Abstract
Approximately 90% of the 2,000 children, adolescents, and young adults enrolled each year in Children’s Oncology Group acute lymphoblastic leukemia (ALL) trials will be cured. However, high-risk subsets with significantly inferior survival remain, including infants, newly diagnosed patients with age ≥10 years, white blood count ≥50,000/µl, poor early response or T-cell ALL, and relapsed ALL patients. Effective strategies to improve survival include better risk stratification, optimizing standard chemotherapy and combining targeted therapies with cytotoxic chemotherapy, the latter of which is dependent upon identification of key driver mutations present in ALL.
Keywords: acute lymphoblastic leukemia (ALL), COG ALL trials
INTRODUCTION
The goals of this Blueprint are to summarize current knowledge regarding the biology and treatment of childhood acute lymphoblastic leukemia (ALL), to review the major findings from recent Children’s Oncology Group (COG) ALL trials and translational research efforts, and to outline plans for COG ALL clinical trials scheduled to open in the near future. The efforts of groups throughout the world have made childhood ALL a highly curable disease, but a review of the results of non-COG ALL trials is beyond the scope of this report.
STATE OF THE DISEASE—CLINICAL
Overview and Incidence
ALL is the most common pediatric malignancy, comprising 25% of cancers occurring before 15 years of age and 19% among those less than 20 years old [1]. Despite steady improvements in outcome, ALL remains the leading cause of childhood cancer death [2].
Staging/Stratification
Clinical trials have identified a number of features predictive of outcome, including initial age and white blood cell count (WBC), immunophenotype, presence/absence of central nervous system (CNS) or testicular leukemia, presence/absence of specific sentinel genetic lesions (good risk: ETV6–RUNX1 fusion or hyperdiploidy with favorable chromosome trisomies; poor risk: MLL-rearrangements (MLL-R), hypodiploidy, intrachromosomal amplification of chromosome 21 (iAMP21), Philadelphia chromosome positive (Ph+) ALL), and early treatment response, which is assessed now by most groups via measurement of minimal residual disease (MRD) at specific timepoints early in therapy. The COG has developed a system that integrates these factors, including National Cancer Institute (NCI) risk group [3] (standard risk (SR): age 1–9.99 years and initial WBC <50,000/µl; high risk (HR): age 10+ years and/or initial WBC ≥50,000/µl), for risk stratification and treatment allocation [4–6]. The current risk stratification system (AALL08B1; Table I) builds on that developed for the first generation COG ALL trials (AALL03B1) with key differences being changing the MRD threshold used to define poor response from ≥0.1% (AALL03B1) to ≥0.01% (AALL08B1) at end Induction, and incorporating Day 8 peripheral blood MRD measurements while eliminating Day 8/15 bone marrow (BM) aspirates for morphology. Most international groups assess MRD via molecular methods, but the COG has utilized flow-based MRD in clinical trials and has the largest experience with this modality with over 12,000 patients allocated to different treatments based on flow MRD response.
TABLE I.
Overview of the AALL08B1 Classification System for B-Precursor ALL
| Risk group | Low | Averagea | Highb | Very high | ||||||
|---|---|---|---|---|---|---|---|---|---|---|
| Projected 5-year EFS | >95% | 90–95% | 88–90% | <80% | ||||||
| NCI risk-group | SR | SR | SR | SR | SR | HR (<13 years) | SR | HR | HR (≥13 years) | SR or HR |
| Favorable geneticsc | Yes | Yes | No | Yes | No | Any | No | Any | Any | Any |
| Unfavorable characteristicsd | No | No | No | No | No | No | No | No | No | Yes |
| Day 8 PB MRD | <0.01% | ≥0.01% | <1% | Any | ≥1% | Any | Any | Any | Any | Any |
| Day 29 BM MRD | <0.01% | < 0.01% | <0.01% | ≥0.01% | <0.01% | <0.01% | ≥0.01% | ≥0.01% | <0.01% | Any |
| Accrual/year | 250 | 350 | 266 | 70 | 89 | 264 | 79 | 64 | 154 | 116 |
| Total accrual/year | 250 | 616 | 423 | 413 | ||||||
| Fraction of patients | 14.7% | 36.2% | 24.8% | 24.3% | ||||||
SR, standard risk; HR, high risk; PB, peripheral blood; MRD, minimal residual disease; BM, bone marrow.
NCI SR patients who are CNS2 may be included in average risk, but will not be eligible for the low risk Arm.
All patients with testicular leukemia will be assigned as HR at start Induction, but may change to VHR if Day 29 MRD ≥0.01% or unfavorable characteristics are present.
“Yes” is defined as the presence of double trisomy 4 and 10 OR ETV6–RUNX1 fusion.
Consists of patients with CNS3, hypodiploidy (<44 chromosomes and/or DNA index <0.81), iAMP21, Induction failure (M3 marrow Day 29), or MLL rearrangement (not MLL deletion). BCR–ABL1 positive patients are eligible for a separate Ph+ ALL study.
Current Outcome
The 5-year overall survival (OS) for ALL increased steadily from 10% in the 1960s to about 80% in the late 1980s [7–14]. The COG recently reported analyses of 21,626 patients enrolled in ALL trials between 1990 and 2005 and showed that the 5-year OS increased from 83.7% in 1990–1994 to 90.4% in 2000–2005 (P < 0.0001) [15]. Current data (May 2012) show that the 5-year OS is 92.3% for 6,662 patients enrolled in COG ALL trials between 2006 and 2009, which is significantly (P = 0.0015) better than OS in the 2000–2005 cohort.
Survival for patients with relapsed ALL remains poor. Prognostic factors for survival post relapse were analyzed in 1961 (of 9,585 total) patients enrolled on COG trials between 1988 and 2002 who relapsed at any site [16]. As previously reported, the major predictors of outcome were time to relapse, site of relapse, and immunophenotype [16–20]. Patients with early BM relapse within 36 months of diagnosis had an abysmal outcome with 5-year OS of about 15% [16].
STATE OF THE DISEASE—BIOLOGICAL
Molecular Targets
Recent studies by the COG and other groups have provided major insights into the genomic landscape of newly diagnosed and relapsed ALL. While a complete description is beyond the scope of this report, these include identification and characterization of the Philadelphia chromosome-like (Ph-like) subtype, discovery of CRLF2 genomic lesions and JAK mutations, development of molecular risk classifiers, and the identification of key pathways targeted by somatic sequence mutations [21–46]. Large scale whole exome and whole genome sequencing studies currently being performed by the COG and other groups will provide data that should further refine knowledge of the genomic landscape of pediatric ALL and identify new potential biomarkers and therapeutic targets. In parallel, the COG and other groups have analyzed the relationship between host germline variability and ALL incidence, biology, treatment response, and adverse side effects using single nucleotide polymorphism (SNP) arrays [47–54]. The COG has the largest experience in this area with SNP germline genotypes completed for 2,000 patients enrolled in the COG P9900 studies, 2,653 patients enrolled in AALL0232 (86.8% of eligible patients), and an additional 2,000–3,000 genotypes that will be completed in the next few years.
A key finding with major therapeutic implications is the discovery of a subtype called Ph-like (or BCR–ABL1-like) ALL that has a gene expression profile similar to that of Ph+ ALL, but lacks BCR–ABL1 fusion [27,29]. Ph-like ALL is three to four times more common than Ph+ ALL, and Ph-like ALL patients have a significantly worse 5-year event free survival (EFS) than other HR patients without this phenotype [55]. About 40–50% of Ph-like ALL cases have genomic alterations leading to CRLF2 over-expression, about half of which have JAK1 or JAK2 mutations that are responsive to JAK inhibitors in vitro and in xenograft models [22,25,28,32,41,42,56,57]. The COG is conducting a phase I trial (ADVL1011) of the JAK inhibitor ruxolitinib, and plans to develop a trial of ruxolitinib combined with chemotherapy in relapsed ALL patients with JAK mutations. COG studies have shown that other kinase gene point mutations occur only rarely in Ph-like ALL [55], but that many Ph-like ALL cases contain fusion genes activating ABL1, JAK2, and PDGFRB that are sensitive to cognate tyrosine kinase inhibitors (TKIs) in vitro, in xenograft models, and anecdotally in pediatric ALL patients with lesions such as EBF1-PDGRFB fusion [41,42,58]. Based on these findings, the COG is developing technologies to identify patients enrolled in with such fusions with the goal of treating patients with ABL1 and PDGFRB fusions with ABL/PDGFRB class TKIs.
There are several other molecular targets with therapeutic potential that the COG has or will study. AALL0631 is testing whether the FLT3 TKI lestaurtinib can improve outcome of infants with MLL-R ALL. AALL1122 is the third COG trial of TKI (dasatinib) + chemotherapy in Ph+ ALL. Monoclonal antibodies and immunoconjugates are also attractive agents to test in ALL. The COG ADVL04P2 phase II study of epratuzumab + chemotherapy in relapsed ALL did not pass the pre-defined screening benchmark for activity; however, improvements in early MRD response were observed. The bispecific T cell engager agent blinatumomab is an antibody construct that directs CD3+ cytotoxic T-cells to CD19 expressing leukemic blasts. Very promising responses have been seen with this agent in relapsed adult ALL and the COG is currently collaborating with international colleagues to conduct a phase I/II trial (AALL1121) of blinatumomab in children with second or greater relapsed or refractory ALL, with plans to test this agent in first relapse of ALL in the near future.
MAJOR RESULTS FROM RECENT COG ALL TRIALS
Prior to the development of TKI therapy, Ph+ ALL was one of the worst prognostic groups in pediatric ALL. Despite treatment with intensive chemotherapy and widespread use of allogeneic stem cell transplantation (SCT) less than half of patients survived [59,60]. COG AALL0031 incorporated imatinib, starting after completion of Induction therapy, into a very intensive chemotherapy regimen [61]. The chemotherapy + imatinib regimen was well tolerated, with no significant increased toxicity attributable to imatinib. Patients treated with continuous imatinib (340 mg/m2/day) therapy had a 3-year EFS of 80% versus 35% (P < 0.0001) for historical controls treated in the pre-TKI era. COG AALL0622 then tested dasatinib (60 mg/m2/day) + the AALL0031 chemotherapy backbone. Enrollment to this study was completed in February 2012, so mature outcome data are not yet available. Continuous dasatinib treatment (starting on Day 15 of Induction) combined with intensive chemotherapy was determined to be safe based on pre-defined criteria. In AALL0031 continuous imatinib treatment during the last year of maintenance therapy was associated with unacceptable elevation in liver transaminases, so imatinib was given on a 2-week on to 2-week off schedule during this phase [61]. Longer follow-up of AALL0622 is needed to see if continuous dasatinib treatment will be feasible throughout maintenance. AALL0622 patients received dasatinib for the last 2 weeks of Induction and had significantly higher complete remission rates and a much lower proportion of patients were MRD-positive as compared to AALL0031 where TKI was started after Induction [62]. To address the challenge of testing targeted therapies in small, molecularly defined childhood ALL subsets, the COG collaborated with the European EsPhALL group to develop a transatlantic pediatric Ph+ ALL trial (AALL1122) of dasatinib + chemotherapy that is now enrolling patients. While the optimal therapy for Ph+ ALL remains to be determined, the results of COG AALL0031 have significant implications for other subsets of very high risk ALL and suggest that outcomes can be improved significantly if: (a) a driver molecular lesion is present that is fundamental to leukemogenesis; (b) the leukemia is dependent on continued activity of this molecular lesion (oncogene addiction); and (c) an effective targeted agent is available that can be combined safely with chemotherapy [6]. Recent discoveries of targetable genomic lesions in Ph-like ALL exemplify this approach.
Historically, outcomes for T-cell ALL (T-ALL) have been worse than those for B cell precursor (BCP) ALL [15]. Nelarabine has differential cytotoxicity for T-cell malignancies and had very promising results in phase I/II trials of children and adults with refractory T-ALL or T-cell lymphoma [63,64]. However, these early phase trials suffered from high rates of serious and sometimes fatal CNS toxicity (Guillain–Barre syndrome, refractory seizures, and coma). COG AALL00P2 established that adding nelarabine to intensive chemotherapy was safe and feasible, with very promising outcomes in children with newly diagnosed T-ALL and high-risk features [65]. This study formed the basis for COG AALL0434, a prospective randomized T-ALL trial of chemotherapy ± nelarabine that should complete enrollment in 2014.
The COG has taken a different approach than many other groups to treating SR ALL, which comprises about two-thirds of BCP ALL patients. COG trials use a 3-drug (dexamethasone (DEX), PEG-asparaginase (ASNase), and vincristine) Induction without anthracyclines, a 4-week low intensity consolidation phase rather than the intensive 8-week Berlin-Frankfurt-Munster (BFM) or COG augmented BFM (ABFM) consolidation, and no high dose methotrexate (HD MTX). CCG 1991 established that escalating IV MTX without leucovorin rescue was superior to oral MTX during two interim maintenance (IM) phases with almost all of the improvement due to decreased CNS/testicular rather than BM relapses [66]. CCG 1961 showed that a second delayed intensification (DI) phase was not beneficial in HR patients with a rapid response [67] and CCG 1991 had the same finding for SR BCP ALL patients. This more limited treatment approach was highly effective for the ~90% of CCG 1991 SR BCP ALL patients that were good responders based on BM morphology during Induction. The 5-year OS for 2,078 rapid responders was 96.0%; for the ~50% of patients with favorable genetic features (ETV6–RUNX1 fusion or trisomy 4 + 10), the 5-year EFS and OS were 94.2% and 98.7%, suggesting that a large group of patients can be identified that are almost certain to be cured with relatively limited therapy that includes only 75 mg/m2 of cumulative anthracycline exposure and 1 g/m2 of cyclophosphamide. This baseline regimen was used in COG AALL0331 (2005–2010) that tested augmented versus standard consolidation (results remain blinded as of September 2012) and the current COG AALL0932 SR-ALL trial.
The COG AALL0232 study was a 2 × 2 randomized trial that enrolled 3,108 eligible HR BCP patients (2004–2011). All patients received the COG ABFM regimen, which was shown to be superior to the prior standard regimen in CCG 1882 and 1961 [67–69]. Patients were randomized to receive DEX 10 mg/m2/day on Days 1–14 (Dex14) versus prednisone 60 mg/m2/day on Days 1–28 (Pred28) during Induction, and HD MTX with leucovorin rescue versus escalating dose Capizzi MTX without rescue + ASNase during the first IM phase. Patients randomized to HD MTX had superior outcomes to those randomized to Capizzi MTX [70]. The Induction steroid question initially included all patients, but was restricted to those <10 years old in 2008 after interim monitoring showed excess rates of osteonecrosis (ON) among patients 10+ years old randomized to Dex14 [71]. For the patients <10 years, the DH (Dex14 + HD MTX) regimen had superior EFS compared to PH (Pred28 + HD MTX) therapy [72]. In contrast, for patients 10+ years old at diagnosis there was no difference between those randomized to Dex14 versus Pred28. Osteonecrosis rates were much higher in those 10+ years old compared to younger patients, and modest changes in chemotherapy administration significantly impacted ON risk, with interrupted dose DEX (Days 1–7 and 15–21 vs. 1–21) during DI significantly decreasing ON risk. These results were practice changing; HD MTX and age-based Induction steroid regimens are the new standards of care in COG HR BCP ALL trials.
The COG and CALGB collaborated to compare outcomes for AYA ALL patients 16–21 years old treated on pediatric versus adult cooperative group trials. The AYA patients treated on pediatric trials had dramatically better survival than those treated on the adult trials [73]. Many other groups have reported similar findings [74]. The COG later reported outcome for 262 AYA ALL patients enrolled on CCG 1961 (the largest cohort reported to date) showing 5-year EFS 71.5% and OS 77.5%, establishing that there is no role for routine use of SCT in AYA ALL [75]. Data were recently presented regarding 601 AYA patients 16–30 years old treated on AALL0232, by far the largest group analyzed to date, and showed a significantly increased risk of death in remission for AYA patients as compared to younger HR children in this trial [76,77]. Osteonecrosis is a particular problem among patients ≥13 years. CCG 1961 established that interrupted dose DEX during DI significantly decreased ON incidence [78]. Parallel genomic studies showed that genetic polymorphisms affect ON risk [47]. To further study risk factors for ON, the current COG HR ALL trial AALL1131 includes pharmacokinetic and genomic studies in parallel with MRI screening.
Despite the use of very intensive therapy, the outcome for infants younger than 1 year with ALL is substantially inferior to that of older children with EFS rates less than 50%, and even lower for the approximately 80% with MLL-R [79]. Given these poor outcomes despite highly toxic therapy, COG AALL0631 (2007-present) is testing targeted therapy. Almost all infants with MLL-R ALL overexpress the wild type FLT3 TK with evidence of autocrine signaling [80–82]. Pre-clinical studies showed that lestaurtinib, a small molecule FLT3 TKI with excellent oral bioavailability, selectively kills MLL-R ALL samples and synergizes with chemotherapy [83,84]. The initial phase of COG AALL0631 has been completed and established that lestaurtinib can be given safely with intensive chemotherapy and has biological activity [85,86]. AALL0631 is now in the efficacy phase with MLL-R infants randomized to receive chemotherapy with/without lestaurtinib.
Patients with an early BM relapse occurring within 36 months of diagnosis have a very poor outcome and few survive despite widespread use of SCT [16]. COG investigators hypothesized that it might be possible to increase complete remission (CR) rates and convert more patients to MRD-negative status thereby improving SCT outcomes by adding new agents to an intensive chemotherapy regimen. COG AALL01P2 established the safety of a 3–4 months triple induction regimen consisting of three 4–5 weeks blocks of intensive chemotherapy [87]. COG ADVL04P2 then tested addition of the anti-CD22 monoclonal antibody epratuzumab to this regimen and showed that epratuzumab could be given safely once or twice weekly during the 4-week Induction, but there was no change in CR2 rates with chemotherapy + epratuzumab as compared to historical controls treated with the chemotherapy regimen alone [88,89]. Based on these results, the COG will not pursue epratuzumab further. COG AALL07P1 is now testing addition of bortezomib to the AALL01P2 chemotherapy regimen.
STRATEGIC APPROACH: TARGETED THERAPY
Newly Diagnosed Population
The second generation COG BCP ALL trials (AALL0932/AALL1131) for Ph-negative ALL use a classification and stratification approach (AALL08B1; Table I) whereby SR ALL patients enter AALL0932 and receive a three-drug DEX-based Induction. At end Induction, clinical, genetic, and MRD response data are integrated to assign patients to different studies for post-Induction therapy. AALL0932 patients with adverse genetic features (hypodiploidy, iAMP21, MLL-R) or poor response are not eligible to continue on AALL0932, but are eligible to cross over to receive COG ABFM post-Induction therapies with randomized interventions on AALL1131. This approach assures that only patients with an excellent survival (baseline expected 5-year EFS 93%, OS ≥97%) will be treated with less intensive therapy. In this population, randomized questions are being conducted that focus on maintenance chemotherapy, and embedded studies are assessing health-related quality of life, and physical functioning.
At diagnosis, HR BCP patients enroll in AALL1131 and receive a four-drug Dex14 (1–9.99 years) or Pred28 (10–30.99 years) Induction. At the end of Induction, patients are stratified into HR and very high risk (VHR) subgroups (and AALL0932 HR/VHR patients join these groups) with different randomized interventions on a similar ABFM backbone that utilizes HD MTX during IM#1 for all patients, a single DI phase, and a second IM phase with Capizzi MTX + ASNase (VHR only). The HR group has an estimated 88% baseline 5-year EFS and is randomized to receive intrathecal (IT) MTX versus IT triple chemotherapy. The VHR group includes patients with older age (≥13 years), adverse genetics (hypodiploidy, iAMP21, MLL-R), or Day 29 MRD ≥0.01% and has an estimated 5-year EFS of ≤75%. Novel therapies are being explored in this group by comparing standard ABFM consolidation/re-consolidation strategies with two different experimental regimens that utilize more myelosuppressive agents commonly used in relapsed ALL, etoposide/cyclophosphamide ± clofarabine. The 3-drug etoposide/cyclophosphamide/clofarabine regimen has shown significant activity in refractory/relapsed ALL [90–92]. The design of this 3-arm trial will allow the effect of clofarabine to be isolated. Embedded studies assess neurocognitive outcomes using a novel computer-based assessment tool and will define the natural history of ON using serial magnetic resonance imaging.
Because the outcome for T-ALL patients is inferior to that of BCP ALL patients when less intensive treatment is administered [15], all patients with T-ALL are considered high risk and treated with a 4-drug Induction and the COG ABFM backbone. AALL0434 employs a 2 × 2 design with one intervention testing refinement of standard agents (HD MTX vs. Capizzi MTX + ASNase) and one testing nelarabine.
Relapsed Population
COG AALL07P1 is testing addition of bortezomib to the AALL01P2 triple re-Induction regimen in patients with early BM relapse occurring within 36 months of diagnosis. AALL0433 is designed to see if outcome for patients with late (≥36 months) BCP marrow and isolated extramedullary (IEM) (≥18 months) relapse, can be improved by using this same triple re-Induction regimen (without bortezomib) followed by 2 years of intensive chemotherapy.
Trial Design Strategies
Because of the size of patient populations, the COG is able to conduct randomized phase III trials in patients with newly diagnosed ALL, often asking two questions with a 2 × 2 design. For some small subsets randomizations with relaxed alpha (infant trial AALL0631), or comparison to historical control outcomes (Ph+ ALL trials AALL0031/AALL0622/AALL1122) have been used. Recent COG ALL trials for first relapse of ALL have used historical control comparisons in an effort to evaluate new agents more efficiently, but a number of confounding factors such as changes in patient populations or supportive care over time have limited the effectiveness of this approach. Because of this, newly developed COG relapse ALL trials will include concurrent randomizations to control versus experimental arms with novel agents.
KEY TRIALS TO BE PURSUED
Pivotal Phase III Trials
Early relapse is a common cause of treatment failure in T-ALL/T-lymphoblastic lymphoma (T-LL), and patients who relapse rarely survive. These factors suggest that early treatment intensification and early addition of active new agents are needed to improve survival. Pre-clinical data show that bortezomib is active in T-ALL and that it has an additive or synergistic effect with cytotoxic chemotherapy [93–95]. Bortezomib has an acceptable toxicity profile in children, and the COG phase I trial ADVL0317 showed a maximum tolerated dose of 1.3 mg/m2 given twice weekly for 2 weeks as a single agent, followed by a 10-day rest [96]. Bortezomib has been incorporated safely into an intensive ALL re-Induction backbone [97,98] and is currently under investigation in ALL patients with early first relapse (AALL07P1). Current plans for the next COG T-ALL/T-LL trial (AALL1231), projected to open when AALL0434 completes accrual in early 2014, are to use a modified COG ABFM regimen (similar to AALL1131) that includes two IM (#1 HD MTX; #2 Capizzi MTX ± ASNase) phases and one DI phase. Based on very promising data regarding DEX efficacy in T-ALL from the AIEOP-BFM 2000 [99] and UKALL 2003 [100] trials, all patients will receive DEX 6 mg/m2/day for 28 days during Induction and two doses of PEG-ASNase given on Days 4 and 18, with a randomization to ± bortezomib 1.3 mg/m2 with four doses in Induction and four in DI, using 4-year EFS as the primary endpoint.
To date, the COG has conducted separate trials for the three major risk groups of first relapse ALL (early BM, early IEM, late BM + late IEM). For early BM relapse a series of single arm, limited accrual/duration phase II re-Induction studies have been conducted testing novel agents (ADVL04P2, epratuzumab; AALL07P1, bortezomib) added to the AALL01P2 chemotherapy regimen with the primary endpoint being CR2 rate at end Block 1 as compared to historical control data with chemotherapy alone. COG first relapse trials for intermediate (early IEM) and low (late BM + late IEM) risk patients have each used unique multiagent chemotherapy regimens with or without randomized interventions with 3-year EFS endpoints. To date, MRD response has not been used to allocate subsets of late BM relapse patients to receive SCT, which is becoming a standard approach. This fragmented approach to relapse trials has been associated with poor accrual that has led to problems in completing studies efficiently. The challenges of executing trials in patients with relapsed LL have been even more problematic. To address these problems and test new agents efficiently in these very high risk populations, COG is developing a comprehensive trial that will include all patients with first relapse of ALL/LL and which will adopt a platform, commonly used in European studies. This approach has been successful in other cooperative groups, including the I-BFM and UK CCLG/Australia/New Zealand consortia [19,101].
Though the design of this comprehensive relapse trial is not yet finalized, it is envisioned that late BCP ALL BM and IEM relapse patients will receive three blocks of a standard chemotherapy backbone that is a slightly modified ALLR3 regimen [101]. These patients will be defined as low (LR; MRD <0.1% end Block 1) and intermediate risk (IR; MRD ≥0.1% end Block 1) based on early MRD response. The IR patients will not continue to receive treatment on this study after Block 3, but will be eligible for SCT trials developed by the COG SCT and ALL committees. The LR patients will continue on study and receive 2 years total treatment with a post Block 3 randomization to ± - blinatumomab replacing 16 weeks of conventional chemotherapy. The approach for high-risk patients with early BM relapse of BCP ALL or any BM relapse of T-ALL/LL has not yet been finalized.
Prioritization Strategy
While overall outcomes are excellent for ALL, it remains the single largest cause of cancer related death in children [2]. Even though good risk subsets of newly diagnosed ALL patients now have 5-year OS approaching 95%, 38% of deaths that occurred in patients diagnosed between 1990 and 2005 were in those with good risk clinical features [15]. Thus, it is imperative to conduct randomized trials testing interventions seeking to improve EFS for subsets with less than 95% 5-year EFS. Identification of patients with expected EFS of at least 95% is now possible at end Induction, so it is likely that future COG ALL trials will enroll all patients with ALL at initial diagnosis, with ultralow risk subsets identified at end Induction that will not participate in randomized questions. The current approach is to ask questions focused on refining the use of standard agents in subsets with expected EFS above about 85%, and to test new agents and targeted therapies in subgroups with a worse outcome. As new agents that are effective and have favorable toxicity profiles are identified, it will be important to test them in good risk subsets to see if they can improve outcome for these patients and/or replace cytotoxic chemotherapy agents associated with adverse short/long term toxicities.
National and International Collaborations in Childhood ALL
As the number of relapses decreases for childhood ALL, national and international collaborations are increasingly required to further improve survival. The major North American (including COG, the Dana Farber Cancer Institute consortium, and St. Jude Children’s Research Hospital), European and Asian childhood ALL treatment groups form the Ponte di Legno Childhood ALL collaborative network and leaders meet periodically to discuss trial results, trial design strategies, and to plan collaborative research projects on rare patient subsets [102]. These collaborations have provided important insights into high-risk subsets with recent major publications on hypodiploid ALL, Ph+ ALL, Induction failure, and near final manuscripts on Down syndrome-ALL and second malignant neoplasms in patients treated for ALL [60,103,104]. Because of these ongoing interactions, the groups have become comfortable sharing unpublished data to facilitate design of new trials by other groups. Critically, international collaboration will be required to test new targeted therapies in relatively uncommon (<5–10% of childhood ALL), molecularly defined patients subsets that have a poor outcome, as exemplified by the current international Ph+ ALL trial (COG AALL1122).
CONCLUSIONS
Cure rates for pediatric ALL are outstanding, but ALL remains the leading cause of childhood cancer death. Optimization of conventional chemotherapy has improved outcome for favorable risk subgroups, but has had limited effect for poor risk subgroups of newly diagnosed and relapsed ALL. Targeted therapy has revolutionized treatment of Ph+ ALL and provides a roadmap for how to approach other poor risk subgroups. Current and future COG ALL trials will focus on using existing and newly discovered biomarkers to stratify patients into different risk groups and testing novel agents and treatment strategies for those predicted to have a poor outcome. With decreasing relapse rates and improving survival, enhancements in supportive care to decrease morbidity and mortality and better measures of short- and long-term quality of life become even more important. Parallel translational research efforts have led to major new insights into ALL biology, host polymorphisms associated with ALL occurrence, genotype, response, and toxicities, and identified new biomarkers and potential therapeutic targets. Emerging data from large-scale next generation sequencing studies should identify additional therapeutic targets to pursue in COG ALL clinical trials.
ACKNOWLEDGMENT
This work was supported by the Chair’s grant U10 CA98543, Statistics and Data Center grant U10 CA98413, CCOP U10 CA95861, Human Specimen Banking U24 CA114766 grants of the Children’s Oncology group from the National Cancer Institute, National Institutes of Health, Bethesda, MD, USA. A complete listing of grant support for research conducted by the Children’s Cancer Group and Pediatric Oncology Group before initiation of the COG grant in 2003 is available online at: http://www.childrensoncologygroup.org/admin/grantinfo.htm. Stephen Hunger is the Ergen Family Chair in Pediatric Cancer.
Grant sponsors: National Cancer Institute; National Institutes of Health, Bethesda, MD.
Footnotes
Conflicts of interest: Mignon Loh is a member of the DSMC for a Bristol Myers Squibb sponsored trail of dasatinib in Ph+ leukemias. Stephen Hunger owns stock in Amgen, which markets blinatumomab.
REFERENCES
- 1.Ries LAG, Smith MA, Gurney JG, et al., editors. National Cancer Institute, SEER Program. Bethesda, MD: 1999. Cancer Incidence and Survival among Children and Adolescents: United States SEER Program 1975–1995. NIH Pub. No. 99-4649. [Google Scholar]
- 2.Smith MA, Seibel NL, Altekruse SF, et al. Outcomes for children and adolescents with cancer: Challenges for the twenty-first century. J Clin Oncol. 2010;28:2625–2634. doi: 10.1200/JCO.2009.27.0421. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.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;14:18–24. doi: 10.1200/JCO.1996.14.1.18. [DOI] [PubMed] [Google Scholar]
- 4.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;109:926–935. doi: 10.1182/blood-2006-01-024729. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.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;111:5477–5485. doi: 10.1182/blood-2008-01-132837. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Hunger SP, Raetz EA, Loh ML, et al. Improving outcomes for high-risk all: Translating new discoveries into clinical care. Pediatr Blood Cancer. 2011;56:984–993. doi: 10.1002/pbc.22996. [DOI] [PubMed] [Google Scholar]
- 7.Conter V, Arico M, Basso G, et al. Long-term results of the Italian Association of Pediatric Hematology and Oncology (AIEOP) studies 82, 87, 88, 91 and 95 for childhood acute lymphoblastic leukemia. Leukemia. 2010;24:255–264. doi: 10.1038/leu.2009.250. [DOI] [PubMed] [Google Scholar]
- 8.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;24:285–297. doi: 10.1038/leu.2009.262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Kamps WA, van der Pal-de Bruin KM, Veerman AJ, et al. Long-term results of Dutch Childhood Oncology Group studies for children with acute lymphoblastic leukemia from 1984 to 2004. Leukemia. 2010;24:309–319. doi: 10.1038/leu.2009.258. [DOI] [PubMed] [Google Scholar]
- 10.Mitchell C, Richards S, Harrison CJ, et al. Long-term follow-up of the United Kingdom Medical Research Council protocols for childhood acute lymphoblastic leukaemia, 1980–2001. Leukemia. 2010;24:416–418. doi: 10.1038/leu.2009.256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.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;24:265–284. doi: 10.1038/leu.2009.257. [DOI] [PubMed] [Google Scholar]
- 12.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;24:371–382. doi: 10.1038/leu.2009.252. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.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;24:355–370. doi: 10.1038/leu.2009.261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.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;24:320–334. doi: 10.1038/leu.2009.253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Hunger SP, Lu X, Devidas M, et al. Improved survival for children and adolescents from 1990 to 2005: A report from the Children’s Oncology Group. J Clin Oncol. 2012;30:1663–1669. doi: 10.1200/JCO.2011.37.8018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Nguyen K, Devidas M, Cheng SC, et al. Factors influencing survival after relapse from acute lymphoblastic leukemia: A Children’s Oncology Group study. Leukemia. 2008;22:2142–2150. doi: 10.1038/leu.2008.251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Einsiedel HG, von Stackelberg A, Hartmann R, et al. Long-term outcome in children with relapsed all by risk-stratified salvage therapy: Results of trial acute lymphoblastic leukemia-relapse study of the Berlin-Frankfurt-Munster group 87. J Clin Oncol. 2005;23:7942–7950. doi: 10.1200/JCO.2005.01.1031. [DOI] [PubMed] [Google Scholar]
- 18.Roy A, Cargill A, Love S, et al. Outcome after first relapse in childhood acute lymphoblastic leukaemia—Lessons from the United Kingdom R2 Trial. Br J Haematol. 2005;130:67–75. doi: 10.1111/j.1365-2141.2005.05572.x. [DOI] [PubMed] [Google Scholar]
- 19.Tallen G, Ratei R, Mann G, et al. Long-term outcome in children with relapsed acute lymphoblastic leukemia after time-point and site-of-relapse stratification and intensified short-course multidrug chemotherapy: Results of trial ALL-REZ BFM 90. J Clin Oncol. 2010;28:2339–2347. doi: 10.1200/JCO.2009.25.1983. [DOI] [PubMed] [Google Scholar]
- 20.van den Berg H, de Groot-Kruseman HA, Damen-Korbijn CM, et al. Outcome after first relapse in children with acute lymphoblastic leukemia: A report based on the Dutch Childhood Oncology Group (DCOG) relapse ALL 98 protocol. Pediatr Blood Cancer. 2011;57:210–216. doi: 10.1002/pbc.22946. [DOI] [PubMed] [Google Scholar]
- 21.Bhojwani D, Kang H, Moskowitz NP, et al. Biologic pathways associated with relapse in childhood acute lymphoblastic leukemia: A Children’s Oncology Group study. Blood. 2006;108:711–717. doi: 10.1182/blood-2006-02-002824. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Harvey RC, Mullighan CG, Chen IM, et al. Rearrangement of CRLF2 is associated with mutation of JAK kinases, alteration of IKZF1, hispanic/latino ethnicity, and a poor outcome in pediatric B-progenitor acute lymphoblastic leukemia. Blood. 2010;115:5312–5321. doi: 10.1182/blood-2009-09-245944. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Harvey RC, Mullighan CG, Wang X, et al. Identification of novel cluster groups in pediatric high-risk B-precursor acute lymphoblastic leukemia with gene expression profiling: Correlation with genome-wide DNA copy number alterations, clinical characteristics, and outcome. Blood. 2010;116:4874–4884. doi: 10.1182/blood-2009-08-239681. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Kang H, Chen IM, Wilson CS, et al. Gene expression classifiers for relapse-free survival and minimal residual disease improve risk classification and outcome prediction in pediatric B-precursor acute lymphoblastic leukemia. Blood. 2010;115:1394–1405. doi: 10.1182/blood-2009-05-218560. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Mullighan CG, Collins-Underwood JR, Phillips LA, et al. Rearrangement of CRLF2 in B-progenitorand Down syndrome-associated acute lymphoblastic leukemia. Nat Genet. 2009;41:1243–1246. doi: 10.1038/ng.469. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Mullighan CG, Phillips LA, Su X, et al. Genomic analysis of the clonal origins of relapsed acute lymphoblastic leukemia. Science. 2008;322:1377–1380. doi: 10.1126/science.1164266. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Mullighan CG, Su X, Zhang J, et al. Deletion of IKZF1 and prognosis in acute lymphoblastic leukemia. N Engl J Med. 2009;360:470–480. doi: 10.1056/NEJMoa0808253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Mullighan CG, Zhang J, Harvey RC, et al. Jak mutations in high-risk childhood acute lymphoblastic leukemia. Proc Natl Acad Sci USA. 2009;106:9414–9418. doi: 10.1073/pnas.0811761106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.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;10:125–134. doi: 10.1016/S1470-2045(08)70339-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Yang JJ, Bhojwani D, Yang W, et al. Genome-wide copy number profiling reveals molecular evolution from diagnosis to relapse in childhood acute lymphoblastic leukemia. Blood. 2008;112:4178–4183. doi: 10.1182/blood-2008-06-165027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Cario G, Zimmermann M, Romey R, et al. Presence of the P2RY8-CRLF2 rearrangement is associated with a poor prognosis in non-high-risk precursor B-cell acute lymphoblastic leukemia in children treated according to the ALL-BFM 2000 protocol. Blood. 2010;115:5393–5397. doi: 10.1182/blood-2009-11-256131. [DOI] [PubMed] [Google Scholar]
- 32.Hertzberg L, Vendramini E, Ganmore I, et al. Down syndrome acute lymphoblastic leukemia, a highly heterogeneous disease in which aberrant expression of CRLF2 is associated with mutated JAK2: A report from the international BFM study group. Blood. 2010;115:1006–1017. doi: 10.1182/blood-2009-08-235408. [DOI] [PubMed] [Google Scholar]
- 33.Zhang J, Mullighan CG, Harvey RC, et al. Key pathways are frequently mutated in high-risk childhood acute lymphoblastic leukemia: A report from the Children’s Oncology Group. Blood. 2011;118:3080–3087. doi: 10.1182/blood-2011-03-341412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Bhojwani D, Kang H, Menezes RX, et al. Gene expression signatures predictive of early response and outcome in high-risk childhood acute lymphoblastic leukemia: A Children’s Oncology Group study [corrected] J Clin Oncol. 2008;26:4376–4384. doi: 10.1200/JCO.2007.14.4519. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Hogan LE, Meyer JA, Yang J, et al. Integrated genomic analysis of relapsed childhood acute lymphoblastic leukemia reveals therapeutic strategies. Blood. 2012;118:5218–5226. doi: 10.1182/blood-2011-04-345595. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Loudin MG, Wang J, Leung HC, et al. Genomic profiling in down syndrome acute lymphoblastic leukemia identifies histone gene deletions associated with altered methylation profiles. Leukemia. 2011;25:1555–1563. doi: 10.1038/leu.2011.128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Ensor HM, Schwab C, Russell LJ, et al. Demographic, clinical and outcome features of children with acute lymphoblastic leukemia and CRLF2 deregulation: Results from the MRC ALL97 clinical trial. Blood. 2011;117:2129–2136. doi: 10.1182/blood-2010-07-297135. [DOI] [PubMed] [Google Scholar]
- 38.Chen IM, Harvey RC, Mullighan CG, et al. Outcome modeling with CRLF2, IKZF1, JAK, minimal residual disease in pediatric acute lymphoblastic leukemia: A Children’s Oncology Group study. Blood. 2012;119:3512–3522. doi: 10.1182/blood-2011-11-394221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Zhang J, Ding L, Holmfeldt L, et al. The genetic basis of early T-cell precursor acute lymphoblastic leukaemia. Nature. 2012;481:157–163. doi: 10.1038/nature10725. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Kang H, Wilson CS, Harvey RC, et al. Gene expression profiles predictive of outcome and age in infant acute lymphoblastic leukemia: A Children’s Oncology Group study. Blood. 2012;119:1872–1881. doi: 10.1182/blood-2011-10-382861. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Tasian SK, Doral MY, Borowitz MJ, et al. Aberrant STAT5 and PI3K/mTOR pathway signaling occurs in human CRLF2-rearranged B-precursor acute lymphoblastic leukemia. Blood. 2012;120:833–842. doi: 10.1182/blood-2011-12-389932. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Maude SL, Tasian SK, Vincent T, et al. Targeting JAK1/2 and mtor in murine xenograft models of Ph-like acute lymphoblastic leukemia. Blood. 2012;120:3510–3518. doi: 10.1182/blood-2012-03-415448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Morak M, Attarbaschi A, Fischer S, et al. Small sizes and indolent evolutionary dynamics challenge the potential role of P2RY8-CRLF2-harboring clones as main relapse-driving force in childhood all. Blood. 2012 doi: 10.1182/blood-2012-07-443218. [Epub ahead of print]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Dorge P, Meissner B, Zimmermann M, et al. IKZF1 deletion is an independent predictor of outcome in pediatric acute lymphoblastic leukemia treated according to the ALL-BFM 2000 protocol. Haematologica. 2012 doi: 10.3324/haematol.2011.056135. [Epub ahead of print]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Moorman AV, Schwab C, Ensor HM, et al. IGH@ translocations, CRLF2 deregulation, and micro-deletions in adolescents and adults with acute lymphoblastic leukemia. J Clin Oncol. 2012;30:3100–3108. doi: 10.1200/JCO.2011.40.3907. [DOI] [PubMed] [Google Scholar]
- 46.Palmi C, Vendramini E, Silvestri D, et al. Poor prognosis for P2RY8-CRLF2 fusion but not for CRLF2 over-expression in children with intermediate risk B-cell precursor acute lymphoblastic leukemia. Leukemia. 2012;26:2245–2253. doi: 10.1038/leu.2012.101. [DOI] [PubMed] [Google Scholar]
- 47.French D, Hamilton LH, Mattano LA, Jr, et al. A PAI-1 (SERPINE1) polymorphism predicts osteonecrosis in children with acute lymphoblastic leukemia: A report from the Children’s Oncology Group. Blood. 2008;111:4496–4499. doi: 10.1182/blood-2007-11-123885. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Trevino LR, Yang W, French D, et al. Germline genomic variants associated with childhood acute lymphoblastic leukemia. Nat Genet. 2009;41:1001–1005. doi: 10.1038/ng.432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Yang JJ, Cheng C, Yang W, et al. Genome-wide interrogation of germline genetic variation associated with treatment response in childhood acute lymphoblastic leukemia. JAMA. 2009;301:393–403. doi: 10.1001/jama.2009.7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Yang JJ, Cheng C, Devidas M, et al. Ancestry and pharmacogenomics of relapse in acute lymphoblastic leukemia. Nat Genet. 2011;43:237–241. doi: 10.1038/ng.763. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Xu H, Cheng C, Devidas M, et al. ARID5B genetic polymorphisms contribute to racial disparities in the incidence and treatment outcome of childhood acute lymphoblastic leukemia. J Clin Oncol. 2012;30:751–757. doi: 10.1200/JCO.2011.38.0345. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Yang JJ, Cheng C, Devidas M, et al. Genome-wide association study identifies germline polymorphisms associated with relapse of childhood acute lymphoblastic leukemia. Blood. 2012;120:4197–4204. doi: 10.1182/blood-2012-07-440107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Ellinghaus E, Stanulla M, Richter G, et al. Identification of germline susceptibility loci in ETV6-RUNX1-rearranged childhood acute lymphoblastic leukemia. Leukemia. 2012;26:902–909. doi: 10.1038/leu.2011.302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Orsi L, Rudant J, Bonaventure A, et al. Genetic polymorphisms and childhood acute lymphoblastic leukemia: GWAS of the ESCALE study (SFCE) Leukemia. 2012 doi: 10.1038/leu.2012.148. [Epub ahead of print]. [DOI] [PubMed] [Google Scholar]
- 55.Loh ML, Zhang J, Harvey RC, et al. Tyrosine kinome sequencing of high-risk Philadelphia chromosome-like pediatric acute lymphoblastic leukemia: A report from the Children’s Oncology Group TARGET Project. Blood. doi: 10.1182/blood-2012-04-422691. in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Bercovich D, Ganmore I, Scott LM, et al. Mutations of JAK2 in acute lymphoblastic leukaemias associated with Down’s syndrome. Lancet. 2008;372:1484–1492. doi: 10.1016/S0140-6736(08)61341-0. [DOI] [PubMed] [Google Scholar]
- 57.Russell LJ, Capasso M, Vater I, et al. Deregulated expression of cytokine receptor gene, CRLF2, is involved in lymphoid transformation in B-cell precursor acute lymphoblastic leukemia. Blood. 2009;114:2688–2698. doi: 10.1182/blood-2009-03-208397. [DOI] [PubMed] [Google Scholar]
- 58.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;22:153–166. doi: 10.1016/j.ccr.2012.06.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Arico M, Valsecchi MG, Camitta B, et al. Outcome of treatment in children with philadelphia chromosome-positive acute lymphoblastic leukemia. N Engl J Med. 2000;342:998–1006. doi: 10.1056/NEJM200004063421402. [DOI] [PubMed] [Google Scholar]
- 60.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;28:4755–4761. doi: 10.1200/JCO.2010.30.1325. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Schultz KR, Bowman WP, Aledo A, et al. Improved early event-free survival with imatinib in philadelphia chromosome-positive acute lymphoblastic leukemia: A Children’s Oncology Group study. J Clin Oncol. 2009;27:5175–5181. doi: 10.1200/JCO.2008.21.2514. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Slayton WB, Schultz KR, Jones T, et al. Outstanding early responses with dasatinib and intensive chemotherapy in pediatric philadelphia chromosome positive acute lymphoblastic leukemia (PH + ALL): Children’s oncology Group (COG) trial AALL0622. Pediatric Blood Cancer (SIOP Proceedings) 2012;59:988a. [Google Scholar]
- 63.Berg SL, Blaney SM, Devidas M, et al. Phase II study of nelarabine (compound 506U78) in children and young adults with refractory T-cell malignancies: A report from the Children’s Oncology Group. J Clin Oncol. 2005;23:3376–3382. doi: 10.1200/JCO.2005.03.426. [DOI] [PubMed] [Google Scholar]
- 64.Kurtzberg J, Ernst TJ, Keating MJ, et al. Phase I study of 506U78 administered on a consecutive 5-day schedule in children and adults with refractory hematologic malignancies. J Clin Oncol. 2005;23:3396–3403. doi: 10.1200/JCO.2005.03.199. [DOI] [PubMed] [Google Scholar]
- 65.Dunsmore KP, Devidas M, Linda SB, et al. Pilot study of nelarabine in combination with intensive chemotherapy in high-risk T-cell acute lymphoblastic leukemia: A report from the Children’s Oncology Group. J Clin Oncol. 2012;30:2753–2759. doi: 10.1200/JCO.2011.40.8724. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Matloub Y, Bostrom BC, Hunger SP, et al. Escalating intravenous methotrexate improves event-free survival in children with standard-risk acute lymphoblastic leukemia: A report for the Children’s Oncology Group. Blood. 2011;118:243–251. doi: 10.1182/blood-2010-12-322909. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.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;111:2548–2555. doi: 10.1182/blood-2007-02-070342. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Nachman J, Sather HN, Gaynon PS, et al. Augmented berlin-frankfurt-munster therapy abrogates the adverse prognostic significance of slow early response to induction chemotherapy for children and adolescents with acute lymphoblastic leukemia and unfavorable presenting features: A report from the Children’s Cancer Group. J Clin Oncol. 1997;15:2222–2230. doi: 10.1200/JCO.1997.15.6.2222. [DOI] [PubMed] [Google Scholar]
- 69.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;338:1663–1671. doi: 10.1056/NEJM199806043382304. [DOI] [PubMed] [Google Scholar]
- 70.Larsen EC, Salzer WL, Devidas M, et al. High dose methotrexate (HD-MTX) as compared to capizzi methotrexate plus asparaginase (c-MTX/ASNASE) improves event-free survival (EFS) in children and young adults with high-rsk acute lymphoblastic leukemia: A report from the Children’s Oncology Group study AALL 023. J Clin Oncol. 2011;29:6s. (abstr.). [Google Scholar]
- 71.Mattano LA, Nachman J, Devidas M, et al. Increased incidence of osteonecrosis (ON) with a dexamethasone (DEX) induction for high-risk acute lymphoblastic leukemia (HR-ALL): A report from the Children’s Oncology Group (COG) Blood (ASH Annual Meeting abstracts) 2008;112:898. [Google Scholar]
- 72.Winick NJ, Salzer WL, Devidas M, et al. Dexamethasone (DEX) versus prednisone (PRED) during induction for children with high risk acute lymphoblastic leukemia (HR-ALL): A report from the Children’s Oncology Group study AALL023. J Clin Oncol. 2011;29:586s. (abstr.). [Google Scholar]
- 73.Stock W, La M, Sanford B, et al. What determines the outcomes for adolescents and young adults with acute lymphoblastic leukemia treated on cooperative group protocols? A comparison of Children’s Cancer Group and cancer and leukemia group b studies. Blood. 2008;112:1646–1654. doi: 10.1182/blood-2008-01-130237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Schafer ES, Hunger SP. Optimal therapy for acute lymphoblastic leukemia in adolescents and young adults. Nat Rev Clin Oncol. 2011;8:417–424. doi: 10.1038/nrclinonc.2011.77. [DOI] [PubMed] [Google Scholar]
- 75.Nachman JB, La MK, Hunger SP, et al. Young adults with acute lymphoblastic leukemia have an excellent outcome with chemotherapy alone and benefit from intensive postinduction treatment: A report from the Children’s Oncology Group. J Clin Oncol. 2009;27:5189–5194. doi: 10.1200/JCO.2008.20.8959. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Larsen E, Raetz E, Winick N, et al. Outcome in adolescent and young adult (AYA) patients compared with younger patients treated for high risk B-precursor acute lymphoblastic leukemia (HR-ALL): A report from the children’s oncology group (COG) study AALL0232. J Clin Oncol (Meeting Abstracts) 2012;30(18 Suppl):CRA9508. [Google Scholar]
- 77.Larsen E, Salzer W, Devidas M, et al. Treatment toxicity in adolescents and young adult (AYA) patients compared with younger patients treated for high-risk B-precursor acute lymphoblastic leukemia (HR-ALL): A report from Children’s Oncology Group (COG) study aall0232. Blood (ASH Annual Meeting Abstracts) 2011;118:1510a. [Google Scholar]
- 78.Mattano LA, Jr, Devidas M, Nachman JB, et al. Effect of alternate-week versus continuous dexamethasone scheduling on the risk of osteonecrosis in paediatric patients with acute lymphoblastic leukaemia: Results from the CCG-1961 randomised cohort trial. Lancet Oncol. 2012;13:906–915. doi: 10.1016/S1470-2045(12)70274-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.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;370:240–250. doi: 10.1016/S0140-6736(07)61126-X. [DOI] [PubMed] [Google Scholar]
- 80.Armstrong SA, Kung AL, Mabon ME, et al. Inhibition of FLT3 in MLL. Validation of a therapeutic target identified by gene expression based classification. Cancer Cell. 2003;3:173–183. doi: 10.1016/s1535-6108(03)00003-5. [DOI] [PubMed] [Google Scholar]
- 81.Brown P, Levis M, Shurtleff S, et al. FLT3 inhibition selectively kills childhood acute lymphoblastic leukemia cells with high levels of FLT3 expression. Blood. 2005;105:812–820. doi: 10.1182/blood-2004-06-2498. [DOI] [PubMed] [Google Scholar]
- 82.Stam RW, den Boer ML, Schneider P, et al. Targeting FLT3 in primary mll-gene-rearranged infant acute lymphoblastic leukemia. Blood. 2005;106:2484–2490. doi: 10.1182/blood-2004-09-3667. [DOI] [PubMed] [Google Scholar]
- 83.Levis M, Allebach J, Tse KF, et al. A FLT3-targeted tyrosine kinase inhibitor is cytotoxic to leukemia cells in vitro and in vivo. Blood. 2002;99:3885–3891. doi: 10.1182/blood.v99.11.3885. [DOI] [PubMed] [Google Scholar]
- 84.Brown P, Levis M, McIntyre E, et al. Combinations of the FLT3 inhibitor CEP-701 and chemotherapy synergistically kill infant and childhood MLL-rearranged all cells in a sequence-dependent manner. Leukemia. 2006;20:1368–1376. doi: 10.1038/sj.leu.2404277. [DOI] [PubMed] [Google Scholar]
- 85.Brown P, Kaeding AJ, Magoon D, et al. Identification of a safe and biologically active dose of the FLT3 inhibitor lestaurtinib in combination with chemotherapy in infants with MLL-rearranged (MLL-R) all: A Children’s Oncology Group study. Pediatr Blood Cancer. 2011;899:302B. (abstr.). [Google Scholar]
- 86.Sun D, Kaeding AJ, Magoon D, et al. Safety and biological activity of the FLT3 inhibitor lestaurtinib in infant MLL-rearranged (MLL-R) all: Children’s oncology group protocol AALL0631. J Clin Oncol (Meeting Abstracts) 2012;30(15 Suppl):9548. [Google Scholar]
- 87.Raetz EA, Borowitz MJ, Devidas M, et al. Reinduction platform for children with first marrow relapse of acute lymphoblastic leukemia: A Children’s Oncology Group study [corrected] J Clin Oncol. 2008;26:3971–3978. doi: 10.1200/JCO.2008.16.1414. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Raetz EA, Cairo MS, Borowitz MJ, et al. Chemoimmunotherapy reinduction with epratuzumab in children with acute lymphoblastic leukemia in marrow relapse: A Children’s Oncology Group pilot study. J Clin Oncol. 2008;26:3756–3762. doi: 10.1200/JCO.2007.15.3528. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Raetz E, Cairo MS, Borowitz MJ, et al. Reinduction chemoimmunotherapy with epratuzumab in relapsed acute lymphoblastic leukemia (ALL) in children, adolescents, and young adults: Results from Children’s Oncology Group (COG) study ADVL04P2. Blood (ASH Annual Meeting Abstracts) 2011;118:573a. doi: 10.1002/pbc.25454. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Hijiya N, Gaynon P, Barry E, et al. A multi-center phase I study of clofarabine, etoposide and cyclophosphamide in combination in pediatric patients with refractory or relapsed acute leukemia. Leukemia. 2009;23:2259–2264. doi: 10.1038/leu.2009.185. [DOI] [PubMed] [Google Scholar]
- 91.Locatelli F, Testi AM, Bernardo ME, et al. Clofarabine, cyclophosphamide and etoposide as single-course re-induction therapy for children with refractory/multiple relapsed acute lymphoblastic leukaemia. Br J Haematol. 2009;147:371–378. doi: 10.1111/j.1365-2141.2009.07882.x. [DOI] [PubMed] [Google Scholar]
- 92.Hijiya N, Thomson B, Isakoff MS, et al. Phase 2 trial of clofarabine in combination with etoposide and cyclophosphamide in pediatric patients with refractory or relapsed acute lymphoblastic leukemia. Blood. 2011;118:6043–6049. doi: 10.1182/blood-2011-08-374710. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Horton TM, Gannavarapu A, Blaney SM, et al. Bortezomib interactions with chemotherapy agents in acute leukemia in vitro. Cancer Chemother Pharmacol. 2006;58:13–23. doi: 10.1007/s00280-005-0135-z. [DOI] [PubMed] [Google Scholar]
- 94.Houghton PJ, Morton CL, Kolb EA, et al. Initial testing (stage 1) of the proteasome inhibitor bortezomib by the pediatric preclinical testing program. Pediatr Blood Cancer. 2008;50:37–45. doi: 10.1002/pbc.21214. [DOI] [PubMed] [Google Scholar]
- 95.Szczepanek J, Pogorzala M, Konatkowska B, et al. Differential ex vivo activity of bortezomib in newly diagnosed paediatric acute lymphoblastic and myeloblastic leukaemia. Anticancer Res. 2010;30:2119–2124. [PubMed] [Google Scholar]
- 96.Horton TM, Pati D, Plon SE, et al. A phase 1 study of the proteasome inhibitor bortezomib in pediatric patients with refractory leukemia: A Children’s Oncology Group study. Clin Cancer Res. 2007;13:1516–1522. doi: 10.1158/1078-0432.CCR-06-2173. [DOI] [PubMed] [Google Scholar]
- 97.Messinger YH, Gaynon PS, Sposto R, et al. Bortezomib with chemotherapy is highly active in advanced B-precursor acute lymphoblastic leukemia: Therapeutic advances in Childhood Leukemia & Lymphoma (TACL) study. Blood. 2012;120:285–290. doi: 10.1182/blood-2012-04-418640. [DOI] [PubMed] [Google Scholar]
- 98.Messinger Y, Gaynon P, Raetz E, et al. Phase I study of bortezomib combined with chemotherapy in children with relapsed childhood acute lymphoblastic leukemia (ALL): A report from the therapeutic advances in childhood leukemia (TACL) consortium. Pediatr Blood Cancer. 2010;55:254–259. doi: 10.1002/pbc.22456. [DOI] [PubMed] [Google Scholar]
- 99.Schrappe M, Zimmermann M, Moricke A, et al. Dexamethasone in induction can eliminate one third of all relapses in childhood acute lymphoblastic leukemia (ALL): Results of an international randomized trial in 3655 patients (trial AEIOP-BFM ALL 2000) Blood (ASH Annual Meeting Abstracts) 2008;112:7. [Google Scholar]
- 100.Vora A, Wade R, Mitchell CD, et al. Improved outcome for children and young adults with t-cell acute lymphoblastic leukemia (ALL): Results of the United Kingdom Medical Research Council (MRC) trial ukall 2003. Blood (ASH Annual Meeting Abstracts) 2008;112:908. [Google Scholar]
- 101.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;376:2009–2017. doi: 10.1016/S0140-6736(10)62002-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Hunger SP, Baruchel A, Biondi A, et al. The thirteenth international childhood acute lymphoblastic leukemia workshop report: La Jolla, CA, USA, December 7–9, 2011. Pediatr Blood Cancer. 2012 doi: 10.1002/pbc.24354. [Epub ahead of print]. [DOI] [PubMed] [Google Scholar]
- 103.Nachman JB, Heerema NA, Sather H, et al. Outcome of treatment in children with hypodiploid acute lymphoblastic leukemia. Blood. 2007;110:1112–1115. doi: 10.1182/blood-2006-07-038299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Schrappe M, Hunger SP, Pui CH, et al. Outcomes after induction failure in childhood acute lymphoblastic leukemia. N Engl J Med. 2012;366:1371–1381. doi: 10.1056/NEJMoa1110169. [DOI] [PMC free article] [PubMed] [Google Scholar]
