For children with acute lymphoblastic leukemia, the identification of those at higher risk of disease recurrence and modifying therapy based on this risk is a critical component to the provision of optimal care. The specific definitions of high-risk ALL vary across cooperative groups, but the themes are consistent, being largely based on leukemia biology and disease response. Intensification of conventional chemotherapy for those with high-risk disease has led to improved outcomes. It is anticipated that the development of rational targeted therapy for specific biologically unique subsets of children with leukemia will contribute to ongoing progress in improving the outcomes for children with acute lymphoblastic anemia.

Learning Objectives
  • To understand current criteria used to identify children with high-risk ALL

  • To understand therapeutic strategies for children with high-risk ALL

The chance of successful treatment for a child or adolescent with acute lymphoblastic leukemia (ALL) has improved dramatically over the last 50 years, with 5-year overall survival (OS) rates of approaching 90% for children receiving therapy in the developed world.1,2  The identification of active chemotherapy agents, understanding how to use them in combination, and treatment of the CSF as a sanctuary site were the steps that moved the disease from one that was uniformly fatal to one with a chance of cure of ∼75% by the 1980s.3-7  The majority of the improvement in outcomes in the last several decades can be attributed to modifications of the use of these original components and, to some extent, to intensifying therapy for those patients identified as having disease that is more difficult to cure. In this brief review, contemporary criteria used to identify patients at higher risk of relapse and the utility of intensification of therapy based on this risk are reviewed.

Risk classification is needed when there is uncertainty regarding a future event with estimates of the probability of the event being made based on past experience. For the purposes of care of a child with newly diagnosed ALL, the future event is relapse of disease and past experience is largely generated from data from cooperative clinical trials. Risk classification is important for determining the average outcome for a group and for determining whether interventions can modify the risk. Risk classification is not the same as being able to predict an individual's experience.

For the care of patients with ALL, the ideal risk classification system uses factors that are simple to measure and therefore easily generalizable and are able to be determined at the time of diagnosis or early on during the treatment course. Such a system then allows for risk-adapted therapy with treatment modifications made that may affect the likelihood of success.

The importance of understanding why certain patients are cured of their disease and others are not has been an integral part of the development of ALL therapy. Reporting in the Journal of the American Medical Association in 1966, Sidney Farber described 15 long-term survivors from the initial group of 1445 pediatric patients with ALL treated with chemotherapy. He notes, “it is still impossible to differentiate, at the time of diagnosis, the >99% whose lives will be prolonged by months or one or 2 years and the <1% who will survive 5 years of longer.”8  In the 1970s and 1980s, with increasing success of ALL therapy, investigators began to identify risk factors for relapsed disease. Patient age, disease burden measured in presenting WBC or “tumor bulk,” and evidence of disease response measured by changes in peripheral blood blast count or early BM morphologic assessment were found to be predictive of outcome.9-11  In the early 1990s, the 4 cooperative groups running clinical trials in pediatric ALL in North America all used risk definitions that included age and WBC, but all used different cutoff points for these continuous variables. The need to be able to compare and apply results across studies lead to a consensus conference in 1993 leading to the creation of the National Cancer Institute (NCI) criteria defining high-risk patients with B-precursor ALL as those 10 years of age or older or those having a presenting WBC count of 50 000/μL or greater.12 

The information used to allocate patients with ALL into different risk groups in contemporary trials can be divided into 3 categories: the patient's clinical features at the time of diagnosis, the disease characteristics, and measurements of initial response to therapy.

Within the Children's Oncology Group (COG), patients with ALL are currently classified into 7 different groups including infants, those with Philadelphia chromosome-positive (Ph+) disease, and those with T-ALL. The classification of the remainder of children with precursor B-ALL into 4 additional groups is described in Table 1. Factors used to define the risk group of children with ALL by other cooperative groups in contemporary clinical trials are similar in themes but vary in their details (Table 2).

Table 1.

COG classification of newly diagnosed B-precursor ALL (adapted from the COG classification system for B precursor ALL, AALL08B1)86 

COG classification of newly diagnosed B-precursor ALL (adapted from the COG classification system for B precursor ALL, AALL08B1)86
COG classification of newly diagnosed B-precursor ALL (adapted from the COG classification system for B precursor ALL, AALL08B1)86

Favorable genetics: hyperdiploidy with trisomies of chromosomes 4 and 10 or ETV6_RUNX1 fusion. Unfavorable characteristics: CNS2, induction failure, age 13 y and older, hypodiploid (<44 chromosomes or DNA index <0.81), MLL rearrangement, iAMP21. Note that infants and those with Ph+ disease are treated on separate trials.

HR, High risk by age and WBC count; PB, peripheral blood; and SR, standard risk by age and WBC count.

Table 2.

Risk classification strategies used by cooperative groups in recent or ongoing clinical trials for children with ALL

Risk classification strategies used by cooperative groups in recent or ongoing clinical trials for children with ALL
Risk classification strategies used by cooperative groups in recent or ongoing clinical trials for children with ALL

BFM, Berlin Frankfurt Munster; DFCI, Dana Farber Cancer Institute; SJCRH, St. Jude Children's Research Hospital; and MRC-UK-ALL, Medical Research Council-United Kingdom.

Age at diagnosis remains an important predictor of risk of relapse. Excluding infants, older age is consistently associated with a worse outcome, which can be explained in large part by variations in the biology of the disease with age. A smaller proportion of older children and adolescents with ALL have favorable cytogenetic findings, including ETV6-RUNX1 or hyperdiploidy, and a higher proportion have unfavorable genetic features including hypodiploidy and BCR-ABL (Ph+) disease.13  Contributing to the inferior OS of older patients is the observation that those >15 years of age treated for relapsed ALL have a significantly worse outcome than their younger peers.14 

Infants, those <1 year of age at the time of diagnosis, remain at very high risk of relapse, even with aggressive contemporary therapy.15,16  Risk stratification within the infant group is based on presence of MLL gene rearrangement, found in ∼75% and in the majority of younger infants. In addition, months of age at the time of diagnosis, with those <3 or 6 months fairing worse and high presenting WBC (>100 000 or >300 000) are important prognostic factors within this group of patients.17 

WBC count at the time of diagnosis of >50 000/μL is associated with less favorable outcome for children with B-lineage ALL.12  Age and WBC count are not accurate predictors of outcome for T-cell ALL.18,19 

Patients meeting the criteria of CNS 3 (having 5 WBCs/μL in the CSF with blasts in the cytospin) and, at least within the context of some studies, those with initial traumatic lumbar punctures with blasts (≥10 RBCs/μL) have an inferior event-free survival (EFS).20,21  Presence of CNS disease at the time of diagnosis is associated with other adverse prognostic factors.22  The prognostic implication of CNS 2 (<5 WBCs/μL with blasts) has been less consistent but likely also confers higher risk.23,24 

Patients with Down's syndrome and ALL have both a higher risk of relapse and a higher rate of treatment-related mortality (TRM) than non-Down's syndrome patients.25  Evaluation of >600 patients across 16 cooperative groups trials suggest that cutoff values for age and presenting WBC count for risk allocation in children with Down's syndrome should not be the same as those used for non-Down's syndrome patients.

Gender and ethnicity have not been used in the allocation of treatment intensity in clinical trials, except for the use of prolonged maintenance therapy for boys in some cooperative group protocols. Boys, irrespective of risk of testicular relapse, have a slightly inferior outcomes compared with girls.26  Black and Hispanic children with ALL have been shown to have an inferior outcome compared with Caucasian and Asian children.27  The reasons underlying these disparities are likely multiple, but include variations in ALL subtypes, host polymorphisms, and sociodemographic factors.28,29 

In earlier treatment eras, T-cell phenotype was considered an unfavorable prognostic feature, but over the last decade, with more intensive therapy and, especially if those with NCI standard risk features are excluded, patients with T-cell disease have been shown to have outcomes similar to those with precursor B-ALL.30-32  Unlike precursor B-ALL, prognostic factors for patients with T-cell ALL are less clear. A subset of T-cell ALL patients with a unique phenotype, described as early T-cell precursor ALL, characterized by being CD1a and CD8 negative with weak CD5 and coexpression of myeloid markers, have been shown to have an highly unfavorable prognosis by some groups and an intermediate-risk outcome by others.19,33-35 

The list of genetic alterations in childhood ALL that are associated with risk of relapse continues to rise.36  Some of these factors are well established as predictive of outcomes, whereas others remain to be validated.

Patients with hypodiploid leukemic cells have an unfavorable prognosis; those with fewer than 44 chromosomes are at the highest risk.37  A significant portion of ALL samples with low hypodiploidy will have additional unfavorable genetic findings, including TP53 alterations in >90% of cases.38 

Patients with Ph+ ALL [t(9:22)] historically had very poor outcomes, especially those with higher WBC and slow early response to therapy.1  A significant portion of patients with Ph+ ALL will have alterations in IKZF1, which is associated with chemotherapy resistance.39  With the introduction of tyrosine kinase inhibitors to intensive conventional therapy, the outlook has changed substantially.

Ph-like or BCR-ABL1-like ALL makes up 10%–15% of pediatric ALL and is identified by having a gene expression profile similar to that of Ph+ disease but without the presence of the BCR-ABL translocation.40,41  It is associated with a high frequency of IZKF1 alterations and is associated with a poor prognosis.42,43  This entity has not been included in risk stratification schemes in clinical trials to date but likely will be in the near future.

In infants with ALL, in whom translocations involving the MLL gene are very common, any MLL rearrangement confers a significantly worse prognosis.15  In older children, the prognostic importance of MLL is somewhat less clear and may be affected by age and translocation partner.44  Some, but not all, cooperative groups use the presence of MLL gene rearrangements as an indication for allocation to higher-risk therapy.

Initial descriptions of patients with intrachromosomal amplification of RUNX1 (iAMP) described very poor outcomes.45  More recent data suggest that intensification of conventional chemotherapy abrogate this risk.

With the availability of newer genomic techniques, multiple recurrent submicroscopic genetic alterations, including gene deletions, mutations, and amplifications, have been identified.46  Many of these alterations have not yet been shown to have clinical meaningfulness. One of the more common of these abnormalities is IKZF1 alterations, which occur in ∼15% of pediatric ALL cases and are associated with an unfavorable prognosis.41,47 IKZF1 alterations are found more commonly in patients meeting NCI high-risk criteria, those with Ph+ and Ph-like disease, and in patients with Down's syndrome.39,48,49 

Response to therapy as a prognostic indicator has been used since the early trials of ALL therapy. Failure to obtain remission after 4-6 weeks on induction therapy is a relatively rare event, occurring in ∼2%-3% of patients with ALL, and is associated with a poor OS.50  Those with induction failure are more likely to have other high-risk features, including older age and higher WBC, having T-cell disease, and having disease with unfavorable genetic features.

The rate of clearance of blasts from peripheral blood and BM by assessment of morphology has been shown to be prognostic and has been used for risk group allocation in many ALL trials.51,52  Prednisone response as measured in peripheral blood after 7 days of monotherapy and day 8 and day 15 of induction therapy assessments of BM morphology have been critical factors in risk assignment strategies in the recent past. These response tools remain valuable, including their use in contemporary trials in countries with variable resources in which minimal residual disease (MRD) measurements are not routinely feasible.53 

For the majority of pediatric ALL subgroups, the measurement of MRD has been shown to provide the most powerful prognostic information. Several recent large clinical trials have cemented this tool as the most important prognostic variable for the majority of children with both precursor B-ALL and T-cell ALL.19,54-56  In a study involving 2143 patients with NCI high-risk and standard-risk ALL, MRD measured by flow cytometry evaluated at the end of induction (day 29) was found to be the most significant prognostic factor, although NCI risk group maintained prognostic significance in multivariate analysis.54  Therapeutic interventions were not made based on MRD in this series of trials. Of the high-risk patients included in this study treated on the POG 9905 regimen, those who had an MRD level >0.01% at the end of induction had a 5-year EFS of 33% ± 8%) compared with 79% ± 4% (p < .001) for those who were negative.

The AIEOP-BFM 2000 study used PCR-based MRD measurements at the end of induction (day 33) and the end of consolidation (day 78) in 3184 patients with precursor B-ALL to prospectively risk stratify patients.55  Those who had MRD levels >10-3 at day 78, as well as those with a prednisone poor response, induction failure, or those with chromosomal translocations of t(4;11) or t(9:22), were allocated to the high-risk group. High-risk patients received intensified blocks of postconsolidation therapy. Even in the context of allocation of more intensive therapy, in multivariate analysis, MRD at the end of induction was the most important factor, although elevated WBC and ETV-RUNX1 status and DNA index retained predictive value. By MRD risk assignment alone, the EFS at 5 years was 91% in the standard-risk group compared with 77% in the intermediate-risk group and 50% in the high-risk group.

In the UKALL 2003 trial, patients were risk stratified based on clinical features, cytogenetics, and early BM response, as well as by MRD evaluation at the end of induction and postconsolidation. MRD risk status, with high risk defined by the presence of 0.001% MRD at the end of induction, was the most significant prognostic factor. Of the patients with evaluable MRD, 81% of all relapses occurred in those who were MRD high risk.56 

Within the context of the AIEOP BFM 2000 study, MRD measurement in patients with T-cell ALL was the most powerful prognostic factor for risk of relapse,19  although prednisone poor response and early T-cell phenotype also maintained prognostic value. The kinetics of MRD clearance in patients with T-ALL is different with than in those with precursor B-ALL, with only a small proportion (16%) being MRD negative by day 33. Conversely ∼50% are negative by day 78 and have a 7-year cumulative incidence of relapse of 8.5%. The incidence of relapse of those who are positive at day 78 was significantly higher and varied by level of MRD positivity, with those with >10-3 having an incidence of relapse of 44.7%.

In addition to the traditional factors used to assess risk of relapse for patients with ALL, consideration of the patient's compliance with therapy may need to be taken into account. In a study evaluating adherence with oral 6 mercaptopurine in the maintenance portion of treatment, an adherence rate of <95% was associated with a significantly increased risk of relapse.29 

Identification of groups at variable risk of relapse is done primarily to be able to inform modifications of therapy to limit short-term and long-term toxicities to those with more easily treatable disease and to intensify therapy for those with a worse prognosis. Improvement in outcome for higher-risk patients to date can largely be attributed to intensification of conventional chemotherapy. For a small percentage of children with very-high-risk ALL, intensification of therapy with the use of stem cell transplantation (SCT) in first remission has been applied. The use of biologically targeted therapy for those with less favorable disease to which a druggable target can be identified is clearly the most appealing intervention. To date, the success of this strategy is largely limited to the subset of patients with Ph+ leukemia.

Therapy for ALL is generally divided into 3 components, including remission induction, consolidation/intensification, and maintenance (also referred to as continuation).

Induction therapy for high-risk patients on most contemporary cooperative group trials use at least 4 drugs, including vincristine, an anthracycline, an asparaginase product, and either prednisone or dexamethasone. The most commonly used anthracyclines are doxorubicin or daunorubicin. Compared in a “therapeutic window” design as part of the induction regimen, the 2 agents were found to be equally effective.57  Mitoxantrone was shown to be superior to idarubicin in induction therapy for patients being treated in the relapsed setting, but to date is not commonly used as part of initial therapy.58  Data to support the choice of asparaginase product in induction is lacking, but most contemporary trials use pegylated asparaginase, with IV administration being demonstrated to be safe.59  Dexamethasone and prednisone are both used in induction regimens. With the limitation of variability on conversion ratios used between the 2 agents, data suggest that dexamethasone may be more effective, but is associated with increased risks of acute and long-term toxicities.60 

Intensification of therapy for high-risk patients has been done primarily during the 6-8 months of therapy after induction. Randomized trials evaluating postinduction intensification in high-risk patients over the last 2 decades have demonstrated significantly improved outcomes with some intensified regimens, but others showed no benefit (Table 3).

Table 3.

Selected randomized clinical trials evaluating intensification of postinduction therapy for patients with higher-risk ALL

Selected randomized clinical trials evaluating intensification of postinduction therapy for patients with higher-risk ALL
Selected randomized clinical trials evaluating intensification of postinduction therapy for patients with higher-risk ALL

CR, Complete remission; MTX, methotrexate; and PPR, pathological partial response.

Improvements in outcomes for higher-risk patients in serial, nonrandomized trials have also been attributed to modifications of postinduction therapy, including the use of dexamethasone as the corticosteroid, the intensive use and formulation of asparaginase,4,56  and intensification of consolidation/reinduction treatment.32 

Intensification of conventional chemotherapy has also been investigated within specific biological subsets of patients with ALL. Intrachromosomal amplification of chromosome 21 (iAMP) was found to be associated with a significantly inferior EFS in several cooperative group trials.45,61  Subsequent analysis identified that the poor outcome was driven primarily by patients with iAMP allocated to a standard-risk group and therefore treated with less intense therapies.62  Intensifying chemotherapy for this group of patients dramatically decreased the risk of relapse at 5 years from 70% to 16% over the course of 2 sequential trials.63 

Similarly, patients with t(1:19) were found to have a significantly inferior outcome when treated with a regimen that was antimetabolite based,64  whereas when treated with intensified therapy, the outcome is significantly more favorable.65,66 

Ongoing trials continue to evaluate the possibility of improving outcomes of children with high-risk ALL by intensifying postinduction chemotherapy, including the COG studies evaluating the impact of the addition of clofarabine for patients with very-high-risk precursor B-ALL and nelarabine for those with high-risk T-cell ALL. As opposed to improving outcomes with ongoing manipulation and intensification of conventional ALL therapies, identification of biologically driven subsets of ALL with unique druggable targets will likely lead to the next significant advances in ALL therapy. To date, this strategy has been shown to be highly effective for one subset of ALL, that of Ph+ ALL, which accounts for ∼3%-4% of cases. Favorable results for treatment of children with Ph+ ALL with the tyrosine kinase inhibitor imatinib in addition to intensive multiagent chemotherapy, leading to a 5-year disease-free survival (DFS) of 70%, has changed the standard of care for this group from that allocation to SCT in first remission (Figure 1).67-69 

Figure 1.

Data from COG protocol AALL0031. Shown are data with long-term follow-up of patients from COG protocol AALL0031 with Ph+ leukemia with no difference in 5-year DFS for patients in Cohort 5 (treated with continuous imatinib plus multiagent chemotherapy) compared with hematopoietic SCT in all cohorts, either from a related or unrelated donor. (Used with permission from Schultz et al, 2014.67 )

Figure 1.

Data from COG protocol AALL0031. Shown are data with long-term follow-up of patients from COG protocol AALL0031 with Ph+ leukemia with no difference in 5-year DFS for patients in Cohort 5 (treated with continuous imatinib plus multiagent chemotherapy) compared with hematopoietic SCT in all cohorts, either from a related or unrelated donor. (Used with permission from Schultz et al, 2014.67 )

Close modal

The identification of a subset of ALL patients who are Ph− but have a similar gene expression to those who are Ph+, called BCR-ABL1-like ALL, begs the question as to whether this group, that has been shown to have a worse prognosis with standard high-risk therapy,70  may benefit from targeted therapy, potentially with tyrosine kinase or JAK2 inhibitors.71  There are several published case reports describing children with ALL with induction failure found to have a BCR-ABL1-like disease who have subsequently had dramatic responses to tyrosine kinase inhibitors.72-74  Mechanisms for screening patients for BCR-ABL-like disease, evaluating for likelihood of response to tyrosine kinase inhibitors or JAK2 inhibitors in vitro, and considering the addition of targeted therapies to conventional chemotherapy are being planned within COG (Mignon Loh, personal communication).

Currently, there remains a population of children that can be identified as having an exceptionally high risk of having relapsed disease, for whom relatively poor outcomes with conventional therapy (including SCT) are expected. In the context of BFM therapy, patients with high MRD levels at the end of induction and consolidation had a 5-year DFS of 45% that was not improved by SCT.75  The 4-year OS for patients with hypodiploid ALL treated in a recent COG study with very intensive chemotherapy was 54%. With the limitations of small numbers, this study did not show an advantage of SCT as a component of therapy or improvement with intensified chemotherapy compared with historical controls.67  A retrospective review combining patients from multiple clinical trials showed a 32% 10-year OS for patients with initial induction failure.50  For those older than 6 years and those with T-cell ALL, there was a trend for improved outcome with SCT. A study of sibling donor allogeneic SCT in first remission versus chemotherapy for a group of patients defined as having very-high-risk disease, including those with induction failure, showed a DFS advantage with SCT.76  Lastly, despite intensified therapy, including high-dose cytarabine and methotrexate, infants with ALL with MLL translocations continue to have a poor outcome, with 5-year EFS between 30% and 40%.15  The use of SCT to intensify therapy for infants has not shown clear benefit, but the risk of relapse may be reduced for those in the highest risk group.16,77 

Morbidity, both short-term and long-term, affects every patient treated for ALL. Despite improvements in supportive care, mortality as a consequence of therapy persists as a cause of death. A recent meta-analysis of randomized trials of newly diagnosed pediatric patients with ALL reported that the proportion of nonrelapse mortality was 3.6% overall and 1.4% in induction. In both periods, those classified as having high-risk leukemia had a significantly increased risk of nonrelapse mortality.78  Definitions of TRM in childhood ALL are variable and the majority of published clinical trials do not include descriptions of the definitions used.79  Variability in defining and reporting of TRM involves both inclusion and exclusion of deaths occurring immediately before the initiation of therapy or after completion of therapy, those that occur after SCT, and those from other causes not strictly related to disease or its therapy. The lack of definition of TRM makes comparison across trials difficult.

In the NOPHO ALL-92 and 2000 trials, 25% of all deaths on study were considered treatment related, with the majority being secondary to infection (72%), primarily bacterial infections, with bleeding or thrombosis, organ toxicity, or complications of tumor burden accounting for most of the remainder.80  Patients with high-risk ALL had a significantly higher risk of TRM than those with standard- or intermediate-risk disease (6.7% ± 0.9%, 1.7% ± 0.4%, and 2.4% ± 0.5%, respectively; p < .001). In the ALL IC-BFM 2002 trial, which expanded the access to trial participation to a large number of centers with relatively more limited resources, TRM accounted for 5% of deaths for those patients in complete remission, affecting 3% of those being treated for standard-risk disease and 13% of those being treated with high-risk regimens.53  Other factors that have been associated with TRM include age (<1 year or >15 years), female sex, SCT in first complete remission, and earlier treatment era.81,82  Patients with Down's syndrome and ALL constitute a unique group, with substantially higher TRM compared with non-Down's syndrome patients (7.7%±1% vs 2% ± <1% respectively, p < .0001) and the majority attributable to infection.25 

The use of antibacterial prophylaxis during periods of prolonged neutropenia in adult patients with acute leukemia is supported by data from a meta-analysis showing its use to be associated with decreased risk of death,83  and is recommended in adult supportive care guidelines from both the National Comprehensive Cancer Network and the American Society of Clinical Oncology.84,85  Data on the use of antibacterial prophylaxis in children are extremely limited. Studies evaluating the utility of levofloxacin prophylaxis during ALL induction and in those being treated with intensive therapy for relapsed disease are being done through the Dana-Farber Cancer Institute consortium and COG, respectively.

The goals of therapy for every child newly diagnosed with ALL include maximizing the likelihood of cure while minimizing the risks of both acute and long-term side effects. Risk stratification, intensification of therapy for higher-risk patients, and, in the case of Ph+ ALL, adding targeted therapy have accounted for significant improvements in outlook for children and adolescents with high-risk disease. Despite progress, stratification schemes remain imperfect, with 1/3 of deaths in children with ALL in those who initially meet the criteria of favorable-risk disease.26  The next substantial steps in the efforts to improve the outcomes of children with ALL will likely include clinically accessible tools for measuring individual patient's disease features, with the potential for the identification of druggable targets. In addition, refining strategies for preventing TRM will allow for the tradition of remarkable progress in the care of children with ALL to continue.

Conflict-of-interest disclosure: The author declares no competing financial interests. Off-label drug use: None disclosed.

Sarah Alexander, MD, The Hospital for Sick Children, 555 University Ave, Toronto, Ontario M5G 1X8, Canada; Phone: (416)813-7654, ext. 204068; Fax: (416)813-5327; e-mail: sarah.alexander@sickkids.ca.

1
Arico
 
M
Schrappe
 
M
Hunger
 
SP
, et al. 
Clinical outcome of children with newly diagnosed Philadelphia chromosome-positive acute lymphoblastic leukemia treated between 1995 and 2005
J Clin Oncol
2010
, vol. 
28
 
31
(pg. 
4755
-
4761
)
2
Schrappe
 
M
Nachman
 
J
Hunger
 
S
, et al. 
Educational symposium on long-term results of large prospective clinical trials for childhood acute lymphoblastic leukemia (1985-2000)
Leukemia
2010
, vol. 
24
 
2
(pg. 
253
-
254
)
3
Salzer
 
WL
Devidas
 
M
Carroll
 
WL
, et al. 
Long-term results of the pediatric oncology group studies for childhood acute lymphoblastic leukemia 1984-2001: a report from the children's oncology group
Leukemia
2010
, vol. 
24
 
2
(pg. 
355
-
370
)
4
Silverman
 
LB
Stevenson
 
KE
O'Brien
 
JE
, et al. 
Long-term results of Dana-Farber Cancer Institute ALL Consortium protocols for children with newly diagnosed acute lymphoblastic leukemia (1985-2000)
Leukemia
2010
, vol. 
24
 
2
(pg. 
320
-
334
)
5
Gaynon
 
PS
Angiolillo
 
AL
Carroll
 
WL
, et al. 
Long-term results of the children's cancer group studies for childhood acute lymphoblastic leukemia 1983-2002: a Children's Oncology Group Report
Leukemia
2010
, vol. 
24
 
2
(pg. 
285
-
297
)
6
Pui
 
CH
Pei
 
D
Sandlund
 
JT
, et al. 
Long-term results of St Jude Total Therapy Studies 11, 12, 13A, 13B, and 14 for childhood acute lymphoblastic leukemia
Leukemia
2010
, vol. 
24
 
2
(pg. 
371
-
382
)
7
Moricke
 
A
Zimmermann
 
M
Reiter
 
A
, et al. 
Long-term results of five consecutive trials in childhood acute lymphoblastic leukemia performed by the ALL-BFM study group from 1981 to 2000
Leukemia
2010
, vol. 
24
 
2
(pg. 
265
-
284
)
8
Farber
 
S
Chemotherapy in the treatment of leukemia and Wilms' tumor
JAMA
1966
, vol. 
198
 
8
(pg. 
826
-
836
)
9
Riehm
 
H
Reiter
 
A
Schrappe
 
M
, et al. 
Corticosteroid-dependent reduction of leukocyte count in blood as a prognostic factor in acute lymphoblastic leukemia in childhood (therapy study ALL-BFM 83) [article in German]
Klin Padiatr
1987
, vol. 
199
 
3
(pg. 
151
-
160
)
10
Mastrangelo
 
R
Poplack
 
D
Bleyer
 
A
Riccardi
 
R
Sather
 
H
D'Angio
 
G
Report and recommendations of the Rome workshop concerning poor-prognosis acute lymphoblastic leukemia in children: biologic bases for staging, stratification, and treatment
Med Pediatr Oncol
1986
, vol. 
14
 
3
(pg. 
191
-
194
)
11
Hammond
 
D
Sather
 
H
Nesbit
 
M
, et al. 
Analysis of prognostic factors in acute lymphoblastic leukemia
Med Pediatr Oncol
1986
, vol. 
14
 
3
(pg. 
124
-
134
)
12
Smith
 
M
Arthur
 
D
Camitta
 
B
, et al. 
Uniform approach to risk classification and treatment assignment for children with acute lymphoblastic leukemia
J Clin Oncol
1996
, vol. 
14
 
1
(pg. 
18
-
24
)
13
Moricke
 
A
Zimmermann
 
M
Reiter
 
A
, et al. 
Prognostic impact of age in children and adolescents with acute lymphoblastic leukemia: data from the trials ALL-BFM 86, 90, and 95
Klin Padiatr
2005
, vol. 
217
 
6
(pg. 
310
-
320
)
14
Freyer
 
DR
Devidas
 
M
La
 
M
, et al. 
Postrelapse survival in childhood acute lymphoblastic leukemia is independent of initial treatment intensity: a report from the Children's Oncology Group
Blood
2011
, vol. 
117
 
11
(pg. 
3010
-
3015
)
15
Pieters
 
R
Schrappe
 
M
De Lorenzo
 
P
, et al. 
A treatment protocol for infants younger than 1 year with acute lymphoblastic leukaemia (Interfant-99): an observational study and a multicentre randomised trial
Lancet
2007
, vol. 
370
 
9583
(pg. 
240
-
250
)
16
Mann
 
G
Attarbaschi
 
A
Schrappe
 
M
, et al. 
Improved outcome with hematopoietic stem cell transplantation in a poor prognostic subgroup of infants with mixed-lineage-leukemia (MLL)-rearranged acute lymphoblastic leukemia: results from the Interfant-99 Study
Blood
2010
, vol. 
116
 
15
(pg. 
2644
-
2650
)
17
Brown
 
P
Treatment of infant leukemias: challenge and promise
Hematology Am Soc Hematol Educ Program
2013
, vol. 
2013
 (pg. 
596
-
600
)
18
Pullen
 
J
Shuster
 
JJ
Link
 
M
, et al. 
Significance of commonly used prognostic factors differs for children with T cell acute lymphocytic leukemia (ALL), as compared to those with B-precursor ALL. A Pediatric Oncology Group (POG) study
Leukemia
1999
, vol. 
13
 
11
(pg. 
1696
-
1707
)
19
Schrappe
 
M
Valsecchi
 
MG
Bartram
 
CR
, et al. 
Late MRD response determines relapse risk overall and in subsets of childhood T-cell ALL: results of the AIEOP-BFM-ALL 2000 study
Blood
2011
, vol. 
118
 
8
(pg. 
2077
-
2084
)
20
Pui
 
CH
Thiel
 
E
Central nervous system disease in hematologic malignancies: historical perspective and practical applications
Semin Oncol
2009
, vol. 
36
 
4 Suppl 2
(pg. 
S2
-
S16
)
21
Shaikh
 
F
Voicu
 
L
Tole
 
S
, et al. 
The risk of traumatic lumbar punctures in children with acute lymphoblastic leukaemia
Eur J Cancer
2014
, vol. 
50
 
8
(pg. 
1482
-
1489
)
22
Levinsen
 
M
Taskinen
 
M
Abrahamsson
 
J
, et al. 
Clinical features and early treatment response of central nervous system involvement in childhood acute lymphoblastic leukemia
Pediatr Blood Cancer
2014
, vol. 
61
 
8
(pg. 
1416
-
1421
)
23
Burger
 
B
Zimmermann
 
M
Mann
 
G
, et al. 
Diagnostic cerebrospinal fluid examination in children with acute lymphoblastic leukemia: significance of low leukocyte counts with blasts or traumatic lumbar puncture
J Clin Oncol
2003
, vol. 
21
 
2
(pg. 
184
-
188
)
24
Dutch Childhood Oncology G
te Loo
 
DM
Kamps
 
WA
van der Does-van den Berg
 
A
van Wering
 
ER
de Graaf
 
SS
Prognostic significance of blasts in the cerebrospinal fluid without pleiocytosis or a traumatic lumbar puncture in children with acute lymphoblastic leukemia: experience of the Dutch Childhood Oncology Group
J Clin Oncol
2006
, vol. 
24
 
15
(pg. 
2332
-
2336
)
25
Buitenkamp
 
TD
Izraeli
 
S
Zimmermann
 
M
, et al. 
Acute lymphoblastic leukemia in children with Down syndrome: a retrospective analysis from the Ponte di Legno study group
Blood
2014
, vol. 
123
 
1
(pg. 
70
-
77
)
26
Hunger
 
SP
Lu
 
X
Devidas
 
M
, et al. 
Improved survival for children and adolescents with acute lymphoblastic leukemia between 1990 and 2005: a report from the children's oncology group
J Clin Oncol
2012
, vol. 
30
 
14
(pg. 
1663
-
1669
)
27
Kadan-Lottick
 
NS
Ness
 
KK
Bhatia
 
S
Gurney
 
JG
Survival variability by race and ethnicity in childhood acute lymphoblastic leukemia
JAMA
2003
, vol. 
290
 
15
(pg. 
2008
-
2014
)
28
Yang
 
JJ
Cheng
 
C
Devidas
 
M
, et al. 
Ancestry and pharmacogenomics of relapse in acute lymphoblastic leukemia
Nat Genet
2011
, vol. 
43
 
3
(pg. 
237
-
241
)
29
Bhatia
 
S
Landier
 
W
Shangguan
 
M
, et al. 
Nonadherence to oral mercaptopurine and risk of relapse in Hispanic and non-Hispanic white children with acute lymphoblastic leukemia: a report from the children's oncology group
J Clin Oncol
2012
, vol. 
30
 
17
(pg. 
2094
-
2101
)
30
Goldberg
 
JM
Silverman
 
LB
Levy
 
DE
, et al. 
Childhood T-cell acute lymphoblastic leukemia: the Dana-Farber Cancer Institute acute lymphoblastic leukemia consortium experience
J Clin Oncol
2003
, vol. 
21
 
19
(pg. 
3616
-
3622
)
31
Pui
 
CH
Evans
 
WE
Treatment of acute lymphoblastic leukemia
N Engl J Med
2006
, vol. 
354
 
2
(pg. 
166
-
178
)
32
Moricke
 
A
Reiter
 
A
Zimmermann
 
M
, et al. 
Risk-adjusted therapy of acute lymphoblastic leukemia can decrease treatment burden and improve survival: treatment results of 2169 unselected pediatric and adolescent patients enrolled in the trial ALL-BFM 95
Blood
2008
, vol. 
111
 
9
(pg. 
4477
-
4489
)
33
Patrick
 
K
Wade
 
R
Goulden
 
N
, et al. 
Outcome for children and young people with Early T-cell precursor acute lymphoblastic leukaemia treated on a contemporary protocol, UKALL 2003
Br J Haematol
2014
, vol. 
166
 
3
(pg. 
421
-
424
)
34
Coustan-Smith
 
E
Mullighan
 
CG
Onciu
 
M
, et al. 
Early T-cell precursor leukaemia: a subtype of very high-risk acute lymphoblastic leukaemia
Lancet Oncol
2009
, vol. 
10
 
2
(pg. 
147
-
156
)
35
Inukai
 
T
Kiyokawa
 
N
Campana
 
D
, et al. 
Clinical significance of early T-cell precursor acute lymphoblastic leukaemia: results of the Tokyo Children's Cancer Study Group Study L99-15
Br J Haematol
2012
, vol. 
156
 
3
(pg. 
358
-
365
)
36
Teachey
 
DT
Hunger
 
SP
Predicting relapse risk in childhood acute lymphoblastic leukaemia
Br J Haematol
2013
, vol. 
162
 
5
(pg. 
606
-
620
)
37
Nachman
 
JB
Heerema
 
NA
Sather
 
H
, et al. 
Outcome of treatment in children with hypodiploid acute lymphoblastic leukemia
Blood
2007
, vol. 
110
 
4
(pg. 
1112
-
1115
)
38
Holmfeldt
 
L
Wei
 
L
Diaz-Flores
 
E
, et al. 
The genomic landscape of hypodiploid acute lymphoblastic leukemia
Nat Genet
2013
, vol. 
45
 
3
(pg. 
242
-
252
)
39
Mullighan
 
CG
Miller
 
CB
Radtke
 
I
, et al. 
BCR-ABL1 lymphoblastic leukaemia is characterized by the deletion of Ikaros
Nature
2008
, vol. 
453
 
7191
(pg. 
110
-
114
)
40
Den Boer
 
ML
van Slegtenhorst
 
M
De Menezes
 
RX
, et al. 
A subtype of childhood acute lymphoblastic leukaemia with poor treatment outcome: a genome-wide classification study
Lancet Oncol
2009
, vol. 
10
 
2
(pg. 
125
-
134
)
41
Mullighan
 
CG
Su
 
X
Zhang
 
J
, et al. 
Deletion of IKZF1 and prognosis in acute lymphoblastic leukemia
N Engl J Med
2009
, vol. 
360
 
5
(pg. 
470
-
480
)
42
van der Veer
 
A
Waanders
 
E
Pieters
 
R
, et al. 
Independent prognostic value of BCR-ABL1-like signature and IKZF1 deletion, but not high CRLF2 expression, in children with B-cell precursor ALL
Blood
2013
, vol. 
122
 
15
(pg. 
2622
-
2629
)
43
Loh
 
ML
Zhang
 
J
Harvey
 
RC
, et al. 
Tyrosine kinome sequencing of pediatric acute lymphoblastic leukemia: a report from the Children's Oncology Group TARGET Project
Blood
2013
, vol. 
121
 
3
(pg. 
485
-
488
)
44
Moorman
 
AV
Ensor
 
HM
Richards
 
SM
, et al. 
Prognostic effect of chromosomal abnormalities in childhood B-cell precursor acute lymphoblastic leukaemia: results from the UK Medical Research Council ALL97/99 randomised trial
Lancet Oncol
2010
, vol. 
11
 
5
(pg. 
429
-
438
)
45
Moorman
 
AV
Richards
 
SM
Robinson
 
HM
, et al. 
Prognosis of children with acute lymphoblastic leukemia (ALL) and intrachromosomal amplification of chromosome 21 (iAMP21)
Blood
2007
, vol. 
109
 
6
(pg. 
2327
-
2330
)
46
Mullighan
 
CG
Molecular genetics of B-precursor acute lymphoblastic leukemia
J Clin Invest
2012
, vol. 
122
 
10
(pg. 
3407
-
3415
)
47
Feng
 
J
Tang
 
Y
Prognostic significance of IKZF1 alteration status in pediatric B-lineage acute lymphoblastic leukemia: a meta-analysis
Leuk Lymphoma
2013
, vol. 
54
 
4
(pg. 
889
-
891
)
48
Mullighan
 
CG
Goorha
 
S
Radtke
 
I
, et al. 
Genome-wide analysis of genetic alterations in acute lymphoblastic leukaemia
Nature
2007
, vol. 
446
 
7137
(pg. 
758
-
764
)
49
Buitenkamp
 
TD
Pieters
 
R
Gallimore
 
NE
, et al. 
Outcome in children with Down's syndrome and acute lymphoblastic leukemia: role of IKZF1 deletions and CRLF2 aberrations
Leukemia
2012
, vol. 
26
 
10
(pg. 
2204
-
2211
)
50
Schrappe
 
M
Hunger
 
SP
Pui
 
CH
, et al. 
Outcomes after induction failure in childhood acute lymphoblastic leukemia
N Engl J Med
2012
, vol. 
366
 
15
(pg. 
1371
-
1381
)
51
Schultz
 
KR
Pullen
 
DJ
Sather
 
HN
, et al. 
Risk- and response-based classification of childhood B-precursor acute lymphoblastic leukemia: a combined analysis of prognostic markers from the Pediatric Oncology Group (POG) and Children's Cancer Group (CCG)
Blood
2007
, vol. 
109
 
3
(pg. 
926
-
935
)
52
Lauten
 
M
Moricke
 
A
Beier
 
R
, et al. 
Prediction of outcome by early bone marrow response in childhood acute lymphoblastic leukemia treated in the ALL-BFM 95 trial: differential effects in precursor B-cell and T-cell leukemia
Haematologica
2012
, vol. 
97
 
7
(pg. 
1048
-
1056
)
53
Stary
 
J
Zimmermann
 
M
Campbell
 
M
, et al. 
Intensive chemotherapy for childhood acute lymphoblastic leukemia: results of the randomized intercontinental trial ALL IC-BFM 2002
J Clin Oncol
2014
, vol. 
32
 
3
(pg. 
174
-
184
)
54
Borowitz
 
MJ
Devidas
 
M
Hunger
 
SP
, et al. 
Clinical significance of minimal residual disease in childhood acute lymphoblastic leukemia and its relationship to other prognostic factors: a Children's Oncology Group study
Blood
2008
, vol. 
111
 
12
(pg. 
5477
-
5485
)
55
Conter
 
V
Bartram
 
CR
Valsecchi
 
MG
, et al. 
Molecular response to treatment redefines all prognostic factors in children and adolescents with B-cell precursor acute lymphoblastic leukemia: results in 3184 patients of the AIEOP-BFM ALL 2000 study
Blood
2010
, vol. 
115
 
16
(pg. 
3206
-
3214
)
56
Vora
 
A
Goulden
 
N
Wade
 
R
, et al. 
Treatment reduction for children and young adults with low-risk acute lymphoblastic leukaemia defined by minimal residual disease (UKALL 2003): a randomised controlled trial
Lancet Oncol
2013
, vol. 
14
 
3
(pg. 
199
-
209
)
57
Escherich
 
G
Zimmermann
 
M
Janka-Schaub
 
G
Co ALLsg. Doxorubicin or daunorubicin given upfront in a therapeutic window are equally effective in children with newly diagnosed acute lymphoblastic leukemia. A randomized comparison in trial CoALL 07-03
Pediatr Blood Cancer
2013
, vol. 
60
 
2
(pg. 
254
-
257
)
58
Parker
 
C
Waters
 
R
Leighton
 
C
, et al. 
Effect of mitoxantrone on outcome of children with first relapse of acute lymphoblastic leukaemia (ALL R3): an open-label randomised trial
Lancet
2010
, vol. 
376
 
9757
(pg. 
2009
-
2017
)
59
Silverman
 
LB
Supko
 
JG
Stevenson
 
KE
, et al. 
Intravenous PEG-asparaginase during remission induction in children and adolescents with newly diagnosed acute lymphoblastic leukemia
Blood
2010
, vol. 
115
 
7
(pg. 
1351
-
1353
)
60
Teuffel
 
O
Kuster
 
SP
Hunger
 
SP
, et al. 
Dexamethasone versus prednisone for induction therapy in childhood acute lymphoblastic leukemia: a systematic review and meta-analysis
Leukemia
2011
, vol. 
25
 
8
(pg. 
1232
-
1238
)
61
Robinson
 
HM
Broadfield
 
ZJ
Cheung
 
KL
, et al. 
Amplification of AML1 in acute lymphoblastic leukemia is associated with a poor outcome
Leukemia
2003
, vol. 
17
 
11
(pg. 
2249
-
2250
)
62
Heerema
 
NA
Carroll
 
AJ
Devidas
 
M
, et al. 
Intrachromosomal amplification of chromosome 21 is associated with inferior outcomes in children with acute lymphoblastic leukemia treated in contemporary standard-risk children's oncology group studies: a report from the children's oncology group
J Clin Oncol
2013
, vol. 
31
 
27
(pg. 
3397
-
3402
)
63
Moorman
 
AV
Robinson
 
H
Schwab
 
C
, et al. 
Risk-directed treatment intensification significantly reduces the risk of relapse among children and adolescents with acute lymphoblastic leukemia and intrachromosomal amplification of chromosome 21: a comparison of the MRC ALL97/99 and UKALL2003 trials
J Clin Oncol
2013
, vol. 
31
 
27
(pg. 
3389
-
3396
)
64
Crist
 
WM
Carroll
 
AJ
Shuster
 
JJ
, et al. 
Poor prognosis of children with pre-B acute lymphoblastic leukemia is associated with the t(1;19)(q23;p13): a Pediatric Oncology Group study
Blood
1990
, vol. 
76
 
1
(pg. 
117
-
122
)
65
Uckun
 
FM
Sensel
 
MG
Sather
 
HN
, et al. 
Clinical significance of translocation t(1;19) in childhood acute lymphoblastic leukemia in the context of contemporary therapies: a report from the Children's Cancer Group
J Clin Oncol
1998
, vol. 
16
 
2
(pg. 
527
-
535
)
66
Andersen
 
MK
Autio
 
K
Barbany
 
G
, et al. 
Paediatric B-cell precursor acute lymphoblastic leukaemia with t(1;19)(q23;p13): clinical and cytogenetic characteristics of 47 cases from the Nordic countries treated according to NOPHO protocols
Br J Haematol
2011
, vol. 
155
 
2
(pg. 
235
-
243
)
67
Schultz
 
KR
Carroll
 
A
Heerema
 
NA
, et al. 
Long-term follow-up of imatinib in pediatric Philadelphia chromosome-positive acute lymphoblastic leukemia: Children's Oncology Group Study AALL0031
Leukemia
2014
, vol. 
28
 
7
(pg. 
1467
-
1471
)
68
Jeha
 
S
Coustan-Smith
 
E
Pei
 
D
, et al. 
Impact of tyrosine kinase inhibitors on minimal residual disease and outcome in childhood Philadelphia chromosome-positive acute lymphoblastic leukemia
Cancer
2014
, vol. 
120
 
10
(pg. 
1514
-
1519
)
69
Biondi
 
A
Schrappe
 
M
De Lorenzo
 
P
, et al. 
Imatinib after induction for treatment of children and adolescents with Philadelphia-chromosome-positive acute lymphoblastic leukaemia (EsPhALL): a randomised, open-label, intergroup study
Lancet Oncol
2012
, vol. 
13
 
9
(pg. 
936
-
945
)
70
Loh
 
M
A BCR-ABL1-like gene expression profile confers a poor prognosis in patients with high-risk acute lymphoblastic leukemia (HR-ALL): a report from Children's Oncology Group (COG) AALL0232 [abstract]
Blood (ASH Annual Meeting Abstracts)
2011
, vol. 
118
 
21
pg. 
743
 
71
Roberts
 
KG
Morin
 
RD
Zhang
 
J
, et al. 
Genetic alterations activating kinase and cytokine receptor signaling in high-risk acute lymphoblastic leukemia
Cancer Cell
2012
, vol. 
22
 
2
(pg. 
153
-
166
)
72
Weston
 
BW
Hayden
 
MA
Roberts
 
KG
, et al. 
Tyrosine kinase inhibitor therapy induces remission in a patient with refractory EBF1-PDGFRB-positive acute lymphoblastic leukemia
J Clin Oncol
2013
, vol. 
31
 
25
(pg. 
e413
-
416
)
73
Mustjoki
 
S
Hernesniemi
 
S
Rauhala
 
A
, et al. 
A novel dasatinib-sensitive RCSD1-ABL1 fusion transcript in chemotherapy-refractory adult pre-B lymphoblastic leukemia with t(1;9)(q24;q34)
Haematologica
2009
, vol. 
94
 
10
(pg. 
1469
-
1471
)
74
Lengline
 
E
Beldjord
 
K
Dombret
 
H
Soulier
 
J
Boissel
 
N
Clappier
 
E
Successful tyrosine kinase inhibitor therapy in a refractory B-cell precursor acute lymphoblastic leukemia with EBF1-PDGFRB fusion
Haematologica
2013
, vol. 
98
 
11
(pg. 
e146
-
148
)
75
Conter
 
V
Valsecchi
 
MG
Parasole
 
R
, et al. 
Childhood high-risk acute lymphoblastic leukemia in first remission: results after chemotherapy or transplant from the AIEOP ALL 2000 study
Blood
2014
, vol. 
123
 
10
(pg. 
1470
-
1478
)
76
Balduzzi
 
A
Valsecchi
 
MG
Uderzo
 
C
, et al. 
Chemotherapy versus allogeneic transplantation for very-high-risk childhood acute lymphoblastic leukaemia in first complete remission: comparison by genetic randomisation in an international prospective study
Lancet
2005
, vol. 
366
 
9486
(pg. 
635
-
642
)
77
Dreyer
 
ZE
Dinndorf
 
PA
Camitta
 
B
, et al. 
Analysis of the role of hematopoietic stem-cell transplantation in infants with acute lymphoblastic leukemia in first remission and MLL gene rearrangements: a report from the Children's Oncology Group
J Clin Oncol
2011
, vol. 
29
 
2
(pg. 
214
-
222
)
78
Blanco
 
E
Beyene
 
J
Maloney
 
AM
, et al. 
Non-relapse mortality in pediatric acute lymphoblastic leukemia: a systematic review and meta-analysis
Leuk Lymphoma
2012
, vol. 
53
 
5
(pg. 
878
-
885
)
79
Ethier
 
MC
Blanco
 
E
Lehrnbecher
 
T
Sung
 
L
Lack of clarity in the definition of treatment-related mortality: pediatric acute leukemia and adult acute promyelocytic leukemia as examples
Blood
2011
, vol. 
118
 
19
(pg. 
5080
-
5083
)
80
Lund
 
B
Asberg
 
A
Heyman
 
M
, et al. 
Risk factors for treatment related mortality in childhood acute lymphoblastic leukaemia
Pediatr Blood Cancer
2011
, vol. 
56
 
4
(pg. 
551
-
559
)
81
Prucker
 
C
Attarbaschi
 
A
Peters
 
C
, et al. 
Induction death and treatment-related mortality in first remission of children with acute lymphoblastic leukemia: a population-based analysis of the Austrian Berlin-Frankfurt-Munster study group
Leukemia
2009
, vol. 
23
 
7
(pg. 
1264
-
1269
)
82
Pichler
 
H
Reismuller
 
B
Steiner
 
M
, et al. 
The inferior prognosis of adolescents with acute lymphoblastic leukaemia (ALL) is caused by a higher rate of treatment-related mortality and not an increased relapse rate–a population-based analysis of 25 years of the Austrian ALL-BFM (Berlin-Frankfurt-Munster) Study Group
Br J Haematol
2013
, vol. 
161
 
4
(pg. 
556
-
565
)
83
Gafter-Gvili
 
A
Fraser
 
A
Paul
 
M
, et al. 
Antibiotic prophylaxis for bacterial infections in afebrile neutropenic patients following chemotherapy
Cochrane Database Syst Rev
2012
, vol. 
1
 pg. 
CD004386
 
84
Baden
 
LR
Bensinger
 
W
Angarone
 
M
, et al. 
Prevention and treatment of cancer-related infections
J Natl Compr Canc Netw
2012
, vol. 
10
 
11
(pg. 
1412
-
1445
)
85
Flowers
 
CR
Seidenfeld
 
J
Bow
 
EJ
, et al. 
Antimicrobial prophylaxis and outpatient management of fever and neutropenia in adults treated for malignancy: American Society of Clinical Oncology clinical practice guideline
J Clin Oncol
2013
, vol. 
31
 
6
(pg. 
794
-
810
)
86
Hunger
 
SP
Loh
 
ML
Whitlock
 
JA
, et al. 
Children's Oncology Group's 2013 blueprint for research: acute lymphoblastic leukemia
Pediatr Blood Cancer
2013
, vol. 
60
 
6
(pg. 
957
-
963
)
87
Nachman
 
JB
Sather
 
HN
Sensel
 
MG
, et al. 
Augmented post-induction therapy for children with high-risk acute lymphoblastic leukemia and a slow response to initial therapy
N Engl J Med
1998
, vol. 
338
 
23
(pg. 
1663
-
1671
)
88
Seibel
 
NL
Steinherz
 
PG
Sather
 
HN
, et al. 
Early postinduction intensification therapy improves survival for children and adolescents with high-risk acute lymphoblastic leukemia: a report from the Children's Oncology Group
Blood
2008
, vol. 
111
 
5
(pg. 
2548
-
2555
)
89
Tower
 
RL
Jones
 
TL
Camitta
 
BM
, et al. 
Dose intensification of methotrexate and cytarabine during intensified continuation chemotherapy for high-risk B-precursor acute lymphoblastic leukemia: POG 9406: A report from the Children's Oncology Group
J Pediatr Hematol Oncol
2014
, vol. 
36
 
5
(pg. 
353
-
361
)
90
Larsen
 
EC
Salzer
 
WL
Devidas
 
M
, et al. 
Comparison of high-dose methotrexate (HD-MTX) with Capizzi methotrexate plus asparaginase (C-MTX/ASNase) in children and young adults with high-risk acute lymphoblastic leukemia (HR-ALL): A report from the Children's Oncology Group Study AALL0232 [abstract]
J Clin Oncol
2011
, vol. 
29
 pg. 
3
 
91
Vora
 
A
Goulden
 
N
Mitchell
 
C
, et al. 
Augmented post-remission therapy for a minimal residual disease-defined high-risk subgroup of children and young people with clinical standard-risk and intermediate-risk acute lymphoblastic leukaemia (UKALL 2003): a randomised controlled trial
Lancet Oncol
2014
, vol. 
15
 
8
(pg. 
809
-
818
)