Sie sind auf Seite 1von 7

CME

Pediatric Acute Lymphoblastic


Leukemia: From Diagnosis
to Prognosis
Warren Alperstein, MD; Mary Boren, MD; and Jennifer L. McNeer, MD, MS

Abstract
Warren Alperstein, MD, is a Resident in
Pediatric acute lymphoblastic leukemia is the most common childhood cancer. Although
Pediatrics, Comer Children’s Hospital, Pritz-
the appearance of the disease is often quite dramatic, there are many patients who pres- ker School of Medicine, University of Chi-
ent much more indolently, creating a diagnostic dilemma for the primary care pediatrician. cago. Mary Boren, MD, is a Resident in Pe-
The appropriate diagnostic work-up assesses the initial extent of disease and stability of a diatrics, Comer Children’s Hospital, Pritzker
patient, and provides information that is important for risk stratification. Such information School of Medicine, University of Chicago.
includes patient age and white blood cell count at diagnosis, leukemia immunophenotype, Jennifer L. McNeer, MD, MS, is an Attending
presence or absence of extramedullary disease, and blast cytogenetic abnormalities. After Physician, Section of Pediatric Hematol-
therapy is initiated, the response of the disease to treatment is key for predicting outcomes. ogy/Oncology/Stem Cell Transplant, Comer
Altogether, this information is used to guide overall treatment intensity. Chemotherapy is Children’s Hospital; and an Assistant Profes-
administered in sequential blocks or phases, and lasts for several years. In general, outcomes sor of Pediatrics, Pritzker School of Medi-
cine, University of Chicago.
are excellent and the majority of patients survive, but there are still subsets of patients who
Address correspondence to Jennifer
do not fare as well, either due to resistant or recurrent disease, or due to long-term and late
L. McNeer, MD, MS, 5841 S. Maryland Av-
effects of therapy. [Pediatr Ann. 2015;44(7):e168-e174.]
enue, MC 4060, Chicago, IL 60637; e-mail:
jmcneer@peds.bsd.uchicago.edu.
Disclosure: Jennifer L. McNeer discloses
consulting fees from Jazz Pharmaceuticals.
The remaining authors have no relevant fi-
nancial relationships to disclose.
10.3928/00904481-20150710-10
© Shutterstock

e168 Copyright © SLACK Incorporated


CME

A
previously healthy 3-year-old requested. Bone marrow aspirate and mately half of patients present with fe-
toddler presented to an outside biopsy were performed, which revealed ver, which is thought to be induced by
emergency room with fever and B-cell acute lymphoblastic leukemia pyrogenic cytokines released from leu-
abdominal pain. Physical examination (B-ALL). kemic cells. Half of patients have firm,
and computed tomography scan of the nontender lymphadenopathy. Young
abdomen suggested appendicitis, and PRESENTATION OF ALL children may present with a limp, bone
the patient underwent an uncomplicated Demographics pain, or unwillingness to walk because
appendectomy. However, on pathologic Acute lymphoblastic leukemia of leukemic infiltration of the perios-
evaluation there was no appendicitis. (ALL) is the most common childhood teum, bone, or a joint. Some may pres-
The boy was diagnosed with mesenteric cancer. There are about 2,500 to 3,000 ent with symptoms of anemia, namely
adenitis and discharged home. The fevers new cases of pediatric ALL diagnosed fatigue and lethargy. Rarely, ALL may
and abdominal pain continued, and bilat- in the United States each year, with a present with bleeding secondary to
eral leg pain developed. Upon evaluation peak incidence in children between thrombocytopenia or a serious infec-
by his primary pediatrician, otitis media ages 2 and 5 years.1 The exact patho- tion secondary to neutropenia. How-
was diagnosed and he was prescribed an- genic events leading to the develop- ever, peripheral blood counts can be
tibiotics. The fevers continued, and labo- ment of ALL are unknown. However, relatively normal, and there are not al-
ratory work was significant for elevated ways circulating blasts. Therefore ALL
inflammatory markers. Echocardiogram cannot be ruled out when the blood
and bone scan were obtained, and due to counts are not overtly abnormal, as in
enhancement of several vertebrae as well
Some may present with the illustrative case.
as the left ankle on bone scan, a referral symptoms of anemia,
to pediatric rheumatology was made. Laboratory Evaluation and
In addition to the above history, the
namely fatigue and Diagnosis
review of systems at the time of the lethargy. The initial evaluation includes a
rheumatology visit was significant for CBC with differential and review of
decreased oral intake over several weeks, the peripheral blood smear, examining
resulting in a 4-lb weight loss. During about 5% of cases are associated with carefully for the presence of leukemic
physical examination, the patient ap- inherited genetic syndromes. Those blasts, which are present at diagnosis
peared generally uncomfortable and was with Down syndrome have a 10- to in about 90% of patients.2 Hyperleu-
lying in bed in a flexed position, but there 30-times greater risk of developing kocytosis (>100 × 103 WBC/mcL)
were no focal findings. any type of leukemia, most commonly occurs at diagnosis in about 12% of
The patient was admitted for further ALL. Children with certain autosomal white children and in about 23% of
evaluation. A complete blood count recessive genetic diseases associated African-American children,2 who are
(CBC) revealed a white blood cell with chromosomal fragility such as more likely to be diagnosed with the
(WBC) count of 5.2 × 103/mcL, hemo- ataxia-telangiectasia, Nijmegen break- T-lymphoblastic immunophenotype
globin of 9.2 g/dL, and a platelet count age syndrome, and Bloom syndrome of ALL (T-ALL). It is important in all
of 102 × 103/mcL. The differential was have an increased risk of ALL. There is patients, but in particular those with
48% neutrophils, 45% lymphocytes, and no clear link between infectious expo- hyperleukocytosis to assess for tumor
7% monocytes. A comprehensive meta- sure and the development of ALL. On lysis syndrome (TLS) by obtaining a
bolic panel (CMP) was unremarkable. the other hand, exposure to high-dose CMP and uric acid level. Tumor lysis
C-reactive protein was elevated at 162 radiation and benzene are recognized syndrome can occur spontaneously or
mg/L (normal <5 mg/L) and erythrocyte risk factors for developing ALL, but with the initiation of chemotherapy,
sedimentation rate was 162 mm/h (refer- these associations are rarely found in and is characterized by hyperphospha-
ence range 0-15 mm/h). Lactate dehy- children.2 temia, hyperkalemia, hypocalcemia,
drogenase (LDH) was elevated at 511 and hyperuricemia that occur when
U/L (normal 116-245 U/L), as was the Symptoms leukemic cells rupture and release their
ferritin, which was 465 ng/mL (normal, The symptoms of ALL can be very contents into the circulation. This most
20-300 ng/mL). nonspecific, may gradually become commonly occurs in tumors with a
Because of the anemia and throm- evident over a period of months, or high proliferation rate, and the risk of
bocytopenia, an oncology consult was they may appear acutely.3 Approxi- TLS can be gauged both by the WBC

PEDIATRIC ANNALS • Vol. 44, No. 7, 2015 e169


CME

count and the initial LDH. All patients lineage proteins such as CD19, CD22, row biopsy is obtained, occasionally
should have a chest radiograph to as- CD20, and CD79a, and negative for diagnostic difficulties arise in children
sess for a mediastinal mass, which is surface immunoglobulin, which appears with severe viral infections. At times,
more commonly observed in patients only on mature B cells. Cases of T-ALL the immune response may increase the
with T-ALL than B-ALL. express TdT and variable combinations production of normal precursor B cells
A bone marrow aspirate and biopsy is of T-lineage antigens such as CD1a, in the bone marrow, leading to suppres-
obtained at initial presentation to docu- CD3, CD4, CD5, CD7, and CD8.3 Be- sion of other hematopoiesis and resulting
ment the extent of marrow involvement sides determining the immunophenotype in modest granulocytopenia. It is usually
and to determine immunophenotype of the leukemia, the marrow sample is not difficult to distinguish such processes
(ie, B-cell versus T-cell disease). Lym- also processed for cytogenetic analysis from B-ALL because reactive popula-
phoblasts contain fine chromatin, small to aid with risk stratification. tions of lymphoblasts include cells from
nucleoli, and scant agranular cytoplasm At diagnosis, the presence or absence all stages of early B-cell development
(Figure 1). Acute lymphoblastic leuke- of extramedullary disease must be docu- and never replace the marrow. In con-
mia is defined as involvement of 25% mented. Central nervous system (CNS) trast, neoplastic lymphoblasts tend to
of marrow cellularity by lymphoblasts, involvement is defined as the presence of have a uniform immunophenotype repre-
but can be diagnosed in cases with fewer cranial nerve palsies by history or physi- senting one or another stage of early B-
blasts if a known recurring cytogenetic cal examination, or the finding of leuke- cell development, the hallmark of acute
abnormality is identified.4 The particu- mic blasts in the cerebrospinal fluid ob- leukemia.3
lar immunophenotype is identified using tained via lumbar puncture. In all boys,
flow cytometry, a process by which cells a testicular examination, and for some a RISK STRATIFICATION
are stained with a panel of antibodies to biopsy, is required to determine whether Risk stratification for children with
determine which markers are expressed there is testicular involvement. Patients ALL is of utmost importance as we are
on the surface of the malignant cells. In with extramedullary disease require increasingly tailoring therapy for each
B-ALL, the tumors cells are positive for modification of their therapy to intensify patient. By using risk-based treatment
terminal deoxynucleotidyl transferase therapy to these immune-privileged sites. we can provide less toxic treatment to
(TdT), an enzyme that is expressed only Although making a diagnosis is usu- patients with more favorable progno-
in immature B and T cells and certain B- ally straightforward once a bone mar- ses, and for those with higher-risk dis-
ease, we can intensify therapy early on
to improve outcomes.

Immunophenotype and National


Cancer Institute Risk Groups
The initial distinction to be made
is between B-cell and T-cell leukemia.
This initial distinction is critical as T-
ALL is more aggressive and requires
higher-intensity treatment. Histori-
cally, patients with T-ALL did not fare
as well as those with B-ALL, but with
modern therapeutic approaches the
outcomes for patients with T-ALL are
approaching those of patients with B-
ALL.5-8
Patients with B-ALL are initially
divided into standard- and high-risk
subsets at the time of diagnosis, based
on age and WBC count. These criteria
were established as prognostically im-
Figure 1. Bone marrow aspirate demonstrating acute lymphoblastic leukemia. Sheets of lymphoblasts portant by the National Cancer Institute
with characteristic smooth chromatin and scant cytoplasm are present. (NCI) in 1996 and retain significance

e170 Copyright © SLACK Incorporated


CME

TABLE 1.

Summary of B-Cell Acute Lymphoblastic Leukemia Risk Classification

Risk Factor Favorable Unfavorable


NCI risk group Age: 1-10 years Age: <1 year and ≥10 years
WBC <50 x 103/mcL WBC ≥50 x 103/mcL
Cytogenetics Hyperdiploidya Hypodiploidyb
Trisomy 4 & 10 t(9;22)
t(12;21) MLL rearrangement
iAMP21
Other characteristics CNS or testicular disease

Minimal residual disease <0.01% on Day 29 BM ≥0.01% on Day 29 BM


Abbreviations: BM, bone marrow; CNS, central nervous system; iAMP, intrachromosomal amplification of chromosome 21; NCI, National Cancer Institute; WBC, white blood cell.
a
Hyperdiploidy: >50 chromosomes or DNA index of ≥1.16.
b
Hypodiploidy: <44 chromosomes or DNA index <0.81.

to this day.9 Patients who are between than age 10 years with B-ALL will served prospectively at times of disease
ages 1 and 10 years at the time of diag- have favorable cytogenetics.11 reassessments to help gauge a patient’s
nosis and have a WBC <50 × 103/mcL Unfavorable cytogenetic abnor- disease status (ie, whether remission
are considered to have standard-risk malities include t(9;22), known as the has been achieved).
disease. As a group, these patients “Philadelphia chromosome,” which The cytogenetics of T-ALL are not
have a 4-year event-free survival (EFS) results in production of the BCR-ABL yet as clearly elucidated, and are not
rate of >80%.10 Patients who are ei- fusion protein, a constitutively active currently used in risk stratification
ther younger than age 1 year or age tyrosine kinase that requires incorpo- schemes. Recurrent abnormalities tend
10 years or older age at diagnosis, or ration of a tyrosine kinase inhibitor to be related to genes within the T-cell
have a WBC ≥50 × 103/mcL are con- (TKI) into therapy. The presence of receptor, located on chromosomes 7
sidered high risk, and require more in- t(9;22) increases with patient age, such and 14.11
tense treatment regimens. In particular, that only 2% to 4% of young patients
infants younger than age 1 year with (younger than age 10 years) harbor this Disease Response
ALL have very aggressive disease, and abnormality in their leukemic blasts, Ultimately, the most important pre-
treatment regimens exist specifically whereas up to 25% of adults with ALL dictor of outcome is disease response
for patients in this age group. do.12 Other unfavorable abnormali- to chemotherapy, and whether remis-
ties include hypodiploidy (<44 chro- sion can be achieved. Classically,
Cytogenetics mosomes per cell), rearrangement of remission was assessed morphologi-
Cytogenetic abnormalities within the MLL gene located at chromosome cally, and defined as <5% blasts in the
the leukemic blasts are an important 11q23, and intrachromosomal ampli- bone marrow. Now with more precise
component of risk stratification for fication of chromosome 21.11 Any of techniques (flow cytometry and poly-
patients with B-ALL, and individual these abnormalities would lead to a merase chain reaction), even minute
abnormalities are considered to be patient’s disease being considered high levels of residual leukemia can be
favorable, unfavorable, or intermedi- risk, regardless of initial age and WBC detected—referred to as minimal re-
ate. Favorable factors include a trans- count. sidual disease (MRD). For patients
location between chromosomes 12 There are other recurrent chromo- with B-ALL, the Children’s Oncology
and 21 [t(12;21)], which results in the somal aberrations that do not currently Group (COG) assesses MRD at two
ETV6-RUNX1 fusion protein, or hy- carry prognostic significance, such as time points during the initial 4-week
perdiploidy (>50 chromosomes per t(1;19) or trisomies of other various induction therapy: (1) in the periph-
leukemic cell), specifically with triso- chromosomes. Although these abnor- eral blood on day 8 and (2) in the bone
mies of chromosomes 4 and 10. Ap- malities may not lead to modification marrow on day 29 (at the end of induc-
proximately 50% of patients younger of treatment regimens, they can be ob- tion therapy), with a goal of <0.01%

PEDIATRIC ANNALS • Vol. 44, No. 7, 2015 e171


CME

account age, WBC, cytogenetics, and


disease response.

Treatment and Prognosis


The treatment for ALL occurs in se-
quential blocks of therapy (Figure 2).
This therapy consists of both initial and
delayed aggressive phases of therapy
(induction/consolidation and delayed
intensification) with an intervening less
intense phase of treatment known as
interim maintenance. After the comple-
tion of these blocks of more aggressive
therapy, which generally takes between
6 and 9 months, there is a prolonged
maintenance therapy that, on COG pro-
tocols, lasts approximately 2 years for
girls and 3 years for boys. During this
time, children feel relatively well, with
most of the therapy being low-dose oral
chemotherapy. Normal school and extra-
curricular activities are often resumed.
For all patients with ALL, both sys-
temic therapy and CNS-directed therapy
are required. Systemic chemotherapy
involves a combination of intravenous
and oral medications, with intramuscu-
lar drugs used in some cases as well.
Similar classes of drugs are used in in-
duction/consolidation and delayed in-
tensification, but with substitutions of
certain medications for one another—
for example, dexamethasone for pred-
nisone or doxorubicin for daunorubicin,
incorporating noncross-resistant agents
Figure 2. Schema of the Children’s Oncology Group therapy for acute lymphoblastic leukemia. Duration into later phases of therapy to eradicate
of consolidation is dependent on risk group (4 weeks for standard risk, 8 weeks for high risk). Protocols any remaining drug-resistant malignant
vary as to whether or not there is a second interim maintenance phase.
cells.14 CNS-directed treatment consists
of intrathecal chemotherapy adminis-
leukemic cells in the marrow by day for survival, but the NCI criteria of age tered at the time of lumbar punctures,
29 of therapy. The 5-year EFS rate for and WBC count, as well as favorable and is necessary regardless of CNS sta-
patients with <0.01% MRD (“nega- cytogenetics, retained prognostic sig- tus at diagnosis to protect against CNS
tive MRD”) at the end of induction is nificance.13 relapses. Those patients with overt in-
nearly 90%, and increasing levels of In summary, an individual patient’s volvement of the CNS often require cra-
MRD portend a worse prognosis, with treatment is adjusted based on the par- nial radiation at some point during their
a 5-year EFS rate of about 30% for the ticular risk group to which that patient treatment, in addition to intrathecal che-
group of patients with MRD >1% at the is ultimately assigned (Table 1). Al- motherapy.
end of induction therapy.13 In a multi- though the nuances of risk stratifica- Besides classic cytotoxic chemother-
variate analysis, MRD was found to be tion schemes vary slightly between dif- apy, in some cases more targeted therapy
the most important prognostic factor ferent cooperative groups, all take into is incorporated into treatment regimens.

e172 Copyright © SLACK Incorporated


CME

The paradigm for this is Philadelphia- up and monitoring for late effects of B, Jemal A. Childhood and adolescent
cancer statistics, 2014. CA Cancer J Clin.
chromosome positive (Ph+) ALL. The therapy. However, during therapy, acute 2014;64(2):83-103.
BCR-ABL fusion protein that is pro- toxicity such as nausea, vomiting, hair 2. Kaushansky K, Lichtman MA, Beutler E,
duced by the t(9;22) is a constitutively loss, myelosuppression, and neuropa- Kipps TJ, Seligsohn U, Prchal J. Williams
Hematology. New York, NY: McGraw-Hill
active tyrosine kinase and is the target thy can significantly impact patients’
Professional; 2010.
of the TKI imatinib mesylate. Although daily lives. At times, certain toxicities 3. Bunn HF, Aster JC. Pathophysiology of
imatinib monotherapy is effective in the such as infection and pancreatitis can Blood Disorders. 1st ed. New York, NY:
treatment of chronic myeloid leukemia be life-threatening, even with optimal McGraw-Hill Professional; 2011.
4. Vardiman JW, Thiele J, Arber DA, et al.
in chronic phase, it does not lead to du- supportive care. Furthermore, despite The 2008 revision of the World Health
rable remissions in Ph+ ALL when given our overall excellent outcomes, 10% to Organization (WHO) classification of
as a single agent.15 However, when used 20% of patients still relapse, many of myeloid neoplasms and acute leukemia:
rationale and important changes. Blood.
in conjunction with an intensive chemo- whom are considered to have standard- 2009;114(5):937-951.
therapy backbone, imatinib improves risk disease at diagnosis.10,13,23,24 Al- 5. Silverman LB, Gelber RD, Dalton VK, et al.
the outcomes for patients with Ph+ ALL though many children who are cured of Improved outcome for children with acute
lymphoblastic leukemia: results of Dana-
significantly.16,17 In fact, hematopoietic their ALL are quite healthy, late effects
Farber Consortium Protocol 91-01. Blood.
stem cell transplant (HSCT) was previ- such as neuropathy, avascular necrosis, 2001;97(5):1211-1218.
ously recommended in first remission cataracts, cardiomyopathy, and neuro- 6. Schrappe M, Reiter A, Ludwig WD, et al.
for all patients with Ph+ ALL, but this is cognitive deficits remain a significant Improved outcome in childhood acute lym-
phoblastic leukemia despite reduced use of
no longer necessary, as use of a TKI in concern. Finally, there are certain che- anthracyclines and cranial radiotherapy:
conjunction with chemotherapy affords motherapies used in leukemia regi- results of trial ALL-BFM 90. German-
patients equivalent, and perhaps better, mens, namely alkylators, topoisomer- Austrian-Swiss ALL-BFM Study Group.
Blood. 2000;95(11):3310-3322.
outcomes compared with HSCT.16,17 ase inhibitors, and anthracyclines that 7. Gaynon PS, Angiolillo AL, Carroll WL,
More recently a group of patients carry a risk of secondary malignancies et al. Long-term results of the children’s
has been identified using genome-wide or the development of myelodyspla- cancer group studies for childhood acute
lymphoblastic leukemia 1983-2002: a Chil-
analysis whose leukemic blasts do not sia. These are rare after ALL therapy;
dren’s Oncology Group Report. Leukemia.
contain the 9;22 translocation, but har- however, appropriate counseling for 2010;24(2):285-297.
bor a gene expression profile similar families at the time of diagnosis and 8. Möricke A, Zimmermann M, Reiter A, et
to that of patients with Ph+ ALL.18,19 monitoring for these complications af- al. Long-term results of five consecutive
trials in childhood acute lymphoblastic
These patients are considered to have ter therapy are required. leukemia performed by the ALL-BFM
BCR-ABL-like or Ph-like ALL, and study group from 1981 to 2000. Leukemia.
generally poor outcomes have been re- CONCLUSION 2010;24(2):265-284.
9. Smith M, Arthur D, Camitta B, et al. Uni-
ported. The majority of these patients In summary, ALL is the most com- form approach to risk classification and
have been found to have constitutive mon childhood cancer, but thankfully treatment assignment for children with
activation of cytokine receptors or tyro- is still rare. Initial symptoms can be acute lymphoblastic leukemia. J Clin On-
col. 1996;14(1):18-24.
sine kinase pathways.20 Further investi- nonspecific and indolent, requiring
10. Hunger SP, Lu X, Devidas M, et al. Im-
gation has revealed that aberrations in astute observation and evaluation by proved survival for children and adoles-
genes that regulate kinase signaling the primary care physician. Once a di- cents with acute lymphoblastic leukemia
are present in the majority of patients agnosis is made, oncologists carefully between 1990 and 2005: a report from the
Children’s Oncology Group. J Clin Oncol.
with Ph-like ALL, many of which are risk stratify patients so that treatment 2012;30(14):1663-1669.
predicted to respond to existing TKIs can be tailored accordingly. Therapy 11. Harrison CJ. Cytogenetics of paediatric and
or other signaling pathway inhibitors.21 is lengthy, with initial intense phases adolescent acute lymphoblastic leukaemia.
Br J Haematol. 2009;144(2):147-156.
Efforts are in progress to evaluate for followed by a prolonged maintenance 12. Bernt KM, Hunger SP. Current concepts in
these alterations at the time of diagno- phase; as our knowledge of the un- pediatric Philadelphia chromosome-pos-
sis, so that for affected patients, target- derlying mechanisms driving leuke- itive acute lymphoblastic leukemia. Front
Oncol. 2014;4:54.
ed therapy can quickly be incorporated mogenesis improves, our therapy will
13. Borowitz MJ, Devidas M, Hunger SP, et al.
into their chemotherapy regimens.21,22 continually become more specific to Clinical significance of minimal residual
With modern treatment, over 80% individual patients. disease in childhood acute lymphoblas-
of children diagnosed with ALL are tic leukemia and its relationship to other
prognostic factors: a Children’s Oncology
cured, and most of these children re- REFERENCES Group study. Blood. 2008;111(12):5477-
main quite healthy with careful follow 1. Ward E, DeSantis C, Robbins A, Kohler 5485.

PEDIATRIC ANNALS • Vol. 44, No. 7, 2015 e173


CME

14. Norton L, Simon R. The Norton-Simon Oncology Group study AALL0031. Leuke- like acute lymphoblastic leukemia. N Engl
hypothesis revisited. Cancer Treat Rep. mia. 2014;28(7):1467-1471. J Med. 2014;371(11):1005-1015.
1986;70(1):163-169. 18. Mullighan CG, Su X, Zhang J, et al. De- 22. Harvey R, Kang H, Roberts K, et al. Devel-
15. Druker BJ, Sawyers CL, Kantarjian H, et letion of IKZF1 and prognosis in acute opment and validation of a highly sensitive
al. Activity of a specific inhibitor of the lymphoblastic leukemia. N Engl J Med. and specific gene expression classifier to
BCR-ABL tyrosine kinase in the blast 2009;360(5):470-480. prospectively screen and identify B-precur-
crisis of chronic myeloid leukemia and 19. Den Boer ML, van Slegtenhorst M, De sor acute lymphoblastic leukemia (ALL)
acute lymphoblastic leukemia with the Menezes RX, et al. A subtype of child- patients with a Philadelphia Chromosome-
Philadelphia chromosome. N Engl J Med. hood acute lymphoblastic leukaemia Like (Ph-like or BCR-ABL-like) signature
2001;344(14):1038-1042. with poor treatment outcome: a genome- for therapeutic targeting and clinical in-
16. Schultz KR, Bowman WP, Aledo A, et al. wide classification study. Lancet Oncol. tervention. Blood ASH Meeting Abstracts.
Improved early event-free survival with 2009;10(2):125-134. 2013;122:826.
imatinib in Philadelphia chromosome- 20. Loh ML, Mullighan CG. Advances in the 23. Pui CH, Robison LL, Look AT. Acute
positive acute lymphoblastic leukemia: a genetics of high-risk childhood B-pro- lymphoblastic leukaemia. Lancet.
Children’s Oncology Group study. J Clin genitor acute lymphoblastic leukemia and 2008;371(9617):1030-1043.
Oncol. 2009;27(31):5175-5181. juvenile myelomonocytic leukemia: im- 24. Nguyen K, Devidas M, Cheng SC, et al.
17. Schultz KR, Carroll A, Heerema NA, et al. plications for therapy. Clin Cancer Res. Factors influencing survival after relapse
Long-term follow-up of imatinib in pedi- 2012;18(10):2754-2767. from acute lymphoblastic leukemia: a Chil-
atric Philadelphia chromosome-positive 21. Roberts KG, Li Y, Payne-Turner D, et al. dren’s Oncology Group study. Leukemia.
acute lymphoblastic leukemia: Children’s Targetable kinase-activating lesions in Ph- 2008;22(12):2142-2150.

e174 Copyright © SLACK Incorporated

Das könnte Ihnen auch gefallen