Vous êtes sur la page 1sur 10

C H A P T E R 64

CLINICAL MANIFESTATIONS AND TREATMENT OF ACUTE


LYMPHOBLASTIC LEUKEMIA IN CHILDREN
Sima Jeha and Ching-Hon Pui

Serial risk-directed clinical trials have optimized the combination of MLL gene at 11q23 with a variety of partner genes but principally
chemotherapeutic agents and, along with advances in supportive care, AF4. Identical twin infant pairs with concordant ALL share the same
have led to current cure rates of childhood acute lymphoblastic leuke- acquired MLL gene rearrangements.
mia (ALL) exceeding 85% compared with long-term survival less than The most common chromosome translocation in childhood ALL,
10% in the 1960s.1 However, because ALL is the most common t(12;21), results in ETV6(TEL)-RUNX1(AML1) fusion. In contrast
cancer in children, relapsed ALL remains the leading cause of death to MLL-rearranged ALL, ETV6-RUNX1 leukemias present after
from a disease in this age group. Over the past decade, minimal infancy and have a concordance rate of only 10% in identical twins.
residual disease (MRD) has become a major determinant in risk strati- ETV6-RUNX1 fusion can be found in as many as 1% of cord blood
fication, cranial irradiation has been successfully omitted from some samples of normal newborn babies, a frequency 100 times higher
frontline protocols, and the tyrosine kinase inhibitor (TKI) imatinib than the prevalence of this subtype of leukemia, suggesting that
has revolutionized the treatment of patients with BCR-ABL1 rear- additional postnatal mutations are necessary for malignant transfor-
rangement. Our understanding of the immunology and molecular mation. Analysis of Guthrie cards of 2- to 6-year old children with
pathways involved in ALL is improving at a rapid pace, and several ALL showed that most did have detectable, clonotypic ETV6-RUNX1
targeted therapies are showing promise in early clinical trials. The sequences at birth. In addition, identical ETV6 and RUNX1 break-
challenge is to successfully incorporate targeted therapy into personal- points were present in each of twin pairs with a very asynchronous
ized ALL regimens aiming at improving cure rates and reducing diagnosis, supporting the requirement for one or more additional
toxicity.2 postnatal events after in utero initiation. This interpretation suggests
that ETV6-RUNX1 initiates leukemogenesis but is insufficient for
overt disease, and further genetic alterations are required. ETV6 dele-
EPIDEMIOLOGY tions on chromosome 12p, the most frequent additional genetic
abnormalities described in cases of ALL with ETV6-RUNX1, appear
Acute lymphoblastic leukemia accounts for approximately 75% of all to be subclonal in fluorescence in situ hybridization (FISH) analysis
cases of childhood leukemia and is the most common pediatric cancer of leukemic cells from nontwin patients. These findings indicate that
in developed countries, representing 23% of cancer diagnoses among ETV6 deletion is a secondary or later event in leukemogenesis and
children younger than 15 years of age. About 2400 children and suggest that leukemia might be initiated in utero but requires an
adolescents are diagnosed with ALL each year in the United States, essential second postnatal event (two hits).6
an annual rate of 30 to 40 per million. The peak incidence of ALL Several genetic, dietary, and environmental factors have been pro-
occurs around 4 years of age. This young age peak historically has posed to modify the risk of leukemia initiation. Children with various
appeared at different times in different countries and has been associ- congenital immunodeficiency diseases, including Wiskott-Aldrich
ated with major periods of industrialization, suggesting a causative syndrome, congenital hypogammaglobulinemia, and ataxia telangiec-
role for environmental carcinogens. ALL occurs more frequently in tasia, have an increased risk of developing lymphoid malignancies, as
boys than in girls at all ages.3 do patients under chronic treatment with immunosuppressive drugs.
Acquired and inherited genetic factors play an essential role in ALL The loss of cellular immune surveillance capability for tumor antigens
as supported by the demonstration of genetic abnormalities in leuke- and the inability to self-regulate lymphoproliferative processes may
mic cells of children with ALL as well as by clinical observations. contribute to malignant transformation in these patients. Absence of
Children with the constitutional chromosomal abnormality trisomy exposure to common infections in the first year of life is associated
21 (Down syndrome) are up to 15 times more likely to develop leu- with a higher risk of developing ETV6-RUNX1positive or hyperdip-
kemia than children without Down syndrome. Genetic instability and loid ALL in older children. A possible explanation is that in the
DNA repair disorders (e.g., Bloom syndrome, ataxia telangiectasia, absence of infection-driven modulation of the naive immune network
Fanconi anemia) are also associated with an increased risk of develop- in infants, subsequent infectious exposures could result in a highly
ing ALL.4 Among identical twins, if one is diagnosed with ALL during dysregulated response to infections in older children, contributing to
the first year of life, the risk that the other twin will develop ALL is leukemogenesis. Exposure to ionizing radiation and certain toxic
more than 70%, approaching 100% in twins with a single monocho- chemicals could also facilitate the development of acute leukemia. The
rionic placenta. Highest in infancy, this risk diminishes with increas- high incidence of leukemia among survivors of atomic bomb explo-
ing age at diagnosis in the first twin to about one in 10 in older sions in Japan during World War II is well documented. Among sur-
children. If the first twin develops ALL by 5 to 7 years of age, the risk vivors of the atomic bomb, there was no increase in the incidence of
to the second twin is at least twice that in the general population, leukemia in children exposed to radiation in utero. This experience
regardless of zygosity.5 After the age of 7 years, the risk to the unaf- contrasts with other reports of an increased incidence of ALL in chil-
fected twin is similar to that for persons in the general population. The dren exposed to medical diagnostic radiation both in utero and in
extraordinarily high concordance rate in monozygotic infants twins childhood. Other evidence linking most environmental exposures to
compared with dizygotic infant twins or nontwinned siblings results risk of childhood ALL has largely been inconsistent. Causation path-
from the metastasis of leukemic cells from one twin to the other ways are likely to be multifactorial, and it is probable that the risk of
through shared placental circulation. The majority of infants with ALL from environmental exposure is influenced by genetic variation
ALL have a chromosome translocation that results in the fusion of the through the coinheritance of multiple low-risk variants.
951
952 Part VII Hematologic Malignancies

PATHOBIOLOGY These patients probably would require more intensive therapy because
this genotype was associated with poor outcome in the reported
Acute leukemias comprise a group of clonal disorders of maturation series, albeit not independently significant in one.13
at an early phase of hematopoietic differentiation. ALL subtypes are Experimental models have established that cooperative mutations
a heterogeneous group of malignancies with distinctive immunophe- are necessary to induce leukemia and contribute to the development
notype and molecular pathogenesis that result in varying clinical of drug resistance. Association studies of ALL based on the candidate
characteristics and response to therapy. Accurate pathobiologic diag- gene approach have evaluated a restricted number of polymorphisms
nosis is not only important for prognostic stratification but can also in genes implicated in the metabolism of carcinogens, folate metabo-
help define patient-specific therapeutic approaches.7 lism, protection of DNA from carcinogen-induced damage, and cell
Lymphoblastic leukemias can arise from either B- or T-cell mutant cycle regulation. Such studies highlight difficulties in conducting
hematopoietic cells capable of indefinite self-renewal. T-cell ALL statistically and methodologically rigorous investigations into ALL
can be classified into several distinct genetic subgroups that corre- risk. Genome-wide association studies of childhood ALL have
spond to specific T-cell development stages and is frequently recently demonstrated that common variation at four genetic loci
associated with translocations of T-cell receptor genes on chromo- [7p12.2 (IKZF1), 9p12 (CDKN2A/CDKN2B), 10q21.2 (ARID5B)
some 14q11 or 7q34 with other gene partners. Recent studies have and 14q11.2 (CEBPE)] confers a modest increase in risk, establishing
identified a novel high-risk immature T-cell subtype termed early a role for genetic susceptibility in the development of ALL. In addi-
T-cell precursor (ETP) ALL with immunologic markers and gene tion to identifying common, low-penetrance susceptibility alleles,
expression reminiscent of double-negative 1 thymocyte that retains these data provide insights into disease causation by identifying risk
the ability to differentiate into both T-cell and myeloid, but not variants and annotating genes involved in transcriptional regulation
B-cell, lineages.8 The discovery of its mutational spectrum recapitu- and differentiation of B-cell progenitors.
lated that of acute myeloid leukemia (AML) by whole-genome
sequencing and a global transcription profile similar to that of normal
hematopoietic stem cells (HSCs) and granulocyte macrophage CLINICAL MANIFESTATIONS
precursors suggests that this subtype of leukemia is a stem cell leuke-
mia. The prevalence of mutations in genes regulating cytokine recep- Children with ALL present with nonspecific symptoms and signs
tor and Ras signaling, and histone modification further suggests reflecting the degree of disruption in bone marrow (BM) function
that the addition of myeloid-directed therapy might improve and the extent of extramedullary infiltration. The most common
the outcome of this subtype.9 Most B-lineage leukemias are early presenting symptoms are fever, fatigue, pallor, petechiae, bruising,
precursor B cell, expressing CD19 and CD10 (or cALLa, the common bleeding from mucosal surfaces, and pain (Table 64-1). Patients,
acute leukemia antigen) but lacking surface or cytoplasmic immuno- especially young children, may present with bone pain, arthralgia,
globulin. The mature B-cell ALLs, or Burkitt-cell leukemia, are strati- or refusal to walk because of leukemic infiltration of the bone or
fied separately and are not included in this chapter (see Chapters 81 joint or because of expansion of the BM cavity by leukemic cells.
and 83). The evolution of symptoms may proceed over a few days, weeks,
Recent advances in global genome analysis have enabled the or months. Less common presenting symptoms include headache,
identification of recurring alterations in genes and pathways with key visual complaints, vomiting, respiratory distress, oliguria, and anuria.
roles in cell growth and tumorigenesis. Several observations suggest Occasionally, patients present with life-threatening infection or
that multiple lesions are acquired subsequent to founding transloca- bleeding.
tions to induce leukemogenesis. Single nucleotide polymorphism On physical examination, fever, pallor, petechiae, and ecchymoses
(SNP) array analysis demonstrated substantial differences in the fre- may be present. The lymphoproliferative nature of the disease may
quency of copy number abnormality (CNA) among various ALL
subtypes. MLL-rearranged cases had less than one CNA per case,
suggesting that MLL is a potent oncogene that requires very few Table 64-1 Clinical Presentation of Acute Lymphoblastic Leukemia
cooperating lesions to induce leukemia transformation, but ETV6-
RUNX1 and BCR-ABL1 leukemias have more than six lesions Symptoms and Signs Etiology Management
per case.
Fever Disease or infection Always conduct fever
Deletion or sequence mutation of the IKZF1, a gene that encodes
workup and provide
the early lymphoid transcription factor IKAROS, is present in more
broad antimicrobial
than 80% of BCR-ABL1positive ALL and in a novel high-risk sub-
coverage until infectious
group of BCR-ABL1negative ALL that has a gene expression profile
etiology is ruled out
significantly similar to that of BCR-ABL1-positive ALL.10 The BCR-
ABL1-like ALL occurs in as many as 7% to 9% of children with Fatigue, pallor Anemia (ALL RBC transfusion (slow if
ALL. Approximately half of the BCR-ABL1like cases have rearrange- infiltrating BM) anemia is severe; avoid
ments of the lymphoid cytokine receptor gene CRLF2, with con- in hyperleukocytosis)
comitant Janus kinases (JAKs) mutations in one-third of the Petechiae, bruising, Thrombocytopenia Transfuse with platelets
CRLF2-rearranged cases.11 This genotype is associated with high risk bleeding (ALL infiltrating
of relapse, independent of age, leukocyte count at diagnosis, cytoge- BM)
netics, and levels of MRD after remission induction. A recent tran-
scriptome sequencing study of 12 cases of BCR-ABL1like cases (with Pain Leukemia infiltrating Establish diagnosis and
whole-genome sequencing in two of them) identified structural bones or joints or start chemotherapy
alterations and mutations activating kinase and cytokine receptor expanding BM
signaling in all cases, including EBF1-PDGFRB, NUP214-ABL1, cavity
RANBP2-ABL1, BCR-JAK2, STRN3-JAK2, and activating mutations Respiratory distress, Mediastinal mass Avoid sedation in the
of IL7R or FLT3. Importantly, preclinical studies showed that primary superior vena presence of tracheal
leukemic cells harboring PDGFRB or ABL1 fusion responded to cava syndrome compression; establish
ABL1 TKIs, and those harboring BCR-JAK2 or mutated IL7R diagnosis as soon as
responded to JAK2 inhibitor, suggesting that these patients would possible and start
benefit from the targeted therapy.12 Another novel subtype of B-cell chemotherapy
precursor ALL, characterized by CRLF2 alterations (PAR1 deletion
and IGH-CRLF2), occurs in 5% to 7% of children with ALL and, ALL, Acute lymphoblastic leukemia; BM, bone marrow; RBC, red blood cell.
remarkably, in approximately 50% of the cases with Down syndrome.
Chapter 64 Clinical Manifestations and Treatment of Acute Lymphoblastic Leukemia in Children 953

be manifested as lymphadenopathy, splenomegaly, or less commonly present at diagnosis. The presenting leukocyte counts range widely
hepatomegaly. Central nervous system (CNS) involvement is uncom- from 0.1 to 1500 109/L. Leukemic blasts may or may not be seen
mon at presentation and in most instances is detected by screening on peripheral smear. Approximately 45% of children have leukocyte
lumbar puncture in high-risk patients who are asymptomatic at the counts less than 10 109/L, and 15% present with hyperleukocytosis
time of the puncture (Fig. 64-1, A and B). Papilledema, retinal hem- (>100 109/L). Patients with hyperleukocytosis are at increased risk
orrhages, and cranial nerve palsies should be ruled out on examina- of CNS disease, tumor lysis syndrome, and leukostasis. Leukostasis
tion. CNS involvement usually is restricted to leptomeninges, and may manifest as dyspnea, chest pain, alterations in mental status,
parenchymal mass lesions are uncommon. Epidural spinal cord com- cranial nerve palsies, or priapism. The majority of childhood ALL are
pression is a rare but serious presenting finding and requires immedi- B cell in derivation with approximately 12% to 15% of children with
ate chemotherapy and high-dose glucocorticoid therapy. Laminectomy ALL having a T-cell immunophenotype. T-cell ALL usually occurs in
or radiotherapy is generally not necessary because leukemias are very patients older than 9 years of age with elevated leukocyte counts and
sensitive to chemotherapy at diagnosis. Overt testicular involvement is associated with CNS involvement. Coagulopathy, usually mild, can
occurs in only 2% of boys and usually presents as painless, asym- occur in T-cell ALL and is only rarely associated with severe bleeding.
metric enlargement that can be distinguished from hydrocele by Elevated serum uric acid and lactate dehydrogenase levels are common
ultrasonography (Fig. 64-1, C and D). Less common presenting in patients with a large leukemic cell burden. Patients with massive
features include ocular involvement, subcutaneous nodules (leukemia renal involvement can have increased levels of creatinine, urea nitro-
cutis) (see Fig. 64-1, E and F) and enlarged salivary glands (Mikulicz gen, uric acid, and phosphorus. Approximately 0.5% of patients have
syndrome). Approximately 55% of T-cell cases present with an ante- hypercalcemia at diagnosis, attributable to the release of parathyroid
rior mediastinal mass. A bulky mediastinal mass can compresses the hormone-like protein from lymphoblasts and leukemic infiltration of
great vessels and trachea, resulting in superior vena cava syndrome bone; these patients tend to be in the older age group and present with
and respiratory distress. Patients with a large mediastinal mass gener- low blast cell counts. This complication generally resolves rapidly with
ally present with cough, dyspnea, orthopnea, dysphagia, stridor, cya- hydration and chemotherapy. Liver dysfunction caused by leukemic
nosis, facial edema, increased intracranial pressure, and sometimes infiltration occurs in 10% to 20% of patients, is usually mild, and has
syncope. When significant tracheal compression is present, general no prognostic consequences. Because vincristine and daunorubicin
anesthesia should be avoided and procedures should be performed are metabolized primarily through biliary excretion, modifications of
under local anesthesia. Immediate diagnosis and initiation of steroids the dosage of these agents are recommended if the direct bilirubin level
and chemotherapy is essential to prevent respiratory failure. is elevated.
Clinical laboratory data often reveal a broad spectrum of abnormal Abnormalities of the bone, such as metaphyseal banding, perios-
findings. Various degrees of anemia and thrombocytopenia are usually teal reactions, osteolysis, osteosclerosis, or osteopenia, can be

A C E

B D F
Figure 64-1 CENTRAL NERVOUS SYSTEM (CNS), TESTICULAR, AND SUBCUTANEOUS INVOLVEMENT
IN CHILDHOOD ACUTE LYMPHOBLASTIC LEUKEMIA (ALL). CNS disease identified in the cerebrospinal
fluid (CSF) by screening lumbar puncture at the time of diagnosis in a 12-year-old boy with high-risk precursor B-cell
ALL. The total count of the CSF specimen was 6131/L with 6076 white blood cells/L and 98% blasts. A and B,
The cytospin preparation shows mostly blasts, slightly altered morphologically by the preparation. In B, there is a small
lymphocyte (middle) for comparison with the blasts. C and D, Testicular disease noted at relapse in a 13-year-old boy
with precursor B-cell ALL. Note the infiltrate of blasts in the parenchyma of the testes, surrounding the seminiferous
tubules. Immunostaining (not shown) demonstrated that the blasts were CD19+, CD10+, and TdT+. E and F, Cutane-
ous disease at diagnosis. The patient was an 8-year-old boy with a scalp lesion for 2 months that was initially treated
with antibiotics. On biopsy, there was much crush artifact, but deep in the specimen, there was an infiltrate of blasts
separating fibers (F) shown to be B-cell lineage. Interestingly, the patient had a normal complete blood count, but bone
marrow was packed with blasts that had a precursor B-cell phenotype and a hyperdiploid karyotype.
954 Part VII Hematologic Malignancies

demonstrated by radiographic studies in half of patients, especially fusion, have been abolished by recent improvements in risk-directed
those with low presenting leukocyte counts. Vertebral compression treatment. Aberrant expression of myeloid-associated antigens has
fracture can be detected in 2% of the cases, usually with low leukocyte been observed with certain genetic subtypes. CD15, CD33, and
count and hyperdiploidy (>50 chromosomes), and is not associated CD65 are expressed in ALL cases with a rearranged MLL gene, and
with adverse prognosis. CD13 and CD33 are expressed in cases with the ETV6-RUNX1
fusion. Once associated with a poor outcome in some studies,
myeloid-associated antigen expression has no prognostic impact
DIFFERENTIAL DIAGNOSIS in contemporary risk-directed treatment programs. The recently
identified ETP ALL subtype is characterized by immature genetic
Children with ALL present with a variety of nonspecific symptoms and immunophenotypic features (CD1a negative, CD8 negative, and
that may mimic other conditions. Pancytopenia and fever are also CD5 weak and the expression of stem cell or myeloid markers) and
presenting symptoms for aplastic anemia. Failure of a single cell line, a dismal prognosis with conventional therapy.9
as in transient erythroblastic anemia, idiopathic thrombocytopenic Acute lymphoblastic leukemia can be classified according to
purpura, and congenital or acquired neutropenia, sometime produces modal chromosomal number (ploidy) and specific genetic abnormali-
a clinical picture that is difficult to distinguish from ALL. Routine ties of the leukemia stem line. Hyperdiploidy (>50 chromosomes per
BM aspiration is not necessary for patients with severe thrombocy- cell) is associated with an age of 1 to 10 years, a lower median leu-
topenia and no other hematologic or physical evidence of leukemia. kocyte count, increased sensitivity to antimetabolite agents, and a
However, BM aspiration should be performed to exclude leukemia favorable prognosis. A hypodiploid karyotype (<44 chromosomes per
in patients who require glucocorticoid treatment. Children with cell), by contrast, predicts a poor outcome. Molecular analysis can
infectious mononucleosis or other acute viral illnesses may present identify prognostically and therapeutically relevant subgroups that
with fever, adenopathy, splenomegaly, lymphocytosis, or pancytope- cannot be identified by karyotyping. ETV6-RUNX1, the most
nia. Fever, arthralgias, or a limp may frequently be confused with common specific genetic rearrangement in childhood ALL, has been
juvenile rheumatoid arthritis, which can also be associated with associated with a good prognosis. With the exception of T-cell ALL
anemia, leukocytosis, and mild splenomegaly. Children with promi- with the t(11;19), the prognosis of cases with 11q23/MLL rearrange-
nent bone pain frequently have nearly normal blood counts, a finding ments is generally poor.
that can contribute to a delay in diagnosis. Immunostains and molec- Genetic features do not entirely account for treatment outcome,
ular studies help differentiate ALL from AML and other small blue and their prognostic impact also depends on the treatment efficacy.
cell malignancies that invade the BM, including neuroblastoma, Although up to 15% of patients with hyperdiploidy greater than 50
rhabdomyosarcoma, Ewing sarcoma, and retinoblastoma. Infants chromosomes per cell or ETV6RUNX1 fusion have recurrences of
may present with subcutaneous nodules (leukemia cutis) that look their leukemia, a substantial proportion of the patients with the
clinically like Langerhans cell histiocytosis. t(9;22) and BCR-ABL1 fusion who are 1 to 9 years old and have low
leukocyte counts at diagnosis may be cured with intensive chemo-
therapy alone.15 Among patients with MLLAF4 fusion, infants and
PROGNOSIS adults have a worse prognosis than children. Interindividual vari-
ability in the pharmacokinetics and pharmacodynamics of many
Contemporary regimens have abolished the prognostic impact of antileukemic agents also contributes to the heterogeneity in treatment
many clinical and biologic features, demonstrating that the single response among patients with specific genetic abnormalities.16
most important prognostic factor in childhood ALL is appropriate The degree of reduction of the leukemic cell clone early during
risk-directed therapy (Table 64-2). Accurate assessment of relapse remission induction therapy is determined by both leukemic
hazard is an integral part of ALL therapy, so that only high-risk cell genetics and host pharmacogenetics and has shown greater prog-
patients are treated aggressively, with less toxic therapy reserved for nostic strength than any other individual biologic or host-related
cases at lower risk of failure. feature.17 Measurements of MRD by flow-cytometric detection
To facilitate comparison of treatment results among different of aberrant immunophenotypes or by polymerase chain reaction
clinical trials, participants in a 1993 workshop sponsored by the (PCR) of clonal antigenreceptor gene rearrangements provides a
United States National Cancer Institute adopted a uniform risk clas- level of sensitivity and specificity that cannot be attained by tradi-
sification based on age and leukocyte count. Two-thirds of the tional morphologic assessment of early treatment response. Patients
patients who were 1 to 9 years old with precursor B-cell ALL and a who achieve an immunologic or molecular remission, defined as
leukocyte count less than 50 109/L were considered to be at stan- leukemic involvement of less than 104 nucleated BM cells on
dard risk of relapse; the other third was classified as high risk. This
classification proved to be of limited prognostic value because up to
one-third of patients designated as standard risk may relapse, and
these criteria cannot be applied to T-cell ALL. Moreover, the prog- Minimal Residual Disease
nostic impact of age and, to a lesser extent, leukocyte count is largely
attributable to their association with specific genetic abnormalities. The rapidity of response to induction therapy is an important
For example, the overall poor prognosis of infants younger than 12 independent predictor of outcome. There is strong concordance
months of age can be explained by the very high frequency of MLL between the assessment of MRD by flow cytometry and by PCR
rearrangements (70%80%) in this age group,14 and the overall favor- methods. We (the authors) monitor MRD using primarily flow
able outcome of patients ages 1 to 9 years is related to the preponder- cytometry methods, which are simple and rapid, and we reserve
ance of cases (70%) with hyperdiploidy (>50 chromosomes) or PCR methods for the few patients (<5%) whose leukemic cells
ETV6-RUNX1 (also known as TEL-AML1) fusion, which are both lack a suitable immunophenotype. About half of all patients show
favorable genetic features. Thus a more reasonable strategy is to a disease reduction to 104 or lower after only 2 weeks of remis-
develop clinical prognostic risk categories based on their major sion induction, and these patients appear to have an exception-
immunophenotypic features and genetic characteristics. ally good treatment outcome. Persistence of MRD of 104 or more
Early pre-B ALL, lacking immunoglobulin synthesis, is the most at 4 months from diagnosis is associated with an especially
common form of acute leukemia in children. The high-risk features dismal outcome. The adverse prognosis of high-risk slow early
previously ascribed to pre-B ALL (presence of cytoplasmic immuno- responders can be improved with intensification of induction and
globulin M) are closely associated with the presence of the t(1;19) consolidation. Standard-risk cases may be spared the increased
translocation and E2APBX1 fusion. Prognostic distinctions among risk of early morbidity and mortality from intensive induction,
ALL immunophenotypes, including the negative prognostic impact provided that they receive postinduction intensification therapy.
once associated with T-cell ALL and pre-B ALL with E2A-PBX1
Chapter 64 Clinical Manifestations and Treatment of Acute Lymphoblastic Leukemia in Children 955

Table 64-2 Prognostic Factors in Acute Lymphoblastic Leukemia

Factor Prognosis Clinical Application


Age
<1yr MLL+ (70%80% infants): Poor outcome MLL: Do well on standard ALL therapy
MLL : Same outcome as for older children Potential role for FLT3 inhibitor for MLL+
19yr Standard risk ALL biology may change risk
>9yr Higher risk ALL biology may change risk
White Blood Cell Count
<50 109/L Standard risk ALL biology may change risk
50 109/L Higher risk ALL biology may change risk
Central Nervous System
CNS3 Higher risk Therapy intensification
CNS2 Higher risk of CNS relapse CNS-directed therapy intensification
Traumatic lumbar puncture with blasts
Testicular Higher risk Therapy intensification
Immunophenotype
T cell Higher risk Poor outcome abolished with current therapy
PreB cell (cIgM+) Higher risk Poor outcome abolished with current therapy
Early preB cell Standard risk Genetics may change risk
ETP Dismal prognosis Ongoing studies exploring effective therapies
Ploidy
>50 (DI >1.16) Low risk Good response to antimetabolites
<44 Higher risk Therapy intensification
Genetic Alterations
t(9;22)/BCR-ABL1 Higher risk BCR-ABL inhibitors
t(4;11)/MLL-AF4 Higher risk Potential role for FLT3 inhibitors and hypomethylating agents
t(1;19)/E2A-PBX1 Higher risk Poor outcome cancelled on current therapy
t(12;21)/ETV6-RUNX1 Low risk
IKZF1 Poor prognosis. Present in 80% BCR-ABL1+ and Potential role for TKI, JAK inhibitors
also in BCR-ABL1 like
NUP214-ABL1 High risk Potential benefits from TKI
CRLF2 In 1/3 BCR-ABL1like, associated with Hispanic or Potential role for JAK inhibitors
Latino ethnicity and poor outcome
CREBBP Associated with drug resistance and relapse Potential benefit from histone deacetylase inhibitors
Minimal Residual Disease
Day 15 <0.01% Excellent outcome No benefit from second delayed intensification
Slow early responders Higher MRD = Higher risk of relapse Benefit from augmented delayed intensification
EOI >1% Dismal prognosis HSCT in first CR
4mo >0.1% Dismal outcome HSCT in first CR

CNS, Central nervous system; CR, complete remission; DI, DNA index; EOI, end of induction; HSCT, hematopoietic stem cell transplantation; MRD, minimal residual
disease; JAK, Janus kinase; TKI, tyrosine kinase inhibitor.

completion of remission induction, have a much more favorable clinical trials have incorporated MRD detection into the risk classi-
prognosis than do those who do not achieve this status. Patients fication system. Although MRD positivity is strongly associated with
who are in morphologic remission but have a postinduction known presenting risk features, it has independent prognostic strength
MRD level of 1% or more fare as poorly as those who do not achieve and is increasingly used in risk stratification of ALL in contemporary
clinical remission by conventional criteria (5% blasts). About half regimens.18
of all patients show a disease reduction to 104 or lower after only 2 Currently, pediatric ALL patients are typically classified into three
weeks of remission induction, and they appear to have an exception- risk groupslow-, intermediate-, and high-risk (also referred to as
ally good treatment outcome. The persistence of MRD (0.01%) standard-, high-, and very high-risk)which are categories based on
beyond 4 months from diagnosis was associated with an estimated age, leukocyte count at diagnosis, blast cell immunophenotype, and
70% cumulative risk of relapse. Patients with 0.1% MRD or more genotype, as well as early treatment response. More recently, gene
at 4 months had an especially dismal outcome. Most contemporary expression profiling of leukemic cells by the DNA microarray method
956 Part VII Hematologic Malignancies

has proved useful in identifying previously unrecognized genes whose corticosteroids to this regimen, although the benefit of these pulses
expression may have prognostic significance. The predictive power of and their optimal duration and frequency of administration in the
these newly identified expression signatures requires validation in context of contemporary therapy has not been established.19 Adjust-
prospective clinical trials. ing chemotherapy doses to maintain a white cell count between 2 to
3 109/L and neutrophil counts between 0.5 and 1.5 109/L has
been associated with a better clinical outcome. Overzealous use of
THERAPY, INCLUDING STEM CELL TRANSPLANTATION mercaptopurine, to the extent that neutropenia necessitates chemo-
therapy interruption, reduces overall dose intensity and is counter-
With the exception of patients with mature B-cell ALL, who are productive. The optimal duration of therapy remains unknown.
treated with short-term intensive chemotherapy, therapy for patients Attempts to shorten therapy duration from 24 months to 12 or 18
with ALL is administered over 2 to 3 years. Treatment starts with months have resulted in a significant increase in relapses. Several
a 4- to 6-week remission-induction phase aimed at eradicating the studies showed no advantage to prolonging treatment beyond 3 years.
initial leukemic cell burden and restoring normal hematopoiesis. The A small number of patients with particularly poor prognostic features
induction phase typically includes the administration of a glucocor- may undergo BM transplantation during first remission.
ticoid (prednisone or dexamethasone), vincristine, and at least a third Radiation therapy was the first modality that was successfully used
drug (asparaginase, anthracycline, or both). A three-drug induction to prevent CNS relapse. The effectiveness of cranial radiation as
regimen appears sufficient for most standard-risk cases, provided they preventive therapy was offset by substantial late effects in long-term
receive intensified postremission therapy. The benefit in long-term survivors, including learning disabilities, multiple endocrinopathies,
survival of using four or more drugs during induction is widely and an increased risk of second malignancies. Subsequent trials dem-
accepted in higher risk patients but less clear in lower risk patients. onstrated that in the context of optimal systemic and intrathecal
With this approach, 98% to 99% of patients can attain remission, as therapy, cranial irradiation can be reduced or even omitted altogether.
defined by fewer than 5% blasts in the BM and a return of neutrophil Patients with high-risk genetic features, large leukemic cell burden,
and platelet counts to near normal levels. Intrathecal chemotherapy T-lineage ALL, and leukemic cells in the cerebrospinal fluid, even
is usually initiated at the start of treatment. from iatrogenic introduction from a traumatic lumbar puncture at
After remission induction, consolidation (or intensification) is diagnosis, are at increased risk of CNS relapse and require more
given to eradicate drug-resistant residual leukemic cells. Therapy is intense CNS-directed therapy.20 Studies have successfully used triple
tailored to the leukemia subtype and risk group. All patients benefit intrathecal therapy with methotrexate, hydrocortisone, and cytara-
from a delayed intensification (or delayed reinduction), consisting of bine or intrathecal methotrexate alone. Systematically administered
using drugs similar to those used in remission induction therapy after agents, including high-dose methotrexate, dexamethasone, and aspar-
a 3-month period of a less intensive, interim maintenance chemo- aginase, also contribute to prevention of extramedullary relapse.
therapy. Double-delayed intensification with a second reinduction at Based on reports of more potent in vitro antileukemic activity and
week 32 of treatment improves outcome in patients with intermediate- better CNS penetration, dexamethasone has replaced prednisone in
risk leukemia but does not benefit patients with rapid early response. many continuation regimens. Prednisone remains the preferred gluco-
An augmented intensification regimen consisting of the administra- corticoid during induction because of the relative increased toxicity
tion of additional doses of vincristine and asparaginase during the associated with dexamethasone use.21 Polyethylene glycolconjugated
myelosuppression period after delayed intensification and sequential asparaginase, a long-acting and less allergenic form, is progressively
escalating-dose parental methotrexate followed by asparaginase replacing the native Escherichia coli and is being increasingly adminis-
improved the outcome of high-risk patients whose disease had tered intravenously instead of intramuscularly. Asparaginase derived
responded slowly to initial multiagent induction therapy. from Erwinia chrysanthemi has a short half-life, and its use is currently
After completion of induction and consolidation, patients receive limited to patients who are allergic to the E. coli formulations. The
a 2- to 2.5-year continuation (or maintenance) phase consisting of dose schedule for asparaginase should take into account the variability
low-intensity metronomic chemotherapy designed to eradicate any in the pharmacokinetic profile and potency among the different
residual leukemic cell burden. Weekly low-dose methotrexate and preparations.22 Intensifying asparaginase therapy during the early
daily oral mercaptopurine form the backbone of most continuation phase of treatment benefits high-risk patients, particularly those with
regimens. Many groups add regular pulses of vincristine and T-cell disease. Significant improvement was also reported in the
outcome of patients receiving early intensification consisting of inter-
mediate- or high-dose antimetabolite therapy.23 The optimal dose of
methotrexate depends on the leukemic cell genotype and phenotype,
Consolidation Therapy as well as host pharmacogenetic and pharmacokinetic parameters.
Methotrexate at 2.5g/m2 is adequate for most patients with standard-
The importance of a consolidation phase after remission induc- risk B-cell precursor ALL, but a higher dose (5g/m2) may benefit those
tion is undisputed, but the treatment regimen and duration vary with T-cell or high-risk B-cell precursor ALL. This observation is
in the different childhood ALL studies. Commonly used strategies consistent with the finding that T-lineage blast cells accumulate meth-
include high-dose methotrexate plus mercaptopurine, frequent otrexate polyglutamates less avidly than do B-lineage blast cells. The
pulses of vincristine and corticosteroid plus high-dose asparagi- increased ability of hyperdiploid ALL blasts cells to accumulate meth-
nase for 20 to 30 weeks, and reinduction treatment with the otrexate polyglutamate could partially explain the excellent outcome
same agents given during initial remission induction. Reinduction
treatment has become an integral component of contemporary
protocols. In one randomized study, double reinduction further High-Risk Central Nervous System Relapse
improved treatment outcome in patients with intermediate-risk
ALL, but additional pulses of vincristine and prednisone after a Patients with the following characteristics are at increased risk
single reinduction course were not beneficial, suggesting that the of CNS relapse and require more intense CNS-directed therapy:
increased dose intensity of other drugs, such as asparaginase, 1. Patients with high-risk genetic features
was responsible for the observed improvement. An augmented 2. Large leukemic cell burden
regimen including Capizzi methotrexate (escalating-dose intrave- 3. T-lineage ALL
nous methotrexate with no rescue followed by asparaginase) and 4. CNS-3 status (>5 WBC/L CSF with presence of
additional doses of vincristine and asparaginase during periods lymphoblasts on Cytospin)
of myelosuppression improved the outcome of patients with a 5. CNS-2 status (<5 WBC/L CSF with lymphoblasts)
slow early response to therapy. 6. Traumatic CSF with blasts
Chapter 64 Clinical Manifestations and Treatment of Acute Lymphoblastic Leukemia in Children 957

of children with hyperdiploid greater than 50 chromosomes per cell refractory leukemia (failure to enter morphologic remission after 46
ALL treated on low-intensity antimetabolite-based regimens. Leu- weeks of induction therapy), high level of MRD (>1%) after remis-
covorin rescue is necessary after treatment with high-dose methotrex- sion induction, persistent MRD after consolidation treatment, and
ate; however, overzealous rescue might counteract the antileukemic early hematologic relapse are candidates for allogeneic transplanta-
activity of methotrexate. Although the intensive asparaginase and tion. It is crucial to reduce residual disease to, or close to, undetect-
high-dose methotrexate treatment has significantly improved the able levels as outcome is superior if MRD is undetectable before
outcome for patients with T-cell ALL, the emergence of specific HSCT and worsens with increasing MRD levels at the time of
therapy such as the purine nucleoside analog nelarabine will likely HSCT. Treatment approaches for adolescents and young adults with
increase the tendency to assign patients with T-cell ALL to a specific ALL have evolved considerably with the widespread adoption of
treatment protocol or strata. pediatric-based protocols, which appears to have significantly
It is generally recommended to give mercaptopurine at bedtime to improved survival and decreased the need for HSCT in this age
patients with an empty stomach and to avoid taking it together with group. The benefit of allogeneic HSCT in infants with t(4;11) ALL
milk or milk products that contain xanthine oxidase, an enzyme that remains controversial and should be evaluated in the context of
can degrade the drug. About 10% of the population inherit one wild- emerging molecular therapies such as FLT3 inhibitors and DNA
type gene encoding thiopurine methyltransferase (TPMT) and one methyltransferase inhibitors. Patients with the recently identified
nonfunctional variant allele, resulting in intermediate enzyme activity, ETP ALL have a dismal prognosis (event-free survival of 22%), even
but one in 300 people inherits two nonfunctional variant alleles and though half of the patients received transplantation because of high
are completely deficient of this enzyme that catalyses the S-methylation MRD levels after remission induction. The therapeutic role of HSCT
of mercaptopurine to its inactive metabolite. Patients with heterozy- in this group of patients remains to be determined by studying a
gous and especially homozygous deficiency of TPMT are at high risk larger number of patients. Matched unrelated-donor or cord blood
of severe myelosuppression. Identification of these patients allows transplantation has yielded outcomes comparable to those obtained
physicians to selectively guide reductions in mercaptopurine dosage with matched related-donor HSCT and should be considered reason-
without modifying the dose of methotrexate. Substituting thiogua- able alternatives if a matched donor is not available. Many advances
nine for mercaptopurine during continuation therapy was associated have been made in stem cell transplantation, such as prevention of
with a high incidence of profound thrombocytopenia and hepatic graft-versus-host disease (GVHD), expansion of the pool of suitable
venoocclusive disease. Thioguanine use has therefore been limited to unrelated or related donors, donor selection and tissue typing, accel-
short pulses administered during consolidation therapy in some trials; eration of engraftment, enhancement of the graft-versus-leukemia
mercaptopurine is selected for prolonged administration. effect, and supportive care. Because improvements in transplantation
Because optimizing the administration of existing therapies is tend to parallel those in chemotherapy, the indications for transplan-
reaching its limit, further improvements in outcome will require the tation in newly diagnosed and relapsed patients should be reevaluated
development of therapeutic approaches directed against rational periodically. For example, the presence of Philadelphia chromosome
therapeutic targets. An example is the significant improvement in the is no longer a clear indication for transplantation with the advent of
outcome of Philadelphia chromosomepositive (Ph+) ALL with the TKIs.
advent of TKIs targeting the constitutively active BCR-ABL1 TKI in
ALL subset. Ph+ ALL has historically had an extremely poor outcome,
but recent studies have demonstrated dramatic improvements in ACUTE LYMPHOBLASTIC LEUKEMIA RELAPSE
treatment outcome with incorporation of BCR-ABL1 inhibitors into
Ph+ ALL treatment.24 Recent identification of novel chimeric fusions Most relapses occur during treatment or within the first 2 years after
involving kinases in ALL (e.g., NUP214ABL1 and STRN3JAK2) its completion, although relapses have been reported as late as 10
suggests that additional high-risk cases may benefit from targeted years after initial ALL diagnosis. The most common site of relapse is
therapies directed at kinase signaling. the BM.27 Relapse in extramedullary sites, such as the CNS and testes,
The expanded understanding of the biologic, immunologic, and has decreased to less than 5% and 2% respectively. Leukemia relapse
genetic heterogeneity of ALL has enabled development of several occasionally occurs at other sites, including the eye, ovary, uterus,
novel therapeutic strategies.25 Various monoclonal antibodies are bone, muscle, tonsil, kidney, mediastinum, pleura, and paranasal
showing promise in early clinical trials and may be incorporated into sinus. Extramedullary relapse in children with ALL frequently pres-
ALL regimens in the future. ents as an isolated clinical finding. However, in studies that included
Autologous transplantation has failed to improve outcome in MRD assays, many extramedullary recurrences were associated with
ALL. Comparisons between allogeneic HSC transplantation (HSCT) MRD in the BM. A small fraction of patients experience a recurrence
and intensive chemotherapy have yielded inconsistent results because of acute leukemia with an immunophenotype different from that
of the small numbers of patients studied and differences in case selec- determined at diagnosis. Some of the cases represent relapse of origi-
tion criteria. It is generally accepted that allogeneic HSCT is a treat- nal leukemic clones with a shift in immunophenotype, but others are
ment modality for patients with ALL who are predicted to respond secondary malignancies caused by the mutagenic effects of leukemia
poorly to intensive chemotherapy.26 At present, patients with treatment, especially from epipodophyllotoxin. Patients with isolated
BM relapse generally fare worse than those with isolated extramedul-
lary relapse.28 Factors indicating an especially poor prognosis are a
short initial remission and a T-cell immunophenotype. Other adverse
Dose Schedule factors include t(9;22). The presence of MRD at the end of second
remission induction is also a strong adverse prognostic indicator.
The biologically equivalent doses among the different formula- Although chemotherapy may secure a prolonged second remission in
tions of corticosteroids, thiopurines, and asparaginase are not children with ALL who experience late relapse (defined as >6 months
clear. Trials comparing such agents should be cautiously inter- after cessation of therapy), allogeneic HSCT is the treatment of
preted, taking into account the dose schedule used and the effect choice for patients who experience hematologic relapse during
of variability in the pharmacokinetic profile and potency among therapy or shortly thereafter and for those with T-cell ALL. Patients
the agents involved. Simple modification of the dose or schedule with late-onset isolated CNS relapse who had not received cranial
may result in significant differences in efficacy and toxicity. Also, irradiation as initial CNS-directed therapy have a very high remission
when comparing regimens containing high-dose intravenous retrieval rate, with a long-term prognosis approaching that of newly
methotrexate, the dose schedule of leucovorin rescue should not diagnosed patients in those who had a long initial remission before
be ignored because it plays a crucial role in modulating the activ- the CNS event.
ity and toxicity of methotrexate. Genome-wide studies using matched diagnosis and relapse samples
from the same patients are exploring the genetic basis of relapse.
958 Part VII Hematologic Malignancies

Although 90% of the patients exhibit differential gaining or losing Down syndrome are at increased risk of hyperglycemia and other
genetic lesions from diagnosis to relapse, most relapse samples are complications. Prophylactic use of trimethoprimsulfamethoxazole
clonally related to diagnosis samples. Relapse clones could be present (or pentamidine or atovaquone in patients with poor tolerance to
as minor populations at diagnosis and selected during treatment to trimethoprimsulfamethoxazole) successfully prevents Pneumocystis
emerge as the predominant clone at relapse displaying alterations of jiroveci (formerly carinii) pneumonia. Dental evaluation at diagnosis
genes that have been implicated in treatment resistance. Almost 20% and meticulous oral hygiene during chemotherapy minimize the oral
of relapsed cases have sequence or deletion mutations of CREBBP, complications of leukemia and its treatment. It is important to dis-
which impair histone acetylation and transcriptional regulation of tinguish between herpes simplex viral infection and chemotherapy-
CREBBP targets, suggesting that the mutations may confer drug induced oral mucositis. Occasionally, patients have nausea and
resistance and raising the possibility of using drugs to reverse the substantial pain on swallowing caused by esophageal herpes simplex
aberrant epigenetic programs, such as histone deacetylase inhibitors.29 viral infection, candidiasis, or both. Oral candidiasis occurs fre-
The next generation of deep sequencing technologies promises to quently, especially in young children. Azole compounds (e.g., fluco-
unravel many more if not the full repertoire of genetic alterations in nazole, itraconazole, ketoconazole) are frequently used to treat fungal
leukemia. Parallel gene expression studies, which have identified a infections. It should be recognized that they can inhibit cytochrome
proliferative gene signature that emerges at relapse and consistent P450 enzymes and increase the toxicities of various antileukemic
upregulation of genes such as survivin, provide attractive targets for agents, especially vincristine. On the other hand, concomitant
novel therapeutic intervention. administration of anticonvulsants that induce cytochrome P450
enzymes (e.g., phenytoin, phenobarbital, carbamazepine) increases
the systemic clearance of several antileukemic agents and may
SUPPORTIVE CARE adversely affect treatment outcome. Anticonvulsants that are less
likely to induce the activity of cytochrome P450 enzymes (e.g.,
Stringent supportive care significantly contributes to a favorable ALL Keppra) are recommended in patients receiving chemotherapy. Pho-
outcome and should be initiated at diagnosis because remission tosensitive skin rash can occur during antimetabolite therapy. The
induction is associated with an increased risk from cardiovascular, rashes are erythematous, maculopapular, similar to atopic eczema,
metabolic, and infectious complications. All febrile patients with or and most prominent on the face. Topical administration of simple
without documented infection should be given broad-spectrum intra- emollients or a weak steroid preparation and avoidance of external
venous antibiotics until an infectious disease can be excluded. Rapid exposure to sunlight should improve the skin condition. Patients with
turnover of leukemia cells before and immediately after the initiation Down syndrome tolerate methotrexate poorly; appropriate dose
of chemotherapy leads to metabolic disturbances, including hyperka- adjustment is indicated. During each clinic visit, a thorough review
lemia, hyperuricemia, hyperphosphatemia, and hypocalcemia. of all drugs should be undertaken because of potential adverse inter-
Patients with high levels of uric acid are at risk for the development actions among them. In fact, chemotherapy can also interact with
of acute renal failure secondary to uric acid deposition in the kidneys. various food (e.g., grapefruit) and supplements (e.g., St. Johns wort,
All patients require intravenous hydration to prevent or treat hyper- folic acid).31
uricemia and hyperphosphatemia. Allopurinol, a xanthine oxidase
inhibitor, can prevent uric acid formation. Rasburicase, a recombi-
nant urate oxidase that breaks down uric acid to allantoin (a readily LATE EFFECTS OF TREATMENT
excretable metabolite with five- to 10-fold higher solubility than uric
acid), is more effective than allopurinol but is associated with met- The most problematic late effects of contemporary ALL therapy
hemoglobinemia or hemolytic anemia in patients with glucose-6- include neuropsychological impairments, bone morbidity, and
phosphate dehydrogenase deficiency because hydrogen peroxide is a obesity. Although neuropsychologic deficits are well-recognized side
byproduct of the uric acid breakdown.30 Phosphate binders should effects of cranial irradiation, intrathecal and systemic chemotherapy
also be used to prevent or treat hyperphosphatemia. Transfusions (especially methotrexate) can also cause brain atrophy and spinal cord
should be administered slowly in patients with severe anemia to dysfunction and contribute to the development of neurocognitive
prevent congestive heart failure. In patients with extreme hyperleu- toxicities. Severe CNS toxicity has been attributed to cranial irradia-
kocytosis, packed red blood cell transfusion should be delayed until tion at doses of 2400 cGy or higher, but lower doses have also been
after leukocyte count is decreased to prevent complications of associated with long-term neuropsychological impairments, espe-
leukostasis. All blood products should be irradiated in patients cially in younger children. Obesity, which is most prevalent among
who are receiving immunosuppressive therapy to prevent GVHD. female survivors of childhood ALL, may be related to cranial radia-
Patients should avoid foods that may be contaminated with patho- tion and corticosteroids. Osteopenia, fractures, and osteonecrosis
gens and reduce salt intake, which could induce hypertension and have been observed in up to 30% of survivors of childhood ALL.
resultant seizure in patients receiving glucocorticoids during induc- Osteonecrosis, which can lead to significant pain, loss of function,
tion. Adolescents, obese individuals, and individuals with and total joint replacement, has been reported in approximately 8%
of children with ALL, with the highest frequency observed in those
diagnosed in adolescence. Ovarian and testicular function are rela-
Drug Interactions tively unaffected by most antileukemic therapy. Offspring of patients
successfully treated for childhood ALL are expected to be as normal
We have not yet reached a full understanding of the contribution as the general population. Second malignant neoplasms, including
of genetic polymorphisms to interindividual differences in drug malignant gliomas, meningiomas, and AML, occur with increased
effects to allow us to translate this new knowledge into clinical frequency in patients treated on regimens that include irradiation,
practice. However, by simply avoiding drug interactions, one can epipodophyllotoxins, or alkylating agents.
prevent increased toxicity or reduced efficacy of chemotherapy.
Phenytoin and phenobarbital induce the activity of cytochrome
P450 enzymes, significantly increasing the systemic clearance of FUTURE DIRECTIONS
several antileukemic agents that may adversely affect treatment
outcome. We substitute these anticonvulsants with gabapentin or As the cure rate approaches 90%, treatment response assessed by
Keppra in patients receiving antileukemic therapy. On the other MRD measurements of submicroscopic leukemia has emerged as a
hand, azole compounds (e.g., fluconazole, itraconazole, ketocon- powerful and independent prognostic indicator for gauging the
azole) can inhibit cytochrome P450 enzymes and increase the intensity of ALL therapy. Children at high risk of relapse may now
toxicities of various antileukemic agents, especially vincristine. benefit from early intensification of therapy. The next goal is to
reduce the intensity of therapy in children at very low risk of relapse,
Chapter 64 Clinical Manifestations and Treatment of Acute Lymphoblastic Leukemia in Children 959

hence avoiding undue toxicity. The successful elimination of preven- 14. Kang H, Wilson CS, Harvey RC, et al: Gene expression profiles predic-
tive cranial irradiation indicates that treatment reduction is feasible tive of outcome and age in infant acute lymphoblastic leukemia: A
if done with caution and appropriate substitution with less toxic Childrens Oncology Group study. Blood 119:1872, 2011.
alternatives. Global genome analysis, in addition to refining leukemia 15. Arico M, Schrappe M, Hunger SP, et al: Clinical outcome of
classification, is helping identify potential molecular targets for children with newly diagnosed Philadelphia chromosome-positive acute
therapy. Expanding the application of pharmacogenomics, a science lymphoblastic leukemia treated between 1995 and 2005. J Clin Oncol
that aims to define the genetic determinants of drug effects, will allow 28:4755, 2010.
further personalized therapy in the future. 16. Evans WE, Relling MV: Moving towards individualized medicine with
pharmacogenomics. Nature 429:464, 2004.
17. Stow P, Key L, Chen X, et al: Clinical significance of low levels of
minimal residual disease at the end of remission induction therapy in
REFERENCES childhood acute lymphoblastic leukemia. Blood 115:4657, 2010.
1. Schrappe M, Nachman J, Hunger S, et al: Educational symposium on 18. Schultz KR, Pullen DJ, Sather HN, et al: Risk- and response-based clas-
long-term results of large prospective clinical trials for childhood acute sification of childhood B-precursor acute lymphoblastic leukemia: A
lymphoblastic leukemia (1985-2000). Leukemia 24:253, 2010. combined analysis of prognostic markers from the Pediatric Oncology
2. Pui CH, Evans WE: Treatment of acute lymphoblastic leukemia. N Engl Group (POG) and Childrens Cancer Group (CCG). Blood 109:926,
J Med 354:166, 2006. 2007.
3. Pui CH, Robison LL, Look AT: Acute lymphoblastic leukaemia. Lancet 19. Eden TO, Pieters R, Richards S: Systematic review of the addition of
371:1030, 2008. vincristine plus steroid pulses in maintenance treatment for childhood
4. Bienemann K, Burkhardt B, Modlich S, et al: Promising therapy results acute lymphoblastic leukaemiaan individual patient data meta-analysis
for lymphoid malignancies in children with chromosomal breakage syn- involving 5,659 children1. Br J Haematol 149:722, 2010.
dromes (Ataxia teleangiectasia or Nijmegen-breakage syndrome): A ret- 20. Pui CH, Howard SC: Current management and challenges of malignant
rospective survey1. Br J Haematol 155:468, 2011. disease in the CNS in paediatric leukaemia. Lancet Oncol 9:257, 2008.
5. Schmiegelow K, Lausten TU, Baruchel A, et al: High concordance of 21. Teuffel O, Kuster SP, Hunger SP, et al: Dexamethasone versus prednisone
subtypes of childhood acute lymphoblastic leukemia within families: for induction therapy in childhood acute lymphoblastic leukemia: A
Lessons from sibships with multiple cases of leukemia. Leukemia 26:675, systematic review and meta-analysis. Leukemia 25:1232, 2011.
2011. 22. van den BH: Asparaginase revisited. Leuk Lymphoma 52:168, 2011.
6. Zelent A, Greaves M, Enver T: Role of the TEL-AML1 fusion gene in 23. Matloub Y, Bostrom BC, Hunger SP, et al: Escalating intravenous metho-
the molecular pathogenesis of childhood acute lymphoblastic leukaemia. trexate improves event-free survival in children with standard-risk acute
Oncogene 23:4275, 2004. lymphoblastic leukemia: A report from the Childrens Oncology Group.
7. Pui CH, Carroll WL, Meshinchi S, et al: Biology, risk stratification, and Blood 118:243, 2011.
therapy of pediatric acute leukemias: An update. J Clin Oncol 29:551, 24. Schultz KR, Bowman WP, Aledo A, et al: Improved early event-free
2011. survival with imatinib in Philadelphia chromosome-positive acute lym-
8. Coustan-Smith E, Mullighan CG, Onciu M, et al: Early T-cell precursor phoblastic leukemia: A childrens oncology group study. J Clin Oncol
leukaemia: A subtype of very high-risk acute lymphoblastic leukaemia1. 27:5175, 2009.
Lancet Oncol 10:147, 2009. 25. Pui CH, Jeha S: New therapeutic strategies for the treatment of acute
9. Zhang J, Ding L, Holmfeldt L, et al: The genetic basis of early T-cell lymphoblastic leukaemia. Nat Rev Drug Discov 6:149, 2007.
precursor acute lymphoblastic leukaemia. Nature 481:157, 2012. 26. Pulsipher MA, Peters C, Pui CH: High-risk pediatric acute lymphoblas-
10. Mullighan CG, Su X, Zhang J, et al: Deletion of IKZF1 and prognosis tic leukemia: To transplant or not to transplant? Biol Blood Marrow
in acute lymphoblastic leukemia. N Engl J Med 360:470, 2009. Transplant 17:S137, 2011.
11. Harvey RC, Mullighan CG, Chen IM, et al: Rearrangement of CRLF2 27. Bailey LC, Lange BJ, Rheingold SR, et al: Bone-marrow relapse in pae-
is associated with mutation of JAK kinases, alteration of IKZF1, diatric acute lymphoblastic leukaemia1. Lancet Oncol 9:873, 2008.
Hispanic/Latino ethnicity, and a poor outcome in pediatric B-progenitor 28. Gaynon PS, Qu RP, Chappell RJ, et al: Survival after relapse in childhood
acute lymphoblastic leukemia. Blood 115:5312, 2010. acute lymphoblastic leukemia: Impact of site and time to first relapsethe
12. Mullighan CG, Zhang J, Harvey RC, et al: JAK mutations in high-risk Childrens Cancer Group Experience1. Cancer 82:1387, 1998.
childhood acute lymphoblastic leukemia. Proc Natl Acad Sci U S A 29. Mullighan CG, Zhang J, Kasper LH, et al: CREBBP mutations in
106:9414, 2009. relapsed acute lymphoblastic leukaemia. Nature 471:235, 2011.
13. Ensor HM, Schwab C, Russell LJ, et al: Demographic, clinical, and 30. Howard SC, Jones DP, Pui CH: The tumor lysis syndrome. N Engl J
outcome features of children with acute lymphoblastic leukemia and Med 364:1844, 2011.
CRLF2 deregulation: Results from the MRC ALL97 clinical trial. Blood 31. Haidar C, Jeha S: Drug interactions in childhood cancer. Lancet Oncol
117:2129, 2011. 12:92, 2011.
Chapter 64 Clinical Manifestations and Treatment of Acute Lymphoblastic Leukemia in Children 959.e1

Key Words
Acute lymphoblastic leukemia
Acute lymphoblastic leukemia (ALL) epidemiology
Acute lymphoblastic leukemia (ALL) pathobiology
Chemotherapy
Central nervous system (CNS)
Concordant acute lymphoblastic leukemia (ALL)
Molecular targets
Minimal residual disease (MRD)
Risk classification

Vous aimerez peut-être aussi