definition, 1 splice site mutations, 53
diagnosis of, 1 bevacizumab, 419
Diamond–Blackfan anemia, 10 biopsy, 273
erythropoietin, 8 bleeding child
hemolytic anemia (see also hemolytic anemia) evaluation of, 85–86
investigation of, 1–2 history, 86
medical history of, 4 initial laboratory evaluation, 86–89
microcytic anemia, 6 management of epistaxis, 90
microcytic causes of, 10 physical examination, 86
oral iron challenge, 2–3 bleomycin, 379
parenteral iron therapy, 3, 5–8 bleomycin, etoposide, and cisplatin (BEP), 269
physical examination, 5, 9 bleomycin sulfate, 419–420
TEC, 11 blinatumomab, 420
transfusion therapy, 8 mechanism of action, 379
angiosarcoma, 253, 278 side effects, monitoring, and treatment, 380
Ann Arbor staging system, 229, 230 bone marrow aspiration, 214–215
anterior mediastinal masses, 166 indications, 398–399
anti‐thymocyte globulin (ATG), 413–414 pretreatment evaluation, 399
aplastic crisis, 41 bone marrow transplantation, 123
aprepitant, 414–416 bone scan, 247
arsenic trioxide (ATO), 416–417 brentuximab vedotin, 380, 418–419
mechanism of action, 377 busulfan, 46, 295, 420–421
side effects, monitoring, and treatment,
377–378 carboplatin, 219, 249, 421–422
asparaginase, 378, 417–418 carmustine, 411, 422
atypical teratoid/rhabdoid Tumor (ATRT), 223 caspofungin, 342, 422
autoimmune hemolytic anemia, 18–19 catheters
autoimmune neutropenia (AIN), 122 CVCs (see central venous catheters (CVCs))
autologous stem cell transplantation (ASCT), insertion of, 324–325
232 central line‐associated blood stream infection
avascular necrosis, 41–42, 198, 472 (CLABSI), 326
azacitidine/decitabine, 418 central nervous system (CNS) tumors
mechanism of action, 378 atypical teratoid/rhabdoid Tumor (ATRT), 223
side effects, monitoring, and treatment, 378 bone marrow aspiration, 214–215
utilization, 378 bone scan, 215
cause of, 211
bacterial prophylaxis, 314 chemotherapy, 216–217
Bard PowerLine, 325 clinical presentation, 211–213
B‐cell depletion, 143 computerized tomography (CT), 213
BEACOPP, 231 germ cell tumors, 223, 269–271
bendamustine, 232, 379 gliomas, 219–223
benzodiazepines, 34, 371 imaging studies, 213
beta‐thalassemia lumbar puncture (LP), 214
children, 54, 55 medulloblastoma, 217–219
chronic transfusion therapy, 56 microscopic criteria, 211
clinical management, 55 MRI, 213
diagnostic problems, 54 neurosurgery, 215
genotypic differentiation, 53 PET imaging, 213–214
Index 491

radiation therapy, 215–216 chimeric antigen receptor T‐cell (CAR‐T) cell


single‐photon emission computed therapy, 235
tomography (SPECT), 214 choriocarcinoma, 266
central venous catheters (CVCs) chromosome analysis, 201
assessment and management, 328–331 cisplatin, 280, 422–423
complications, 326–328 cisplatin/carboplatin, 380–381
external and multilumen devices, 323 cis‐retinoic acid/all‐trans retinoic acid, 381
external CVCs, 323 cladribine, 423–424
insertion of catheters, 324–325 cladribine/clofarabine, 382
maintenance, 325–326 clofarabine, 424
subcutaneous ports, 323 coagulopathy, 204
tunneled catheters, 323 Codman triangle, 260
type of, 324 complementary therapies, 35
cerebral vascular disease, 28 complete blood count (CBC), 137
cervical masses, 246 concomitant malignancies, 271
Chédiak Higashi syndrome (CHS), 126 congenital amegakaryocytic thrombocytopenia
chelation therapy (CAMT), 150
lead toxicity, 81–83 congenital deficiency, 103
transfusional iron overload, 79–81 cryoprecipitate, 69–70
chemotherapeutic agents, 270 cyclic neutropenia, 123
chemotherapy, 250, 280 cyclophosphamide, 23, 144, 148, 175, 187, 197,
extravasation 200, 219, 231, 235, 242, 243, 246, 251,
causes of, 404 382–383, 424–425
clinical follow‐up, 407 cyclophosphamide/ifosfamide, 382–383
DNA‐binding agents, 405 cyclosporine, 425
incidence of, 403 cytarabine, 383
non‐DNA‐binding antineoplastics, 405 cytarabine hydrochloride, 425–426
prevention of, 404
signs and symptoms of, 405 dacarbazine, 426
treatment, 405–406 dactinomycin, 426–427
immunization, 316–317 dactinomycin (Actinomycin‐D), 383
intra‐ommaya reservoir tap dapsone, 427
indications, 397 darbepoetin alfa, 427–428
materials, 397 dasatinib, 428
postprocedure monitoring and daunorubicin and doxorubicin, 383–384
complications, 398 daunorubicin hydrochloride, 428–429
pretreatment evaluation, 397 deferasirox, 430–431
procedure, 397–398 deferiprone, 431–432
LP/IT chemotherapy deferoxamine mesylate, 429–430
indications, 393 defibrotide, 431
materials, 393–394 delta beta thalassemia, 56
patient’s medical record, 393 dermatofibrosarcoma protuberans, 253
postprocedure monitoring and desmoid tumors, 253
complications, 396–397 desmopressin acetate, 432–433
pretreatment evaluation, 393 dexamethasone, 318, 433
procedure, 395–396 dexrazoxane, 433–434
prevention of, 317–318 diabetes insipidus (DI), 270
chemotherapy‐induced nausea and vomiting Diamond–Blackfan anemia (DBA), 10, 126, 130,
(CINV), 317–318 200
chest radiography, 240 dimercaprol, 434–435
492 Index

dimethyl sulfoxide, 435 ferrous gluconate, 443


dinutuximab, 384–385, 435–436 ferrous sulfate, 443–444
diphenhydramine, 436–437 fever, 206
disseminated intravascular coagulation (DIC), antibiotic treatment, 338–341
20, 112, 138, 148 definition, 334
Doppler abdominal ultrasound, 240 duration of therapy, 341
doxorubicin, 280 empiric antifungal therapy, 341–343
doxorubicin hydrochloride, 437 initial management, 337–338
dronabinol, 437–438 and neutropenia, 334–337
dyskeratosis congenita (DKC), 126, 130, 200 new sites of infection, 344–345
dyspnea non‐neutropenic oncology patient, 345
cause of, 367 P. jiroveci pneumonia, 344
pathological conditions, 367 rectal temperatures, 334
respiratory failure, 369 supportive measures, 345
treatment for, 369 viral infection, 343–344
fibromyxoid sarcoma, 253
echocardiogram, 36, 174, 187, 240, 263, 322 fibrosarcoma, 253
edetate calcium disodium, 438 filgrastim, 444
ektacytometry, 20 fluconazole, 445
electrocardiograms (EKGs), 204 fludarabine, 445–446
eltrombopag, 438–439 Fluorine‐18‐fluorodeoxyglucose positron
Embden‐Meyerhof pathway (EMP), 17 emission tomography (FDG‐PET), 255
end‐of‐life care folic acid, 446–447
parental dissatisfaction, 363 fondaparinux, 446
symptoms foscarnet, 447
constipation, 365 Free erythrocyte protoporphyrin (FEP), 54
nausea and vomiting, 364–365 fresh frozen plasma, 69
pain, 363 fungal prophylaxis, 314–315
engraftment syndrome, 298
enoxaparin, 439–440 ganciclovir, 447–448
epistaxis, 106 gastrointestinal GI bleeding, 96
epoetin alfa, 440–441 gastrointestinal (GI) tract, 364
Epstein‐Barr virus (EBV) virus, 227 gemcitabine, 448
etoposide, 385, 441 gemtuzumab, 385–386
ETV6–RUNX1 fusion gene, 192 gemtuzumab ozogamicin, 448
Ewing sarcoma family of tumors (ESFT), gene expression profiling, 192, 202
261–262 gene therapy, 46–47, 109
germ cell tumors (GCTs), 223
Face, Legs, Activity, Cry, Consolability (FLACC) central nervous system, 269–271
Behavior Scale, 349 clinical presentation, 267–268
factor V (FV) deficiency, 103 concomitant malignancies, 271
factor VIII (FVIII) deficiency, 103 diagnostic evaluation and risk stratification,
factor XI (FXI) deficiency, 104 268
factor XIII (FXIII) deficiency, 104 epidemiology, 265–266
Fanconi anemia, 123 paraneoplastic syndromes, 271
fatigue, 366–367 pathology and serum tumor markers,
febrile neutropenia (FN), 333 266–267
ferric carboxymaltose, 442 treatment and prognosis, 268–269
ferric gluconate, 442–443 germinomas, 266
ferrous fumarate, 444 Gilbert syndrome, 24
Index 493

gliomas, 219–223 hemophagocytic lymphohistiocytosis (HLH),


glucarpidase, 448–449 287
glucose‐6‐phosphate dehydrogenase (G6PD) clinical findings, 290
deficiency, 15, 17, 18 cyclosporine, 290
glutamine, 45–46 diagnostic criteria, 290
gonadal tumors, 269 feature in, 289
graft‐versus‐host disease, 296, 300, 302–304 macrophages and T‐lymphocytes, 290
granisetron, 449–450 NK cells, 290
granulocyte colony‐stimulating factor (G‐CSF), hemophilia
122, 318, 345 afibrinogenemia, 103
granulocyte immunofluorescence test (GIFT), alpha2‐antiplasmin deficiency, 104
122 clinical presentation, 104–106
granulocyte transfusion, 70–71 definition, 103
gross total resection (GTR), 268 dental care, 108
diagnosis, 106
hematopoietic growth factors, 318 factor VIII (FVIII) deficiency, 103
hematopoietic stem cell transplantation (HSCT), factor XI (FXI) deficiency, 104
46, 333 gene therapy, 109
complications of, 298 hemophilia A, 103
diet, 305 hemophilia B, 103
donor matching, 296–298 heterozygotes, 104
engraftment and graft failure, 298 hypoprothrombinemia, 103
graft‐versus‐host disease, 296, 300, 302–304 incidence, 103
idiopathic pneumonia syndrome (IPS), 304 inhibitors, 108
immune reconstitution, 306–307 novel agents, 108–109
infection prophylaxis, 304, 306 prothrombin deficiency, 103
infections, 298–300 psychological support, 108
late sequelae, 304, 305 rare coagulation factor deficiencies, 104
multidrug therapy, 295 treatment, 106–108
pretransplant preparative regimens, 298, 299 hemophilia A, 103
supportive care, 304 hemophilia B, 103
transfusion guidelines, 305–306 heparin‐induced thrombocytopenia (HIT), 147
transplantable conditions, 295–296 heparin sodium, 450–451
transplant‐associated thrombotic hepatoblastoma (HBL)
microangiopathy (TA‐TMA), 304 clinical presentation, 278–279
types of transplantation, 296 diagnosis and staging, 279
veno‐occlusive disease/sinusoidal obstructive factors, 278
syndrome, 300, 301 incidence of, 278
hemolytic anemia, 8, 162 malignant hepatic tumors, 278
autoimmune hemolytic anemia, 18–19 treatment, 279–280
evaluation, 20–21 hereditary neutropenias, 122
immature reticulocytes, 23 heterozygotes, 104
lab studies, 22 histiocytic disorders
microangiopathic hemolytic anemias, 20 classification, 287, 288
newborn period, 19 LCH (see langerhans cell histiocytosis (LCH))
red cell enzyme deficiencies, 17–18 hodgkin lymphoma
red cell membrane disorders, 15–16 childhood and adolescent/young adult (AYA),
reticulocyte index (RI), 22–23 227
treatment, 21–22 classical HL, 227, 230–231
hemolytic uremic syndrome (HUS), 147 clinical presentation, 228
494 Index

hodgkin lymphoma (cont’d ) inotuzumab ozogamicin, 386, 454–455


evaluation, 228–229 intensitymodulated radiation therapy (IMRT),
nodular lymphocytepredominant HL, 227, 231 257
non‐hodgkin lymphoma (see non‐hodgkin International Neuroblastoma Staging System,
lymphoma) 247, 248
novel agents, 231–232 intracranial hemorrhage (ICH), 106, 135
staging system, 229 invasive fungal disease, 341
treatment strategy, 229–230 irinotecan, 386–387, 455–456
tumor cells, 227 iron complex, polysaccharide, 456
host factors, 202 iron dextran complex, 456–457
human leukocyte antigen (HLA) system, 296 iron overload, 79–81
hyaluronidase, 451 iron sucrose, 457
hydrocortisone, 451 isotretinoin, 458
hydromorphone hydrochloride, 451–452 IV hydration, 32–33
hydroxyurea, 452
hydroxyurea therapy, 45 Kasabach–Merritt syndrome, 148, 249
hyperbilirubinemia/gallstones, 42–43 ketamine, 352
hypercalcemia ketorolac, 32, 458
evaluation, 184–185
treatment, 185 lactate dehydrogenase (LDH), 22
hyperleukocytosis langerhans cell histiocytosis (LCH)
clinical findings and evaluation, 180 brain MRI imaging, 289
complication, 179 diagnosis, 287
definition, 179 large/symptomatic solitary bone lesions, 288
treatment, 180–181 neoplastic process, 287
hypoprothrombinemia, 103 pathogenesis of, 287
skin lesions, 288
ibuprofen, 32 solitary bone lesions, 288
idarubicin, 452–453 systemic disease, 288–289
idiopathic pneumonia syndrome (IPS), 304 vertebral lesions, 288
ifosfamide, 453 lead toxicity, 81–83
imatinib, 386 leiomyosarcoma, 253
imatinib mesylate, 453–454 leucovorin calcium, 458–459
immune globulin, 454 lidocaine, 441–442
immune hemolytic anemias, 22 liposarcoma, 253
immune reconstitution, 306–307 lomustine, 459
immune thrombocytopenic purpura, 122 low‐molecular‐weight heparin (LMWH), 330
diagnosis of, 133 lumbar puncture (LP), 214
emergency therapy, 142 lymph nodes, 205
evaluation, 135–139 lymphocyte‐predominant (LP) cells, 227
mucosal bleeding, 133
treatment, 139–142 malignant peripheral nerve sheath tumors,
immunization, 28, 68, 71, 108, 144, 169 253
chemotherapy, 316–317 massive hepatomegaly, 177–178, 250
immunological disease, 162 mean platelet volume (MPV), 87
immunophenotyping, 205, 206 Measles‐Mumps‐Rubella (MMR), 135
increased intracranial pressure (ICP), 364 mechlorethamine, 459–460
Individualized Care Planning and Coordination medulloblastoma, 217–219
Model, 361–363 melphalan, 460
infection prophylaxis, 304, 306 meperidine, 352
Index 495

mercaptopurine, 387, 460–461 nextgeneration sequencing technology, 192


6‐mercaptopurine (6MP), 192 nilotinib, 468–469
mesna, 461 nivolumab and pembrolizumab, 389
metabolic depletion, 15 N‐methyl‐d‐aspartic acid (NMDA) receptor
methadone hydrochloride, 461–462 antagonist, 33
methotrexate, 387–388, 462–463 nongerminomatous germ cell tumors (NGGCT),
methylene blue, 463 269
methylprednisolone, 463–464 non‐Hodgkin lymphoma, 199
metoclopramide, 464 non‐Hodgkin lymphoma, 199
micafungin, 464–465 clinical presentation, 233
Microangiopathic hemolytic anemias, 20 congenital conditions, 232
microarray technology, 202 diagnostic evaluation, 233–234
microcytic anemia, 6 DLBCL and ALCL, 232–233
middle mediastinal masses, 166 histologic classification, 232
mitoxantrone, 465 incidence of, 232
morphine, 371 novel agent, 235
morphine sulfate, 465–466 staging, 234
mycophenolate mofetil (MMF), 466–467 treatment, 234–235
noninvasive positive pressure ventilation
naloxone, 467–468 (NIPPV), 369
nelarabine, 388–389, 468 nonopioid analgesics, 350–351
neonatal alloimmune neutropenia, 122 nonrhabdomyomatous soft tissue sarcomas
Neonatal Infant Pain Scale (NIPS), 348 bone sarcomas, 258
neonatal isoimmune neutropenia, 122 clinical presentation, 259
neuroblastoma diagnostic evaluation, 259–260
clinical presentation, 245–246 genetics, 258–259
diagnostic studies, 247 pathologic diagnosis, 260
genetic risk factors, 245 prognosis for, 262–263
history, 246 treatment, 260–262
laboratory studies, 246–247 nonrhabdomyosarcoma soft tissue sarcomas
physical examination, 246 (NRSTS), 253
staging, 247, 248
treatment, 247, 248 oncologic emergencies
neuron‐specific enolase (NSE), 247 anemia, 181
neutropenia hypercalcemia, 184–185
febrile neutropenia, 335–337 hyperleukocytosis, 179–181
and fever intracranial pressure and brain herniation,
bacteremia and sepsis, 334–335 176–177
septic shock, 334 massive hepatomegaly, 177–178
neutropenic colitis neutropenic colitis, 178–179
clinical findings and evaluation, 178–179 post‐test, 188–190
treatment, 179 spinal cord compression, 175–176
neutrophils superior vena cava and superior mediastinal
child management, 127–129 syndrome, 172–175
component, 119 tumor lysis syndrome, 181–184
etiology of, 120–122 ondansetron, 469
inherited causes of, 122–126 opioids, 364
initial evaluation, 126–127 opsoclonusmyoclonus‐ataxia syndrome
post‐test, 130–132 (OMAS), 246
risk assessment, 119–120 oseltamivir, 469–470
496 Index

osteosarcoma, 260–261 palonosetron, 470


oxaliplatin, 470 paraneoplastic syndromes, 271
oxycodone, 352 parenteral iron therapy, 3, 5–8
paroxysmal nocturnal hemoglobinuria
packed red blood cell transfusion (PRBC), 23 (PNH), 16
acute blood loss and nonimmune hemolytic patient‐controlled analgesia (PCA), 352
anemia, 66 pazopanib, 470–471
autoimmune hemolytic anemia, 66 pediatric antiemetics, 318, 319
blood transfusion guidelines, 64 pediatric cancers
chronically transfused patients, 66–67 abdominal mass, 168
cytomegalovirus (CMV)‐negative blood, 65 adenopathy, 160, 161
dosing of, 67 back pain, 163
exchange transfusion/erythrocytapheresis, 67 bicytopenia and pancytopenia, 164
neonates, 65–66 bleeding, 165
oncology patients, 66 bone marrow examination, 165
platelets and white blood cells, 65 bone or joint pain, 163
severe chronic anemia, 66 bone pain, 164
pain management, 32 cytopenias/abnormalities, 164
analgesics, 347, 348 diagnosis of, 157
assessment of, 348 diffuse/multifocal bone pain, 163
dosing of pain medication, 347 excisional biopsy, 162
neonates and infants, 348 fever, 158, 159
nonpharmacologic approaches, 354 headache, 159
oral form of medication, 347 incidence, 157
pain pharmacology, 350 laboratory and imaging findings, 158, 162
post‐test, 356–357 lymphadenopathy, 159
preverbal/nonverbal children, 348–349 mediastinal masses, 166
school‐aged and older children, 349–350 musculoskeletal pain, 163
step 1 therapy, 350–351 neuroblastoma and Wilms tumor, 158, 166
step 2 therapy, 351, 352 palpable abdominal mass, 166
step 3 therapy, 351–353 post‐test, 170–172
step 4 therapy, 352, 354 splenomegaly, 162–163
palliative care surgical staging, 167
anorexia and weight loss, 366 suspected leukemia, 165
assessment symptoms and signs, 157, 158
history, 365 pelvic masses, 246
medications, 366 pentamidine isethionate, 471
physical examination, 365–366 perioperative management, 43
definition, 361 perioperative transfusion, 44–45
dyspnea (see dyspnea) peripheral blood stem cell transplant (PBSCT),
end‐of‐life care 296
parental dissatisfaction, 363 platelet surface glycoprotein, 85
symptoms, 363–365 platelet transfusions, 306
fatigue, 366–367 cryoprecipitate, 69–70
gas exchange, 371 dosing of, 68–69
individualized care planning and fresh frozen plasma, 69
coordination, 361–363 indications, 68
nonpharmacologic interventions, 371 posaconazole, 471–472
nutritional supplementation, 366 positron emission tomography (PET), 229
supporting respiration, 369–371 posterior mediastinal masses, 166
Index 497

postoperative care, 43–44 sickle cell disease (SCD)


prednisone, 472 abnormal hemoglobins, 27
preoperative care, 43 acute anemia, 40–41
pretransplant preparative regimens, 298, 299 acute chest syndrome/pneumonia, 35–38
priapism, 38 alpha‐thalassemia, 27
prilocaine, 441–442 analgesics, 34–35
primary mediastinal B‐cell lymphoma (PMBCL), aplastic crisis, 41
232 avascular necrosis, 41–42
primordial germ cells (PGCs), 265 complementary therapies, 35
procarbazine hydrochloride, 472–473 fever and infection in, 29–31
prophylactic antibiotics, 128 general management, 34
prophylaxis hyperbilirubinemia/gallstones, 42–43
bacterial prophylaxis, 314 inpatient, 33–34
fungal prophylaxis, 314–315 mild pain, 32
invasive procedures, 315 novel agents, 45–46
nutrition and dental hygiene, 313 pain management, 32
prevention of, 313 perioperative management, 43
rectal suppositories and rectal temperatures, perioperative transfusion, 44–45
314 postoperative care, 43–44
trimethoprim/sulfamethoxazole (TMP/SMX) preoperative care, 43
prophylaxis, 314 preventive care, 28–29
prothrombin deficiency, 103 priapism, 38
protracted fever, 340 retinopathy/hyphema, 42
pyruvate kinase (PK) deficiency, 17 severe pain, 32–33
stroke, 38–40
radiation therapy, 215–216, 261, 280 transfusion therapy, 44
radiotherapy, 235 vaso‐occlusive episodes, 27, 31–32
rasburicase, 473 single‐photon emission computed tomography
recombinant factor VIIa, 85 (SPECT), 214
red blood cells (RBCs), 15 sirolimus, 476–477
red cell membrane disorders, 15–16 skeletal metastases, 229
retinoblastoma skin irritation, 327
clinical presentation, 276 skin lesions, 246
diagnosis and staging, 276 sodium thiosulfate, 477
follow‐up, 277–278 soft tissue hemorrhages, 105
secondary mutations, 275 soft tissue sarcomas
treatment, 276–277 clinical presentation, 254–255
retinopathy/hyphema, 42 diagnostic evaluation, 255
rhabdomyosarcoma, 253 genetics, 254
RhoGAM, 141 late effects in, 263
Rho(D) immune globulin, 473–474 pathologic diagnosis, 255–256
Rituximab, 199 prognosis, 257–258
rituximab, 143, 389, 474–475 rhabdomyosarcoma, 253
rivaroxaban, 475 staging and classification, 256
romiplostim, 475–476 treatment, 256–257
spinal cord compression
sacrococcygeal tumors, 267, 269 cause of, 175
sargramostim, 476 clinical findings and evaluation, 176
severe congenital neutropenia (SCN), 123 management, 176
severe pain, 32–33 splenectomy, 16, 17, 144
498 Index

splenomegaly, 122 novel anticoagulants, 116


staging, 241 prevention and treatment, 111
steroids, 389–390 testing for, 112–113
storage disease, 162 thrombolysis, 114
stroke, 38–40 thyroid tumors
Stroke Prevention Trial in Sickle Cell Anemia clinical presentation, 283
(STOP), 39 diagnosis and staging, 283–284
succimer, 477–478 genetic predisposition, 283
superior vena cava (SVC), 173 incidence of, 282
superior vena cava syndrome (SVCS), 173, 228 treatment, 284
surgical procedures, 229 tisagenlecleucel, 390–391
synovial sarcoma, 253 tissue plasminogen activator (TPA), 329, 338
systemic antibiotic therapy, 328 topical anesthesia, 323
topotecan, 391, 480–481
tacrolimus, 478 total parenteral nutrition (TPN), 305, 366
temozolomide, 390, 478–479 tranexamic acid, 481
teratomas, 266 transdermal fentanyl, 352
thalassemias transfusion‐associated circulatory overload, 73
alpha‐globin chains, 51 transfusion‐associated graft versus‐host disease,
alpha‐thalassemias, 51–53 73–74
beta‐globin chains, 51 transfusion medicine
beta‐thalassemia, 53–56 clerical errors and misidentification, 63
delta beta thalassemia, 56 complications, 63
globin chain production/deletion, 51 cryoprecipitate, 69–70
HPFH, 56 granulocyte transfusion, 70–71
newborn screening, 56–58 platelet transfusions, 67–69
thioguanine, 390, 479 PRBC (see packed red blood cell transfusion
thiotepa, 479 (PRBC))
thoracic masses, 246 transfusion reactions, 71–74
thrombin, 480 transfusion reactions
thrombin time (TT), 103 allergic transfusion reactions, 73
thrombocytopenia, 123 bacterial infections, 74
acute immune thrombocytopenic purpura, hemolytic transfusion reactions, 71–73
133–135 transfusion‐associated circulatory overload, 73
chronic immune thrombocytopenic purpura, transfusion‐associated graftversus‐host
142–144 disease, 73–74
decreased platelet production, 149–151 transfusion‐related acute lung injury, 73
differential diagnosis of, 134–135 transfusion‐related acute lung injury, 73
drug‐induced thrombocytopenia, 147 transfusion therapy, 44
neonatal alloimmune thrombocytopenia, transient myeloproliferative disorder (TMD), 199
144–146 transplant‐associated microangiopathy, 20
neonatal autoimmune thrombocytopenia, transplant‐associated thrombotic
146–147 microangiopathy (TA‐TMA), 304
nonimmune thrombocytopenia, 147–149 tretinoin, 481–482
post‐testt, 151–154 trimethoprim/sulfamethoxazole, 314, 482–483
thrombophilia tumor dissemination, 267
anticoagulation, 114–115 tumor lysis syndrome, 192
duration of therapy, 115–116 clinical findings and evaluation, 182
evaluation of, 111–112 definition, 181–182
management of, 113–114 prevention and intervention, 182–184
Index 499

undifferentiated sarcoma, 253 Von Willebrand disease (VWD)


uridine diphosphate glucuronosyltransferase acquired von willebrand syndrome, 101
(UGT1A1), 24 autosomal dominant manner, 95
urine catecholamines, 250 clinical presentation, 96
diagnosis, 96–98
valacyclovir, 483 treatment, 98–101
valganciclovir, 483–484 types, 95
vancomycin, 339–340 von Willebrand factor (VWF), 85
vancomycin‐resistant enterococcus (VRE), 340 voriconazole, 486–487
vaso‐occlusive episodes, 27, 31–32
vaso‐occlusive events (VOE), 355 warfarin, 147
venlafaxine, 357 warfarin sodium, 487–488
veno‐occlusive disease (VOD), 298 white blood cell (WBC), 119
venous thromboembolism, 328 Wilms tumor
vinblastine sulfate, 484–485 clinical presentation, 240
vinca alkaloids, 144 evaluation of, 240
vincristine and vinblastine, 391–392 incidence and prevalence, 239
vincristine sulfate, 485 pathologic diagnosis, 240–241
vinorelbine, 485–486 staging, 241
viral prophylaxis and treatment, 315–316 treatment, 241–242
visceral pain, 357 Wiskott–Aldrich syndrome (WAS), 126, 150
vitamin K1/phytonadione, 486 Wong‐Baker FACES Pain Rating Scale, 350

You might also like