• SCT in first complete remission is associated with 69.5% 3-year overall survival in high-risk ALL adult patients treated with intensified pediatric-like protocol.

  • Poor early MRD response is a powerful tool to select patients who may benefit from SCT in first complete remission.

Because a pediatric-inspired Group for Research on Adult Acute Lymphoblastic Leukemia (GRAALL) protocol yielded a markedly improved outcome in adults with Philadelphia chromosome–negative ALL, we aimed to reassess the role of allogeneic stem cell transplantation (SCT) in patients treated in the GRAALL-2003 and GRAALL-2005 trials. In all, 522 patients age 15 to 55 years old and presenting with at least 1 conventional high-risk factor were candidates for SCT in first complete remission. Among these, 282 (54%) received a transplant in first complete remission. At 3 years, posttransplant cumulative incidences of relapse, nonrelapse mortality, and relapse-free survival (RFS) were estimated at 19.5%, 15.5%, and 64.7%, respectively. Time-dependent analysis did not reveal a significant difference in RFS between SCT and no-SCT cohorts. However, SCT was associated with longer RFS in patients with postinduction minimal residual disease (MRD) ≥10−3 (hazard ratio, 0.40) but not in good MRD responders. In B-cell precursor ALL, SCT also benefitted patients with focal IKZF1 gene deletion (hazard ratio, 0.42). This article shows that poor early MRD response, in contrast to conventional ALL risk factors, is an excellent tool to identify patients who may benefit from allogeneic SCT in the context of intensified adult ALL therapy. Trial GRAALL-2003 was registered at www.clinicaltrials.gov as #NCT00222027; GRAALL-2005 was registered as #NCT00327678.

Medscape Continuing Medical Education online

This activity has been planned and implemented in accordance with the Essential Areas and policies of the Accreditation Council for Continuing Medical Education through the joint providership of Medscape, LLC and the American Society of Hematology.

Medscape, LLC is accredited by the ACCME to provide continuing medical education for physicians.

Medscape, LLC designates this Journal-based CME activity for a maximum of 1.0 AMA PRA Category 1 Credit(s)™. Physicians should claim only the credit commensurate with the extent of their participation in the activity.

All other clinicians completing this activity will be issued a certificate of participation. To participate in this journal CME activity: (1) review the learning objectives and author disclosures; (2) study the education content; (3) take the post-test with a 75% minimum passing score and complete the evaluation at http://www.medscape.org/journal/blood; and (4) view/print certificate. For CME questions, see page 2586.

Disclosures

The authors, Associate Editor Jacob M. Rowe, and CME questions author Laurie Barclay, freelance writer and reviewer, Medscape, LLC, declare no competing financial interests.

Learning objectives
  1. Describe outcomes of allogeneic stem cell transplantation (SCT) in first complete remission (CR1) among adults with high-risk acute lymphoblastic leukemia treated with intensified pediatric-like protocol, based on a review of clinical trial data.

  2. Describe the use of poor early minimal residual disease as a tool to select patients who may benefit from SCT in CR1, based on a review of clinical trial data.

  3. Describe the use of other risk factors as a tool to select patients who may benefit from SCT in CR1, based on a review of clinical trial data.

Release date: April 16, 2015; Expiration date: April 16, 2016

Many studies have recently reported that adolescents and young adults with acute lymphoblastic leukemia (ALL) may benefit from pediatric rather than adult chemotherapy protocols, leading to evaluation of pediatric-like approaches in adults.1-7  Several groups are now using pediatric-inspired or even unmodified pediatric protocols in younger adults.8-12  In the first study of the Group for Research on Adult ALL (GRAALL), we showed that a pediatric-inspired protocol markedly improved the outcome of adult patients up to 60 years of age.13  In this new setting, we reassessed the role of allogeneic hematopoietic stem cell transplantation (SCT) in first complete remission (CR1) in patients treated in the GRAALL-2003 and GRAALL-2005 trials.

As in childhood ALL, minimal residual disease (MRD) levels significantly correlate with clinical outcomes in adult ALL trials.13-21  They might thus be considered as a stratification tool, with the option to offer allogeneic SCT to patients with persistent molecular disease.16,18,20,22  Conversely, novel genetic alterations originally described in children have also been reported to have an impact on the outcome of adult patients. In the B-cell precursor (BCP-ALL) subgroup, focal deletions of the IKZF1 gene, observed in up to 80% of Philadelphia chromosome (Ph)-positive ALL, have been associated with a poor outcome in adult patients with Ph-negative ALL.23-29  In the T-cell ALL (T-ALL) subgroup, we and others have found that mutations of the NOTCH1 pathway are associated with a better outcome that could be altered by the presence of additional NRAS and/or KRAS gene mutation or PTEN gene alteration.30-34  We have recently shown that MRD levels combined with these new genetic markers may predict relapse more efficiently than conventional risk factors.21  We thus sought to retrospectively assess the potential contributions of MRD and oncogenetic profiles to identify patients who may significantly benefit from SCT in CR1.

GRAALL-2003 and GRAALL-2005 trials

The GRAALL-2003 and GRAALL-2005 trials were conducted between 2003 and 2011 in 70 centers in France, Belgium, and Switzerland. Protocols are provided in the supplemental Data, available online at the Blood Web site. Results of the GRAALL-2003 trial have already been reported.13  The subsequent GRAALL-2005 trial was very similar to the 2003 trial with the addition of randomized evaluation of hyperfractionated cyclophosphamide during induction and late intensification and rituximab during all phases of therapy in CD20+ BCP-ALL patients. Patients’ outcomes were last updated in January 2013. Per protocol, allogeneic SCT was offered in CR1 to patients age 55 years or younger who presented with at least 1 conventional ALL high-risk factor, as detailed below. Transplantation was planned after 3 or 6 blocks of consolidation, depending on the delay needed for donor identification. Most patients who received transplants from a related donor received 3 blocks of consolidation whereas those who received transplants from an unrelated donor mostly received 6 blocks. Informed consent was obtained from all patients at trial entry. Both trials were conducted in accordance with the declaration of Helsinki and approved by local and multicenter research ethical committees.

Risk factors used in the GRAALL-2003 and GRAALL-2005 trials

High-risk factors used to select patients eligible for allogeneic SCT in CR1 in these trials included (1) central nervous system involvement; (2) low hypodiploidy/near triploidy on karyotype and/or DNA index analysis; (3) early resistance to steroid prephase, defined as a peripheral blood (PB) blast cell count higher than 1.0 × 109/L after the prephase; (4) poor early bone marrow (BM) blast clearance, defined by morphologic BM blast cells of more than 5% after the first week of induction chemotherapy; (5) late CR, meaning that there was a need for the planned salvage course to reach CR; and (6) Ig/TCR MRD ≥10–2 after the first induction course in the GRAALL-2003 trial only. Additional factors were used in BCP-ALL patients, including white blood cell count (WBC) ≥30 × 109/L; MLL gene rearrangement, t[4;11] chromosomal translocation, and/or MLL-AF4 gene fusion, or other MLL rearrangement; and t(1;19) chromosomal translocation and/or E2A-PBX1 gene fusion. Two other factors were introduced in the GRAALL-2005 trial: complex karyotype and immature CD10 immunophenotype in BCP-ALL.

Study population

Overall, 955 patients age 15 to 60 years old with newly diagnosed Ph-negative ALL were enrolled in the GRAALL-2003 and GRAALL-2005 trials. Of the 880 patients (92%) who achieved CR, 811 were age 55 years or younger. Risk classification was available for 743 patients: 221 had standard-risk and 522 had high-risk ALL (Figure 1). The 5-year overall survival (OS) was 74% in the standard-risk group vs 58% in the high-risk group (supplemental Figure 1). These 522 high-risk patients were all candidates for SCT in CR1 and represented our study population.

Figure 1

Patient flowchart.

Figure 1

Patient flowchart.

Close modal

Transplant modalities

In the GRAALL-2003 trial, SCT modalities depended on age, type of risk factor, and donor availability. Three SCT levels were defined, with potentially increasing toxicity. Patients with t(1;19) and/or E2A-PBX1 could receive only an HLA-identical sibling transplant up to the age of 45 years. Other eligible patients could receive an HLA-identical sibling transplant up to the age of 55 years or a 10/10 HLA-matched unrelated transplant up to the age of 45 years. Patients with MRD level ≥10–2 after the first consolidation course could receive either an HLA-identical sibling or a 10/10 HLA-matched unrelated transplant up to the age of 55 years. In the GRAALL-2005 trial, all eligible patients could receive an HLA-identical sibling or a 10/10 HLA-matched unrelated donor transplant until age 55 years, but transplant from a 9/10 HLA-mismatched donor was acceptable in patients with t(4;11)/MLL abnormality, low hypodiploidy/near triploidy, or late CR. The planned conditioning regimen for GRAALL-2003 and GRAALL-2005 was cyclophosphamide 60 mg/kg on 2 days and 12 Gy fractionated or 10 Gy unfractionated total body irradiation. Graft-versus-host disease prophylaxis consisted of cyclosporine and short-course methotrexate.

MRD analysis

MRD-level quantification was based on patient-specific Ig/TCR gene rearrangement monitoring, centrally performed on BM samples after the first induction course (MRD1; 6 weeks after induction initiation) and after the first 3 blocks of consolidation (MRD2; 12 weeks after induction initiation), as described.21  Briefly, DNA was extracted from BM samples, and its quality was assessed by albumin gene quantification by using standardized quantitative real-time polymerase chain reaction (qRT-PCR).35  Potential Ig/TCR targets were identified by using the standardized multiplex PCR established within the BIOMED-2/EuroClonality network.36  For each patient, preferably 2 independent Ig/TCR targets with a sensitivity of at least 10−4 and a quantitative range of 10−4 for at least 1 of the 2 targets were selected for MRD-level monitoring. All MRD data were assessed according to the guidelines developed within the EuroMRD group.35 

New oncogenetic markers

BCP-ALL patients were centrally studied for focal IKZF1 gene deletion by using break point–specific multiplex PCR and multiplex ligation-probe assay, as described.21  T-ALL patients were centrally studied for the NOTCH1/FBXW7 gene mutation, NRAS and/or KRAS gene mutation, and PTEN gene alteration, as described.21  A T-ALL favorable genotype was defined by the presence of NOTCH1/FBXW7 mutation without NRAS and/or KRAS mutation or PTEN alteration, whereas the other combinations defined unfavorable genotypes.34 

Statistical methods

Binary variable comparisons were performed by using Fisher’s exact test. Median comparisons were performed with the Mann-Whitney 2-sample test. The primary end point was relapse-free survival (RFS). Other end points were cumulative incidence of relapse (CIR), nonrelapse-related mortality (NRM), and OS from CR. RFS was defined as the time between achievement of hematologic CR and relapse or death in CR1, other patients being censored at the time of last contact. When evaluating CIR (events being hematologic relapses) and NRM (events being deaths in CR1), estimations took into account deaths in CR1 and relapses as competing events, respectively. Outcome comparisons were performed by Cox models.37  To evaluate the effect of SCT, we performed time-dependent analyses considering SCT in CR1 as a time-dependent event. Outcome data were estimated by the Mantel-Byar method38  and graphically illustrated by Simon-Makuch plots,39  t0 being the time of hematologic CR achievement. This time-dependent methodology allowed avoidance of the bias caused by the time to transplant, at least when analyzing CIR, NRM, and RFS. To avoid the bias related to early relapses when evaluating OS with this method, because patients must be alive and in CR1 to receive a transplant, we used a 45-day landmark period for OS comparisons. Outcome comparisons were performed by Andersen-Gill models.40  Interactions between SCT effect and covariables were tested by introducing interaction terms in multivariable models. We have recently reported that when MRD response and lineage-specific genetic markers were introduced in a multivariable risk model, only these 2 factors remained independent relapse predictors in patients treated with GRAALL protocols.21  Advanced age had an impact only on NRM.

On the basis of these observations, the 2 covariates retained for multivariable analyses were as follows: (1) the classical age, WBC, and resistance to steroid prephase covariates in the entire patient population; and (2) age, MRD1 response, lineage-specific genetic markers (ie, t(4;11)/MLL abnormality and focal IKZF1 gene deletion in BCP-ALL patients, high-risk genetic classifier in T-ALL patients) in the subsets of patients evaluated for MRD1 level and genetic markers. We also performed a landmark donor vs no-donor analysis (supplemental Data). Type 1 error was fixed at the 5% level. All tests were 2-tailed. Hazard ratios (HRs) were given with 95% confidence intervals (CIs). Statistical analysis was performed on the STATA/IC 12.1 software package (STATA, College Station, TX).

Patient characteristics

Of the 522 study patients, 330 had a donor according to protocol criteria and 269 were actually transplanted in CR1. In addition, 13 patients received an unrelated cord blood (UCB) transplant in CR1, not planned by the protocol, leading to 282 patients (54%) in the SCT cohort. The reasons for 61 patients with a donor not receiving SCT in CR1 were early relapse (37 patients), investigator’s decision (11 patients), poor medical condition as a result of intercurrent adverse event (8 patients), early death in CR1 (2 patients), patient or donor refusal (2 patients), and unknown in the remaining patient. Characteristics of the 282 SCT and the 240 no-SCT patients are detailed in Table 1. There were no differences in baseline and early response characteristics between the two cohorts, except that more patients with t(4;11)/MLL abnormalities received SCT. Supplemental Table 1 shows a comparison between no-SCT patients with a donor and SCT patients or no-SCT patients without a donor.

Table 1

Patient characteristics (N = 522)

CharacteristicSCTno-SCTP
Patients 282 240  
Trial   .42 
 GRAALL-2003 65 63  
 GRAALL-2005 217 177  
Gender   .18 
 Male 165 155  
 Female 117 85  
Age, y   .53 
 Median 31.3 32.3  
 Range 16.3-55.9 16.6-55.9  
ECOG PS   .13 
 0 112 78  
 1 146 134  
 2 20 18  
 3  
 NA  
ALL lineage   .71 
 B 183 160  
 T 99 80  
WBC, 109/L   .30 
 Median 25.0 18.0  
 Range 0.7-456 0.5-573  
Identified donor*(n= 522)    
 Sibling 139 35 <.001 
 Unrelated 130 26 <.001 
 None 13 179 <.001 
B-lineage ALL patients (n = 343)    
 No. of patients 183 160  
 WBC ≥30 × 109 /L 73 58 .51 
 CNS disease at diagnosis   .52 
  Yes 13 10  
  No 169 147  
  NA  
 EGIL immunophenotype 20 18 .15 
  I 60 44  
  II 61 61  
  III 27 32  
  IV  
  NA 11  
 t(4;11)/MLL gene rearrangement§   .015 
  Yes 46 23  
  No 136 133  
  NA  
 t(1;19)   .40 
  Yes 12 11  
  No 167 141  
  NA  
 Complex karyotype   .09 
  Yes 12 16  
  No 152 117  
  NA 19 27  
 Low hypodiploidy/near triploidy   .21 
  Yes 13  
  No 160 131  
  NA 16 16  
 Focal IKZF1 gene deletion   .87 
  Yes 29 29  
  No 80 68  
  NA 74 63  
 Resistance to steroid prephase   .14 
  Yes 52 34  
  No 131 126  
 Poor early BM blast clearance   .22 
  Yes 104 79  
  No 68 74  
  NA 11  
 Late CR   .10 
  Yes 10  
  No 173 155  
 Post-induction MRD1 level ≥10−3   .71 
  Yes 27 26  
  No 64 51  
  NA 82 80  
T-ALL patients (n= 179)    
 Patients 99 80  
 WBC ≥100 × 109/L 24 25 .32 
 CNS disease at diagnosis   .87 
  Yes 19 13  
  No 78 66  
  NA  
 EGIL immunophenotype   .68 
  I  
  II 42 31  
  III 33 27  
  IV 10 11  
  NA  
 Complex karyotype   .86 
  Yes 13 10  
  No 74 58  
  NA 12 12  
TLX1 gene overexpression   .43 
  Yes 11  
  No 69 49  
  NA 19 22  
NOTCH1/FBXW7 gene mutation   .20 
  Yes 53 33  
  No 28 25  
  NA 18 22  
 High-risk 4-gene classifier   .37 
  Yes 35 34  
  No 37 22  
  NA 27 24  
 Resistance to steroid prephase   .74 
  Yes 63 50  
  No 36 29  
  NA  
 Poor early BM blast clearance   .34 
  Yes 71 53  
  No 26 27  
  NA  
 Late CR   .99 
  Yes  
  No 96 77  
 Postinduction MRD1 level ≥10−3   .78 
  Yes 19 14  
  No 30 28  
  NA 47 35  
CharacteristicSCTno-SCTP
Patients 282 240  
Trial   .42 
 GRAALL-2003 65 63  
 GRAALL-2005 217 177  
Gender   .18 
 Male 165 155  
 Female 117 85  
Age, y   .53 
 Median 31.3 32.3  
 Range 16.3-55.9 16.6-55.9  
ECOG PS   .13 
 0 112 78  
 1 146 134  
 2 20 18  
 3  
 NA  
ALL lineage   .71 
 B 183 160  
 T 99 80  
WBC, 109/L   .30 
 Median 25.0 18.0  
 Range 0.7-456 0.5-573  
Identified donor*(n= 522)    
 Sibling 139 35 <.001 
 Unrelated 130 26 <.001 
 None 13 179 <.001 
B-lineage ALL patients (n = 343)    
 No. of patients 183 160  
 WBC ≥30 × 109 /L 73 58 .51 
 CNS disease at diagnosis   .52 
  Yes 13 10  
  No 169 147  
  NA  
 EGIL immunophenotype 20 18 .15 
  I 60 44  
  II 61 61  
  III 27 32  
  IV  
  NA 11  
 t(4;11)/MLL gene rearrangement§   .015 
  Yes 46 23  
  No 136 133  
  NA  
 t(1;19)   .40 
  Yes 12 11  
  No 167 141  
  NA  
 Complex karyotype   .09 
  Yes 12 16  
  No 152 117  
  NA 19 27  
 Low hypodiploidy/near triploidy   .21 
  Yes 13  
  No 160 131  
  NA 16 16  
 Focal IKZF1 gene deletion   .87 
  Yes 29 29  
  No 80 68  
  NA 74 63  
 Resistance to steroid prephase   .14 
  Yes 52 34  
  No 131 126  
 Poor early BM blast clearance   .22 
  Yes 104 79  
  No 68 74  
  NA 11  
 Late CR   .10 
  Yes 10  
  No 173 155  
 Post-induction MRD1 level ≥10−3   .71 
  Yes 27 26  
  No 64 51  
  NA 82 80  
T-ALL patients (n= 179)    
 Patients 99 80  
 WBC ≥100 × 109/L 24 25 .32 
 CNS disease at diagnosis   .87 
  Yes 19 13  
  No 78 66  
  NA  
 EGIL immunophenotype   .68 
  I  
  II 42 31  
  III 33 27  
  IV 10 11  
  NA  
 Complex karyotype   .86 
  Yes 13 10  
  No 74 58  
  NA 12 12  
TLX1 gene overexpression   .43 
  Yes 11  
  No 69 49  
  NA 19 22  
NOTCH1/FBXW7 gene mutation   .20 
  Yes 53 33  
  No 28 25  
  NA 18 22  
 High-risk 4-gene classifier   .37 
  Yes 35 34  
  No 37 22  
  NA 27 24  
 Resistance to steroid prephase   .74 
  Yes 63 50  
  No 36 29  
  NA  
 Poor early BM blast clearance   .34 
  Yes 71 53  
  No 26 27  
  NA  
 Late CR   .99 
  Yes  
  No 96 77  
 Postinduction MRD1 level ≥10−3   .78 
  Yes 19 14  
  No 30 28  
  NA 47 35  

CNS, central nervous system; ECOG PS, Eastern Cooperative Oncology Group performance status; EGIL, European Group for the Immunological Characterization of Leukemias; NA, not applicable; WBC, white blood cell count.

*

Numbers of patients with a donor identified by the GRAALL-2003 or GRAALL-2005 protocol criteria (supplemental Data).

These 13 patients received cord blood SCT.

Conventional high-risk factor used in the GRAALL trials.

§

59 of the 69 patients with MLL gene rearrangement had t(4;11) translocation.

In patients who reached hematologic CR after the first induction cycle.

Overall, SCT was performed from a matched sibling donor in 139 patients and an unrelated donor in 143 patients, including 92 fully 10/10 HLA-matched, 38 9/10 HLA-mismatched, and 13 with UCB. The source of hematopoietic stem cells was BM in 184 patients, PB in 85, and UCB in 13. Of note, 10 patients received reduced-intensity conditioning regimen and 17 patients were conditioned without total body irradiation. Median time between CR1 achievement and SCT was 106 days (range, 24 to 380 days) after a median number of 4 consolidation blocks (range, 0 to 6 consolidation blocks). As expected, this time was significantly shorter in patients who received SCT from a sibling donor compared with other patients (87 vs 123 days; P < .001). Accordingly, patients from the sibling donor subgroup received fewer pretransplant consolidation blocks (median, 3 vs 6 consolidation blocks; P < .001).

Outcome of patients who received allogeneic SCT in CR1

Among the 282 patients who were transplanted in CR1 and had a median posttransplant follow-up of 3.5 years, 53 patients relapsed and 89 patients died, including 46 deaths in CR1. At 3 years, posttransplant CIR and NRM were 19.5% (95% CI, 15% to 25%) and 15.5% (95% CI, 12% to 20%), respectively. This resulted in 3-year posttransplant RFS estimates of 64.7% (95% CI, 595 to 70%) and OS estimates of 69.5% (95% CI, 63% to 75%). As shown in Table 2, no difference was observed between BCP-ALL and T-ALL patients. The type of donor did not influence RFS (supplemental Figure 2). Advanced age and administration of a higher number of pretransplant consolidation cycles were both associated with a significantly higher posttransplant NRM (Table 2). Upon adjustment of donor type, age ≥45 years (HR, 1.9; P = .013) and more than 3 pretransplant consolidation cycles (HR, 2.0; P = .004) both remained independently associated with a higher NRM without significant impact on RFS.

Table 2

Outcome of SCT patients

3-year RFS (%)95% CIP3-year CIR (%)95% CIP3-year NRM (%)95% CIP3-year OS (%)95% CIP
All patients (N = 282) 64.7 59-70  19.5 15-25  15.5 12-20  69.5 63-75  
Age, y   .12   .85   .048   .082 
 15-44 67.0 60-73  19.8 15-26  12.9 9-18  71.6 65-77  
 45-55 55.6 41-68  18.2 10-31  25.8 16-40  60.7 46-72  
Donor   .91   .12   .057   .80 
 HLA-identical sibling 63.6 55-71  23.0 17-31  12.8 8-20  69.3 60-77  
 Unrelated 65.8 57-73  16.0 11-23  17.9 12-25  69.6 61-77  
ALL lineage   .55   .80   .74   .34 
 BCP 62.8 55-70  20.1 15-27  16.7 12-23  67.7 60-74  
 T 68.1 58-76  18.5 12-28  13.3 8-22  72.5 62-80  
No. of pre-SCT consolidation blocks   .49   .27   .0009   .38 
 1-3 66.5 58-74  22.2 16-30  11.0 7-18  71.8 62-79  
 4-6 62.9 54-71  16.9 12-24  19.9 14-28  67.1 58-74  
3-year RFS (%)95% CIP3-year CIR (%)95% CIP3-year NRM (%)95% CIP3-year OS (%)95% CIP
All patients (N = 282) 64.7 59-70  19.5 15-25  15.5 12-20  69.5 63-75  
Age, y   .12   .85   .048   .082 
 15-44 67.0 60-73  19.8 15-26  12.9 9-18  71.6 65-77  
 45-55 55.6 41-68  18.2 10-31  25.8 16-40  60.7 46-72  
Donor   .91   .12   .057   .80 
 HLA-identical sibling 63.6 55-71  23.0 17-31  12.8 8-20  69.3 60-77  
 Unrelated 65.8 57-73  16.0 11-23  17.9 12-25  69.6 61-77  
ALL lineage   .55   .80   .74   .34 
 BCP 62.8 55-70  20.1 15-27  16.7 12-23  67.7 60-74  
 T 68.1 58-76  18.5 12-28  13.3 8-22  72.5 62-80  
No. of pre-SCT consolidation blocks   .49   .27   .0009   .38 
 1-3 66.5 58-74  22.2 16-30  11.0 7-18  71.8 62-79  
 4-6 62.9 54-71  16.9 12-24  19.9 14-28  67.1 58-74  

Overall effect of SCT

Among the 240 no-SCT patients, 107 relapsed and 107 died, including 21 deaths in CR1. When analyzing SCT in CR1 as a time-dependent event in the whole study population, RFS (HR, 0.80; 95% CI, 0.60 to 1.06; P = .12) and OS (HR, 0.76; 95% CI, 0.57 to 1.02; P = .069) were not significantly improved in the SCT cohort. The lower CIR (HR, 0.50; 95% CI, 0.35 to 0.70; P < .001) observed in the SCT cohort was counterbalanced by a higher NRM (HR, 1.46; 95% CI, 1.09 to 1.95; P = .011) (Table 3). This is illustrated in Figure 2, which shows Simon-Makuch plots for RFS and OS in both SCT and no-SCT cohorts.

Table 3

Comparison of SCT and no-SCT patient outcomes in prespecified patient subsets (time-dependent analysis)

PatientsSCT patientsCIR HR*95% CIP*RFS HR*95% CIP*OS HR95% CIP
All patients 522 282 0.50 0.35-0.70 <.001 0.80 0.60-1.06 .12 0.76 0.57-1.02 .069 
B-ALL 343 183 0.54 0.35-0.84 .006 0.81 0.57-1.15 .23 0.75 0.53-1.0 .11 
T-ALL 179 99 0.42 0.23-0.76 .004 0.76 0.46-1.26 .29 0.79 0.46-1.35 .39 
Age 15-44 y 414 226 0.45 0.31-0.66 <.001 0.77 0.55-1.06 .11 0.74 0.53-1.04 .085 
Age 45-55 y 108 56 0.74 0.32-1.68 .46 0.93 0.52-1.66 .81 0.84 0.46-1.52 .56 
CNS disease at diagnosis 55 32 0.40 0.14-1.18 .096 1.20 0.50-2.90 .68 1.13 0.50-2.57 .77 
Complex karyotype 51 25 0.80 0.25-2.55 .70 1.60 0.65-3.92 .30 1.36 0.55-3.40 .50 
WBC ≥3 × 109/L (B-ALL) 131 73 0.65 0.35-1.21 .18 0.74 0.43-1.28 .28 0.60 0.35-1.01 .056 
CD10 immature ALL (B-ALL) 125 77 1.14 0.50-2.60 .75 1.64 0.86-3.12 .14 1.09 0.59-2.02 .78 
t(4;11)/MLL gene rearrangement (B-ALL) 69 46 0.50 0.20-1.26 .14 0.71 0.33-1.56 .40 0.48 0.24-0.98 .044 
t(1;19) (B-ALL) 23 12 0.26 0.05-1.30 .10 0.37 0.09-1.49 .16 0.41 0.10-1.72 .22 
Resistance to steroid prephase 199 115 0.63 0.37-1.05 .077 0.85 0.55-1.32 .48 0.81 0.51-1.30 .39 
Poor early BM blast clearance 307 175 0.57 0.37-0.86 .008 0.67 0.47-0.97 .034 0.65 0.44-0.95 .028 
Late CR 19 13 0.46 0.20-1.02 .055 0.40 0.19-0.83 .014 0.21 0.05-0.80 .023 
PatientsSCT patientsCIR HR*95% CIP*RFS HR*95% CIP*OS HR95% CIP
All patients 522 282 0.50 0.35-0.70 <.001 0.80 0.60-1.06 .12 0.76 0.57-1.02 .069 
B-ALL 343 183 0.54 0.35-0.84 .006 0.81 0.57-1.15 .23 0.75 0.53-1.0 .11 
T-ALL 179 99 0.42 0.23-0.76 .004 0.76 0.46-1.26 .29 0.79 0.46-1.35 .39 
Age 15-44 y 414 226 0.45 0.31-0.66 <.001 0.77 0.55-1.06 .11 0.74 0.53-1.04 .085 
Age 45-55 y 108 56 0.74 0.32-1.68 .46 0.93 0.52-1.66 .81 0.84 0.46-1.52 .56 
CNS disease at diagnosis 55 32 0.40 0.14-1.18 .096 1.20 0.50-2.90 .68 1.13 0.50-2.57 .77 
Complex karyotype 51 25 0.80 0.25-2.55 .70 1.60 0.65-3.92 .30 1.36 0.55-3.40 .50 
WBC ≥3 × 109/L (B-ALL) 131 73 0.65 0.35-1.21 .18 0.74 0.43-1.28 .28 0.60 0.35-1.01 .056 
CD10 immature ALL (B-ALL) 125 77 1.14 0.50-2.60 .75 1.64 0.86-3.12 .14 1.09 0.59-2.02 .78 
t(4;11)/MLL gene rearrangement (B-ALL) 69 46 0.50 0.20-1.26 .14 0.71 0.33-1.56 .40 0.48 0.24-0.98 .044 
t(1;19) (B-ALL) 23 12 0.26 0.05-1.30 .10 0.37 0.09-1.49 .16 0.41 0.10-1.72 .22 
Resistance to steroid prephase 199 115 0.63 0.37-1.05 .077 0.85 0.55-1.32 .48 0.81 0.51-1.30 .39 
Poor early BM blast clearance 307 175 0.57 0.37-0.86 .008 0.67 0.47-0.97 .034 0.65 0.44-0.95 .028 
Late CR 19 13 0.46 0.20-1.02 .055 0.40 0.19-0.83 .014 0.21 0.05-0.80 .023 
*

HR in SCT vs no-SCT cohort by the Andersen-Gill model.

OS comparisons were performed using a 45-day landmark period, excluding patients who died or relapsed during the first 45 days following CR achievement from comparisons (see “Statistical methods” section).

Figure 2

Effect of SCT on (A) RFS and (B) OS. The effect of SCT in CR1 is represented on Simon-Makuch plots (SCT as a time-dependent covariable). Time t0 is CR achievement time. A 45-day landmark period was used for OS comparison. The black line represents the no-SCT patient cohort, and the gray line represents the SCT cohort.

Figure 2

Effect of SCT on (A) RFS and (B) OS. The effect of SCT in CR1 is represented on Simon-Makuch plots (SCT as a time-dependent covariable). Time t0 is CR achievement time. A 45-day landmark period was used for OS comparison. The black line represents the no-SCT patient cohort, and the gray line represents the SCT cohort.

Close modal

As shown in Table 3, no significant effect of SCT on RFS was observed in patients younger or older than age 45 years. No effect was seen in patients with BCP-ALL or T-ALL when analyzed separately or in the different patient subsets defined by the prespecified baseline risk factors used in the protocols. Conversely, RFS and OS were significantly longer in the SCT than in the no-SCT cohort in patients who presented with morphologic poor early BM blast clearance or in late CR patients.

Effect of SCT according to early MRD level

This latter observation led us to analyze the time-dependent effect of SCT according to postinduction MRD1 level (Table 4). Among the 522 study patients, 259 had MRD1 evaluation and 19 additional patients were not in CR at this time (late CR patients), meaning that 278 patients (53%) showed the presence of residual disease. As shown in supplemental Table 2, characteristics of these 278 patients did not differ from those of the 244 other patients. MRD1 level was ≥10−3 in 105 (37.7%) of those patients including the 19 late CR patients, with no difference between the SCT and the no-SCT cohorts (38.5% and 36.8%, respectively).

Table 4

Comparison of SCT and no-SCT patient outcomes according to oncogenetic markers and early MRD response (time-dependent analysis)

PatientsSCT patientsCIR HR*95% CIP*PinteractionRFS HR*95% CIP*PinteractionOS HR95% CIPPinteraction
Focal IKZF1 deletion (BCP-ALL)               
 Yes 58 29 0.43 0.18-1.07 .07 .72 0.42 0.19-0.89 .025 .078 0.35 0.16-0.75 .007 .071 
 No 148 80 0.44 0.22-0.90 .025  0.83 0.48-1.44 .51  0.87 0.49-1.55 .64  
NOTCH1/FBXW7 mutation (T-ALL)               
 Yes 86 53 0.36 0.13-1.01 .053 .46 0.68 0.28-1.65 .39 .61 0.67 0.26-1.73 .41 .68 
 No 53 28 0.60 0.26-1.41 .25  1.03 0.45-2.19 .93  1.01 0.44-2.30 .98  
4-gene classifier (T-ALL)               
 High-risk 69 35 0.58 0.28-1.23 .15 .41 1.01 0.53-1.95 .97 .98 0.96 0.46-1.98 .91 .93 
 Low-risk 59 37 0.27 0.05-1.45 .13  0.82 0.23-2.91 .76  0.75 0.20-2.79 .66  
MRD1 level ≥10−3 or late CR (all)               
 Yes 105 59 0.39 0.21-0.71 .002 .24 0.40 0.23-0.69 .001 .001 0.41 0.23-0.74 .003 .002 
 No 173 94 0.63 0.33-1.20 .16  1.37 0.81-2.32 .24  1.47 0.85-2.54 .16  
MRD1 level ≥10−3 or late CR (BCP-ALL)               
 Yes 66 37 0.50 0.24-1.06 .069 .99 0.36 0.18-0.73 .004 .007 0.43 .21-.90 .025 .050 
 No 115 64 0.55 0.24-1.29 .17  1.34 0.70-2.54 .38  1.13 .61-2.11 .70  
MRD1 level ≥10−3 or late CR (T-ALL)               
 Yes 39 22 0.25 0.08-0.75 .014 .053 0.48 0.19-1.19 .11 .038 0.40 .15-1.06 .066 .010 
 No 58 30 0.83 0.30-2.30 .72  1.42 0.57-3.53 .46  2.67 .83-8.55 .10  
PatientsSCT patientsCIR HR*95% CIP*PinteractionRFS HR*95% CIP*PinteractionOS HR95% CIPPinteraction
Focal IKZF1 deletion (BCP-ALL)               
 Yes 58 29 0.43 0.18-1.07 .07 .72 0.42 0.19-0.89 .025 .078 0.35 0.16-0.75 .007 .071 
 No 148 80 0.44 0.22-0.90 .025  0.83 0.48-1.44 .51  0.87 0.49-1.55 .64  
NOTCH1/FBXW7 mutation (T-ALL)               
 Yes 86 53 0.36 0.13-1.01 .053 .46 0.68 0.28-1.65 .39 .61 0.67 0.26-1.73 .41 .68 
 No 53 28 0.60 0.26-1.41 .25  1.03 0.45-2.19 .93  1.01 0.44-2.30 .98  
4-gene classifier (T-ALL)               
 High-risk 69 35 0.58 0.28-1.23 .15 .41 1.01 0.53-1.95 .97 .98 0.96 0.46-1.98 .91 .93 
 Low-risk 59 37 0.27 0.05-1.45 .13  0.82 0.23-2.91 .76  0.75 0.20-2.79 .66  
MRD1 level ≥10−3 or late CR (all)               
 Yes 105 59 0.39 0.21-0.71 .002 .24 0.40 0.23-0.69 .001 .001 0.41 0.23-0.74 .003 .002 
 No 173 94 0.63 0.33-1.20 .16  1.37 0.81-2.32 .24  1.47 0.85-2.54 .16  
MRD1 level ≥10−3 or late CR (BCP-ALL)               
 Yes 66 37 0.50 0.24-1.06 .069 .99 0.36 0.18-0.73 .004 .007 0.43 .21-.90 .025 .050 
 No 115 64 0.55 0.24-1.29 .17  1.34 0.70-2.54 .38  1.13 .61-2.11 .70  
MRD1 level ≥10−3 or late CR (T-ALL)               
 Yes 39 22 0.25 0.08-0.75 .014 .053 0.48 0.19-1.19 .11 .038 0.40 .15-1.06 .066 .010 
 No 58 30 0.83 0.30-2.30 .72  1.42 0.57-3.53 .46  2.67 .83-8.55 .10  
*

HR in SCT vs no-SCT cohort by the Andersen-Gill model.

OS comparisons were performed by using a 45-day landmark period, excluding patients who died or relapsed during the first 45 days following CR achievement from comparisons (see “Statistical methods” section).

As detailed in Table 4, patients with MRD1 level <10−3 did not benefit from SCT in CR1 in term of RFS and OS, whereas those with a poor MRD1 response did benefit (Figure 3A-B). Tests for interactions were highly significant (RFS: P = .001; OS: P = .002; Table 4). Similar significant interactions were observed in the BCP-ALL and T-ALL subgroups when analyzed separately, even if the impact of SCT in poor MRD1 responders did not reach statistical significance in T-ALL patients (Table 4; illustrated for RFS in supplemental Figures 3 and 4). Supplemental Figure 5 illustrates that this interaction was also observed in the subset of 178 patients with poor early morphologic BM blast clearance, suggesting that MRD1 was a better tool than earlier blast clearance to define patients who may benefit from SCT. Similar results were found when excluding the 19 late CR patients from the analysis (data not shown).

Figure 3

Effect of SCT on (A) RFS and (B) OS, according to early MRD1 response. The effect of SCT in CR1 is represented on Simon-Makuch plots (SCT as a time-dependent covariable). Time t0 is CR achievement time. A 45-day landmark period was used for OS comparison. Solid lines represent patients who had MRD1 level ≥10−3 at t0 or late CR patients, and dashed lines represent patients who had MRD1 level <10−3 at t0. Black lines represent the no-SCT patient cohort, and gray lines represent the SCT cohort.

Figure 3

Effect of SCT on (A) RFS and (B) OS, according to early MRD1 response. The effect of SCT in CR1 is represented on Simon-Makuch plots (SCT as a time-dependent covariable). Time t0 is CR achievement time. A 45-day landmark period was used for OS comparison. Solid lines represent patients who had MRD1 level ≥10−3 at t0 or late CR patients, and dashed lines represent patients who had MRD1 level <10−3 at t0. Black lines represent the no-SCT patient cohort, and gray lines represent the SCT cohort.

Close modal

Effect of SCT according to lineage-specific oncogenetic markers

In BCP-ALL patients, focal IKZF1 deletion was detected in 58 (28%) of 206 evaluable patients. Patients with IKZF1 deletion appeared to significantly benefit from SCT in terms of RFS and OS, whereas those without IKZF1 deletion did not (Table 4 and supplemental Figure 6 for RFS). As previously reported,21  the presence of IKZF1 deletion was strongly correlated with poor MRD1 response (60% vs 29% in poor MRD1 responders; P < .001). The role of SCT in the small subset of patients with MRD1 level <10−3 despite IKZF1 deletion (n = 18) could not be evaluated.

In T-ALL patients, a high-risk genetic profile according to our 4-gene classifier was found in 69 (54%) of 128 evaluable patients. Even if patient numbers were lower, RFS and OS appeared to be comparable in both SCT and no-SCT cohorts for both high-risk and low-risk genetic subsets, suggesting that high-risk profiles retained similar unfavorable prognostic value whether patients received SCT or not (Table 4 and supplemental Figure 7 for RFS).

Multivariable models

In the whole patient population, the time-dependent effect of SCT on RFS and OS still did not reach the significance level after adjustment on age, WBC, and resistance to steroid prephase in a multivariable model (RFS: HR, 0.79; 95% CI, 0.59 to 1.05; P = .10; OS: HR, 0.76; 95% CI, 0.57 to 1.03; P = .074). The same was observed when analyzing BCP-ALL and T-ALL patients separately. For BCP-ALL patients, HR was 0.77 (95% CI, 0.55 to 1.10; P = .15) for RFS and HR was 0.72 (95% CI, 0.51 to 1.03; P = .071) for OS. For T-ALL patients, HR was 0.83 (95% CI, 0.50 to 1.39; P = .47) for RFS and HR was 0.89 (95% CI, 0.51 to 1.56; P = .68) for OS. In the 278 patients studied for MRD1 levels, the significant interaction between MRD response and SCT effect was still observed after adjustment on the same covariates. For poor MRD1 responders, HR was 0.37 (95% CI, 0.20 to 0.69; P = .001) for RFS and HR was 0.41 (95% CI, 0.22 to 0.76; P = .005) for OS. For good MRD1 responders, HR was 1.38 (95% CI, 0.82 to 2.33; P = .23) for RFS and HR was 1.47 (95% CI, 0.85 to 2.54; P = .17) for OS. For RFS, Pinteraction = .001 and for OS, Pinteraction = .002.

Although we were limited by patient numbers, we performed multivariable analyses that included MRD1 response, lineage-specific genetic markers, age, and SCT as a time-dependent covariate in the 146 BCP-ALL and 76 T-ALL patients tested for MRD1 and genetic markers. In BCP-ALL patients, MRD1 response (RFS: P = .001; OS: P = .048), t(4;11)/MLL abnormality (RFS: P = .012; OS: P = .003), and IKZF1 deletion (RFS: P = .006; OS: P = .008) were the 3 factors that influenced RFS and OS. The effect of SCT remained apparent in patients with MRD1 ≥10−3 only (RFS: HR, 0.27; 95% CI, 0.12 to 0.60; P = .001; OS: HR, 0.32; 95% CI, 0.14 to 0.76; P = .01), with interactions between MRD1 response and SCT effect (RFS: P = .028; OS: P = 0.12). In T-ALL patients, MRD1 response (RFS: P = .019; OS: P = .038) and the 4-gene classifier (RFS: P = .009; OS: P = .086) were the two factors influencing RFS and OS. The effect of SCT in patients with MRD1 ≥10−3 did not reach the significance level, but interactions between MRD1 response and SCT effect were still observed (RFS: P = .061; OS: P = .034).

In this study, we reassessed the role of allogeneic SCT in CR1 in adult patients with high-risk Ph-negative ALL treated with a pediatric-inspired protocol. We highlighted the value of early MRD quantification and IKZF1 gene deletion to discriminate patients who may currently benefit from SCT. To date, this study is the largest one that specifically investigated the relationship between early MRD response and SCT effect in a time-dependent manner.

Despite prior intensive 5-drug induction and dose-dense consolidation, posttransplant outcome was relatively good. With a 3-year NRM of 15.5% and a 3-year post-SCT survival of almost 70%, this outcome compared favorably with that of previous cohorts of ALL patients receiving myeloablative SCT in CR1.18,41-43  A higher NRM was nonetheless observed in patients age 45 years or older, which was consistent with the UKALL-12/ECOG E2993 trial and the recent donor vs no donor meta-analysis, both of which used the cutoff of age 35 years.44,45  Remarkably, patients who received transplants from related and unrelated donors displayed similar RFS. Similar results have been reported by other groups, which provided reassurance for considering unrelated as well as related donors for patients with an indication of SCT in CR1.42  Conversely, the excess of NRM observed in patients age 45 years or older encouraged us to prospectively evaluate the role of reduced-intensity conditioning SCT in these patients.

Because of the high proportion of SCT performed by using unrelated donors who are frequently not identified at the time of CR achievement, we used a time-dependent rather than a donor vs no-donor methodology to analyze the effect of SCT. We also performed a donor vs no-donor analysis (supplementary Table 3). We failed to find evidence of a beneficial SCT effect in the overall study population of patients defined as eligible for SCT by conventional risk factors. As is frequently observed, the lower CIR observed in the SCT cohort (HR, 0.50) was counterbalanced by a higher NRM resulting in comparable RFS (HR, 0.80). Furthermore, the analysis of the baseline high-risk factors did not identify any subset of patients for whom SCT significantly prolonged RFS. This is not totally surprising because those conventional factors were thought to be associated with a higher relapse risk rather than to interfere with transplant-related mortality. This nonetheless indicated that those factors were not the appropriate ones to define patients who could really benefit from SCT. This result appears to be conflicting with previous reports that show a superiority of allogeneic SCT over chemotherapy or autologous SCT.44-48  In several studies, however, allogeneic SCT was offered to all suitable patients with a donor, whereas it was offered only to selected high-risk patients in the GRAALL trials. Of importance, differences in the high-risk ALL definition, which could include age or not, might also have an impact or even yield opposite conclusions.44  Finally, improvement in the results of chemotherapy through the use of an intensified pediatric-like protocol is probably the main explanation for the lack of superiority of allogeneic SCT over chemotherapy in this article. The 50% RFS estimate at 5 years observed in our no-SCT cohort appears to be higher than that usually reported in similar patients after chemotherapy and compares favorably with results of allogeneic SCT in other studies.44-48 

Nevertheless, we observed a significant effect of SCT in patients with poor early blast clearance or in late CR patients. This led us to evaluate the SCT effect according to early MRD response. In adult ALL, MRD response has been evaluated at various time points using different techniques and threshold cutoffs.16-18,20,49  Good MRD response was generally defined as an MRD level of <10−3 to 10−4 between weeks 5 and 10 and/or MRD level of <10−4 or not detectable between weeks 11 and 17. For logistical reasons, an earlier time point is more convenient for risk-oriented SCT decision. We thus used the 10−3 cutoff after the first induction course in this study. Using this cutoff at this time point, we showed that SCT erased the unfavorable prognostic impact of poor MRD response, whereas good MRD responders did not benefit from transplantation with significant interactions. Similar interactions were observed in BCP-ALL and T-ALL patients when analyzed separately. By using landmark analysis, the German Multicenter Study Group for Adult ALL (GMALL) has also reported that poor MRD responders receiving SCT displayed a better outcome than those treated with chemotherapy.18  Conversely, no superiority of SCT over chemotherapy was observed in patients with a poor early cytologic/MRD response in the Programa Español de Tratamientos en Hematología (PETHEMA) ALL-AR-03 trial, a result that could be hampered by a lower 5-year DFS after allogeneic transplant (32%) than expected in ALL in CR1.20 

We also investigated the role of SCT in patient subsets defined by newly described oncogenetic markers. In BCP-ALL patients, the presence of poor-prognosis IKZF1 gene deletions was also predictive of a positive SCT effect but was strongly related to poor early MRD response. Conversely, in T-ALL patients, unfavorable genetic profiles were not predictive of any positive SCT effect and were not significantly related to poor early MRD response. From that point of view, it is very interesting to observe that the same T-cell genetic classification has recently been demonstrated as a strong outcome predictor in children and adults with T-cell lymphoblastic lymphoma, a thymic disease in which BM MRD evaluation is obviously of lower interest.50,51  For all these reasons, in the next GRAALL-2014 trial, we will base the decision of SCT in CR1 on early MRD response only.

In summary, we here report relatively good results associated with myeloablative SCT in CR1 in adults with Ph-negative high-risk ALL treated with a pediatric-inspired protocol. In this new setting, conventional baseline risk factors did not appear to satisfactorily identify SCT indications. Conversely, we confirm in a large prospective study that early MRD response is the best and maybe the unique tool to optimally select the patients who may currently benefit from SCT in CR1.

The online version of this article contains a data supplement.

There is an Inside Blood Commentary on this article in this issue.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

The authors thank the patients and their families for participating in the Group for Research on Adult Acute Lymphoblastic Leukemia (GRAALL) trials and the GRAALL investigators (listed in the supplemental Data) for submitting clinical data and samples; Elodie Boucher, Nathalie Klein, Cyril Melot, Mathieu Sauvezie, and Francis Daniel for their help in data monitoring; Gérard Socié, Xavier Thomas, Elizabeth Macintyre, and Marie-Christine Béné for reviewing the manuscript; Nicole Raus, Ibrahim Yakoub-Agha, and the Société Française de Greffe de Moelle et de Thérapie Cellulaire for sharing data; and André Baruchel and Gérard Socié for their long-standing intellectual support.

This work was supported by grants 0200701, AOM 04144, and AOM 08106 from the Programme Hospitalier de Recherche Clinique, French Ministry of Health and Institut National du Cancer (H.D.), and by the Swiss State Secretariat for Education, Research and Innovations in Switzerland. Samples were collected and processed by the Assistance Publique-Hôpitaux de Paris Direction de Recherche Clinique Tumor Bank at Necker-Enfants Malades.

Written on behalf of the Group for Research on Adult Acute Lymphoblastic Leukemia (GRAALL). The GRAALL includes the former France-Belgium Group for Lymphoblastic Acute Leukemia in Adults, the French Western-Eastern Group for Lymphoblastic Acute Leukemia, and the Swiss Group for Clinical Cancer Research. GRAALL participating centers and investigators are listed in the supplemental Data.

Contribution: N.D., A.H., S.M., X.T., C.G., Y.C., J.-Y.C., V.L., H.D., and N.I. conceived and designed the study; V.L. provided administrative support; N.D., A.H., S.M., R.T., X.T., P.C., S.N., V.C., J.-H.B., Y.H., M.E.-B., O.R., C.G., Y.C., D.B., J.-Y.C., H.D., and N.I. provided study materials or patients; N.D., A.H., S.M., K.B., V.A., V.L., H.D., and N.I. collected and assembled data; N.D., A.H., S.M., K.B., V.A., V.L., H.D., and N.I. analyzed and interpreted data; N.D., A.H., S.M., H.D., and N.I. helped write the manuscript; and all authors gave final approval to the manuscript.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Norbert Ifrah, Service des Maladies du Sang, Centre Hospitalier Universitaire, 4 rue Larrey, 49933 Angers Cedex 9, France; e-mail: noifrah@chu-angers.fr; Hervé Dombret, Université Paris Diderot, Institut Universitaire d’Hématologie, EA-3518, Hôpital Saint-Louis, 1 avenue Claude Vellefaux, 75475 Paris Cedex 10, France; e-mail: herve.dombret@sls.aphp.fr.

1
Boissel
 
N
Auclerc
 
M-F
Lhéritier
 
V
, et al. 
Should adolescents with acute lymphoblastic leukemia be treated as old children or young adults? Comparison of the French FRALLE-93 and LALA-94 trials.
J Clin Oncol
2003
, vol. 
21
 
5
(pg. 
774
-
780
)
2
de Bont
 
JM
Holt
 
B
Dekker
 
AW
van der Does-van den Berg
 
A
Sonneveld
 
P
Pieters
 
R
Significant difference in outcome for adolescents with acute lymphoblastic leukemia treated on pediatric vs adult protocols in the Netherlands.
Leukemia
2004
, vol. 
18
 
12
(pg. 
2032
-
2035
)
3
Hallböök
 
H
Gustafsson
 
G
Smedmyr
 
B
Söderhäll
 
S
Heyman
 
M
Swedish Adult Acute Lymphocytic Leukemia Group; Swedish Childhood Leukemia Group
Treatment outcome in young adults and children >10 years of age with acute lymphoblastic leukemia in Sweden: a comparison between a pediatric protocol and an adult protocol.
Cancer
2006
, vol. 
107
 
7
(pg. 
1551
-
1561
)
4
Ramanujachar
 
R
Richards
 
S
Hann
 
I
, et al. 
Adolescents with acute lymphoblastic leukaemia: outcome on UK national paediatric (ALL97) and adult (UKALLXII/E2993) trials.
Pediatr Blood Cancer
2007
, vol. 
48
 
3
(pg. 
254
-
261
)
5
Stock
 
W
La
 
M
Sanford
 
B
, et al. 
Children’s Cancer Group; Cancer and Leukemia Group B studies
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
, vol. 
112
 
5
(pg. 
1646
-
1654
)
6
Haïat
 
S
Marjanovic
 
Z
Lapusan
 
S
, et al. 
Outcome of 40 adults aged from 18 to 55 years with acute lymphoblastic leukemia treated with double-delayed intensification pediatric protocol.
Leuk Res
2011
, vol. 
35
 
1
(pg. 
66
-
72
)
7
Advani
 
AS
Sanford
 
B
Luger
 
S
, et al. 
Frontline-Treatment Of Acute Lymphoblastic Leukemia (ALL) In Older Adolescents and Young Adults (AYA) Using a Pediatric Regimen Is Feasible: Toxicity Results of the Prospective US Intergroup Trial C10403 (Alliance) [abstract].
Blood
2013
, vol. 
122
 
21
 
Abstract 3903
8
Ribera
 
J-M
Oriol
 
A
Sanz
 
M-A
, et al. 
Comparison of the results of the treatment of adolescents and young adults with standard-risk acute lymphoblastic leukemia with the Programa Español de Tratamiento en Hematología pediatric-based protocol ALL-96.
J Clin Oncol
2008
, vol. 
26
 
11
(pg. 
1843
-
1849
)
9
Rijneveld
 
AW
van der Holt
 
B
Daenen
 
SMGJ
, et al. 
Dutch-Belgian HOVON Cooperative group
Intensified chemotherapy inspired by a pediatric regimen combined with allogeneic transplantation in adult patients with acute lymphoblastic leukemia up to the age of 40.
Leukemia
2011
, vol. 
25
 
11
(pg. 
1697
-
1703
)
10
Rytting
 
ME
Thomas
 
DA
O’Brien
 
S
, et al. 
 
Augmented Berlin-Frankfurt- Münster therapy in adolescents and young adults (AYAs) with acute lymphoblastic leukemia (ALL). Cancer. 2014;120(23):3660-3668
11
Gökbuget
 
N
Hoelzer
 
D
Treatment of adult acute lymphoblastic leukemia.
Hematology Am Soc Hematol Educ Program
2006
(pg. 
133
-
141
)
12
Faderl
 
S
Thomas
 
DA
O’Brien
 
S
, et al. 
Augmented hyper-CVAD based on dose-intensified vincristine, dexamethasone, and asparaginase in adult acute lymphoblastic leukemia salvage therapy.
Clin Lymphoma Myeloma Leuk
2011
, vol. 
11
 
1
(pg. 
54
-
59
)
13
Huguet
 
F
Leguay
 
T
Raffoux
 
E
, et al. 
Pediatric-inspired therapy in adults with Philadelphia chromosome-negative acute lymphoblastic leukemia: the GRAALL-2003 study.
J Clin Oncol
2009
, vol. 
27
 
6
(pg. 
911
-
918
)
14
Brüggemann
 
M
Raff
 
T
Flohr
 
T
, et al. 
German Multicenter Study Group for Adult Acute Lymphoblastic Leukemia
Clinical significance of minimal residual disease quantification in adult patients with standard-risk acute lymphoblastic leukemia.
Blood
2006
, vol. 
107
 
3
(pg. 
1116
-
1123
)
15
Holowiecki
 
J
Krawczyk-Kulis
 
M
Giebel
 
S
, et al. 
Status of minimal residual disease after induction predicts outcome in both standard and high-risk Ph-negative adult acute lymphoblastic leukaemia. The Polish Adult Leukemia Group ALL 4-2002 MRD Study.
Br J Haematol
2008
, vol. 
142
 
2
(pg. 
227
-
237
)
16
Bassan
 
R
Spinelli
 
O
Oldani
 
E
, et al. 
Improved risk classification for risk-specific therapy based on the molecular study of minimal residual disease (MRD) in adult acute lymphoblastic leukemia (ALL).
Blood
2009
, vol. 
113
 
18
(pg. 
4153
-
4162
)
17
Patel
 
B
Rai
 
L
Buck
 
G
, et al. 
Minimal residual disease is a significant predictor of treatment failure in non T-lineage adult acute lymphoblastic leukaemia: final results of the international trial UKALL XII/ECOG2993.
Br J Haematol
2010
, vol. 
148
 
1
(pg. 
80
-
89
)
18
Gökbuget
 
N
Kneba
 
M
Raff
 
T
, et al. 
German Multicenter Study Group for Adult Acute Lymphoblastic Leukemia
Adult patients with acute lymphoblastic leukemia and molecular failure display a poor prognosis and are candidates for stem cell transplantation and targeted therapies.
Blood
2012
, vol. 
120
 
9
(pg. 
1868
-
1876
)
19
Nagafuji
 
K
Miyamoto
 
T
Eto
 
T
, et al. 
 
Monitoring of minimal residual disease (MRD) is useful to predict prognosis of adult patients with Ph-negative ALL: results of a prospective study (ALL MRD2002 Study). J Hematol Oncol. 2013;6:14
20
Ribera
 
J-M
Oriol
 
A
Morgades
 
M
, et al. 
Treatment of high-risk Philadelphia chromosome-negative acute lymphoblastic leukemia in adolescents and adults according to early cytologic response and minimal residual disease after consolidation assessed by flow cytometry: final results of the PETHEMA ALL-AR-03 trial.
J Clin Oncol
2014
, vol. 
32
 
15
(pg. 
1595
-
1604
)
21
Beldjord
 
K
Chevret
 
S
Asnafi
 
V
, et al. 
Group for Research on Adult Acute Lymphoblastic Leukemia (GRAALL)
Oncogenetics and minimal residual disease are independent outcome predictors in adult patients with acute lymphoblastic leukemia.
Blood
2014
, vol. 
123
 
24
(pg. 
3739
-
3749
)
22
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
)
23
Mullighan
 
CG
Su
 
X
Zhang
 
J
, et al. 
Children’s Oncology Group
Deletion of IKZF1 and prognosis in acute lymphoblastic leukemia.
N Engl J Med
2009
, vol. 
360
 
5
(pg. 
470
-
480
)
24
Iacobucci
 
I
Storlazzi
 
CT
Cilloni
 
D
, et al. 
Identification and molecular characterization of recurrent genomic deletions on 7p12 in the IKZF1 gene in a large cohort of BCR-ABL1-positive acute lymphoblastic leukemia patients: on behalf of Gruppo Italiano Malattie Ematologiche dell’Adulto Acute Leukemia Working Party (GIMEMA AL WP).
Blood
2009
, vol. 
114
 
10
(pg. 
2159
-
2167
)
25
Martinelli
 
G
Iacobucci
 
I
Storlazzi
 
CT
, et al. 
IKZF1 (Ikaros) deletions in BCR-ABL1-positive acute lymphoblastic leukemia are associated with short disease-free survival and high rate of cumulative incidence of relapse: a GIMEMA AL WP report.
J Clin Oncol
2009
, vol. 
27
 
31
(pg. 
5202
-
5207
)
26
Kuiper
 
RP
Waanders
 
E
van der Velden
 
VHJ
, et al. 
IKZF1 deletions predict relapse in uniformly treated pediatric precursor B-ALL.
Leukemia
2010
, vol. 
24
 
7
(pg. 
1258
-
1264
)
27
Iacobucci
 
I
Iraci
 
N
Messina
 
M
, et al. 
IKAROS deletions dictate a unique gene expression signature in patients with adult B-cell acute lymphoblastic leukemia.
PLoS ONE
2012
, vol. 
7
 
7
pg. 
e40934
 
28
Mullighan
 
CG
 
The molecular genetic makeup of acute lymphoblastic leukemia. Hematology Am Soc Hematol Educ Program. 2012;2012:389-396
29
Dörge
 
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
2013
, vol. 
98
 
3
(pg. 
428
-
432
)
30
Breit
 
S
Stanulla
 
M
Flohr
 
T
, et al. 
Activating NOTCH1 mutations predict favorable early treatment response and long-term outcome in childhood precursor T-cell lymphoblastic leukemia.
Blood
2006
, vol. 
108
 
4
(pg. 
1151
-
1157
)
31
Kox
 
C
Zimmermann
 
M
Stanulla
 
M
, et al. 
The favorable effect of activating NOTCH1 receptor mutations on long-term outcome in T-ALL patients treated on the ALL-BFM 2000 protocol can be separated from FBXW7 loss of function.
Leukemia
2010
, vol. 
24
 
12
(pg. 
2005
-
2013
)
32
Ben Abdelali
 
R
Asnafi
 
V
Leguay
 
T
, et al. 
Group for Research on Adult Acute Lymphoblastic Leukemia
Pediatric-inspired intensified therapy of adult T-ALL reveals the favorable outcome of NOTCH1/FBXW7 mutations, but not of low ERG/BAALC expression: a GRAALL study.
Blood
2011
, vol. 
118
 
19
(pg. 
5099
-
5107
)
33
Jenkinson
 
S
Koo
 
K
Mansour
 
MR
, et al. 
Impact of NOTCH1/FBXW7 mutations on outcome in pediatric T-cell acute lymphoblastic leukemia patients treated on the MRC UKALL 2003 trial.
Leukemia
2013
, vol. 
27
 
1
(pg. 
41
-
47
)
34
Trinquand
 
A
Tanguy-Schmidt
 
A
Ben Abdelali
 
R
, et al. 
Toward a NOTCH1/FBXW7/RAS/PTEN-based oncogenetic risk classification of adult T-cell acute lymphoblastic leukemia: a Group for Research in Adult Acute Lymphoblastic Leukemia study.
J Clin Oncol
2013
, vol. 
31
 
34
(pg. 
4333
-
4342
)
35
van der Velden
 
VHJ
Panzer-Grümayer
 
ER
Cazzaniga
 
G
, et al. 
Optimization of PCR-based minimal residual disease diagnostics for childhood acute lymphoblastic leukemia in a multi-center setting.
Leukemia
2007
, vol. 
21
 
4
(pg. 
706
-
713
)
36
van Dongen
 
JJM
Langerak
 
AW
Brüggemann
 
M
, et al. 
Design and standardization of PCR primers and protocols for detection of clonal immunoglobulin and T-cell receptor gene recombinations in suspect lymphoproliferations: report of the BIOMED-2 Concerted Action BMH4-CT98-3936.
Leukemia
2003
, vol. 
17
 
12
(pg. 
2257
-
2317
)
37
Cox
 
DR
Regression models and life tables.
J R Stat Soc B
1972
, vol. 
34
 
2
(pg. 
187
-
220
)
38
Mantel
 
N
Byar
 
DP
Evaluation of response-time data involving transient states: an illustration using heart-transplant data.
J Am Stat Assoc
1974
, vol. 
69
 
345
(pg. 
81
-
86
)
39
Simon
 
R
Makuch
 
RW
A non-parametric graphical representation of the relationship between survival and the occurrence of an event: application to responder versus non-responder bias.
Stat Med
1984
, vol. 
3
 
1
(pg. 
35
-
44
)
40
Andersen
 
PK
Gill
 
RD
Cox’s regression model for counting processes: a large sample study.
Ann Stat
1982
, vol. 
10
 
4
(pg. 
1100
-
1120
)
41
Gorin
 
N-C
Labopin
 
M
Polge
 
E
, et al. 
Acute Leukaemia Working Party of the European Cooperative Group for Blood and Marrow Transplantation (EBMT)
Risk assessment in adult acute lymphoblastic leukaemia before early haemopoietic stem cell transplantation with a geno-identical donor: an easy clinical prognostic score to identify patients who benefit most from allogeneic haemopoietic stem cell transplantation.
Leukemia
2003
, vol. 
17
 
8
(pg. 
1596
-
1599
)
42
Kiehl
 
MG
Kraut
 
L
Schwerdtfeger
 
R
, et al. 
Outcome of allogeneic hematopoietic stem-cell transplantation in adult patients with acute lymphoblastic leukemia: no difference in related compared with unrelated transplant in first complete remission.
J Clin Oncol
2004
, vol. 
22
 
14
(pg. 
2816
-
2825
)
43
Giebel
 
S
Labopin
 
M
Holowiecki
 
J
, et al. 
Outcome of HLA-matched related allogeneic hematopoietic stem cell transplantation for patients with acute leukemia in first complete remission treated in Eastern European centers. Better results in recent years.
Ann Hematol
2009
, vol. 
88
 
10
(pg. 
1005
-
1013
)
44
Goldstone
 
AH
Richards
 
SM
Lazarus
 
HM
, et al. 
In adults with standard-risk acute lymphoblastic leukemia, the greatest benefit is achieved from a matched sibling allogeneic transplantation in first complete remission, and an autologous transplantation is less effective than conventional consolidation/maintenance chemotherapy in all patients: final results of the International ALL Trial (MRC UKALL XII/ECOG E2993).
Blood
2008
, vol. 
111
 
4
(pg. 
1827
-
1833
)
45
Gupta
 
V
Richards
 
S
Rowe
 
J
Acute Leukemia Stem Cell Transplantation Trialists’ Collaborative Group
Allogeneic, but not autologous, hematopoietic cell transplantation improves survival only among younger adults with acute lymphoblastic leukemia in first remission: an individual patient data meta-analysis.
Blood
2013
, vol. 
121
 
2
(pg. 
339
-
350
)
46
Hunault
 
M
Harousseau
 
J-L
Delain
 
M
, et al. 
GOELAMS (Groupe Ouest-Est des Leucémies Airguës et Maladies du Sang) Group
Better outcome of adult acute lymphoblastic leukemia after early genoidentical allogeneic bone marrow transplantation (BMT) than after late high-dose therapy and autologous BMT: a GOELAMS trial.
Blood
2004
, vol. 
104
 
10
(pg. 
3028
-
3037
)
47
Cornelissen
 
JJ
van der Holt
 
B
Verhoef
 
GEG
, et al. 
Dutch-Belgian HOVON Cooperative Group
Myeloablative allogeneic versus autologous stem cell transplantation in adult patients with acute lymphoblastic leukemia in first remission: a prospective sibling donor versus no-donor comparison.
Blood
2009
, vol. 
113
 
6
(pg. 
1375
-
1382
)
48
Kako
 
S
Morita
 
S
Sakamaki
 
H
, et al. 
A decision analysis of allogeneic hematopoietic stem cell transplantation in adult patients with Philadelphia chromosome-negative acute lymphoblastic leukemia in first remission who have an HLA-matched sibling donor.
Leukemia
2011
, vol. 
25
 
2
(pg. 
259
-
265
)
49
Brüggemann
 
M
Raff
 
T
Kneba
 
M
Has MRD monitoring superseded other prognostic factors in adult ALL?
Blood
2012
, vol. 
120
 
23
(pg. 
4470
-
4481
)
50
Callens
 
C
Baleydier
 
F
Lengline
 
E
, et al. 
Clinical impact of NOTCH1 and/or FBXW7 mutations, FLASH deletion, and TCR status in pediatric T-cell lymphoblastic lymphoma.
J Clin Oncol
2012
, vol. 
30
 
16
(pg. 
1966
-
1973
)
51
Lepretre
 
S
Touzart
 
A
Vermeulin
 
T
, et al. 
 
Pediatric ALL-like Therapy in Adults with T-cell Lymphoblastic Lymphoma: Results of the GRAALL-LYSA LL03 study [abstract]. Blood 2014;124(21). Abstract 371

Author notes

N.D. and A.H. contributed equally to this study.

H.D. and N.I. contributed equally to this study.

Sign in via your Institution