To define a role for hematopoietic stem cell transplantation (HSCT) in infants with acute lymphoblastic leukemia and rearrangements of the mixed-lineage-leukemia gene (MLL+), we compared the outcome of MLL+ patients from trial Interfant-99 who either received chemotherapy only or HSCT. Of 376 patients with a known MLL status in the trial, 297 (79%) were MLL+. Among the 277 of 297 MLL+ patients (93%) in first remission (CR), there appeared to be a significant difference in disease-free survival (adjusted by waiting time to HSCT) between the 37 (13%) who received HSCT and the 240 (87%) who received chemotherapy only (P = .03). However, the advantage was restricted to a subgroup with 2 additional unfavorable prognostic features: age less than 6 months and either poor response to steroids at day 8 or leukocytes more than or equal to 300 g/L. Ninety-seven of 297 MLL+ patients (33%) had such high-risk criteria, with 87 achieving CR. In this group, HSCT was associated with a 64% reduction in the risk of failure resulting from relapse or death in CR (hazard ratio = 0.36, 95% confidence interval, 0.15-0.86). In the remaining patients, there was no advantage for HSCT over chemotherapy only. In summary, HSCT seems to be a valuable option for a subgroup of infant MLL+ acute lymphoblastic leukemia carrying further poor prognostic factors. The trial was registered at www.clinicaltrials.gov as #NCT00015873 and at www.controlled-trials.com as #ISRCTN24251487.

Acute lymphoblastic leukemia (ALL) diagnosed within the first 12 months of life accounts for 2.5% to 5% of pediatric ALL cases and displays unique biologic, clinical, and prognostic features that vary from those of older children with ALL.1-5  Approximately 80% of infant cases harbor rearrangements of the mixed-lineage-leukemia (MLL) gene at chromosome band 11q23 (MLL+).1,6,7  The prognosis is relatively poor with only approximately half of cases achieving a cure with current therapies.1,6  Whether the proportion of patients cured can be increased by the more frequent use of concepts, such as hematopoietic stem cell transplantation (HSCT) in first remission or intensification of contemporarily applied hybrid protocols, including both lymphoid- and myeloid-directed treatment elements, is a matter of debate.8  Moreover, possible long-term adverse effects of the preparative regimen before and of graft-versus-host disease (GVHD) after HSCT, disturbing the physical development in such a young and vulnerable patient population, demand for a very strict indication only left for infants with a predictably very poor chance to maintain first remission with chemotherapy alone.9,10 

In the first large international multicenter trial for the treatment of infant ALL, Interfant-99, allogeneic HSCT was a possible option (but not mandated) for patients with initially poorly responding leukemia, defined by a poor response to steroids at day 8 of chemotherapy.6  However, no significant benefit of HSCT was observed in this cohort of high-risk patients when disease-free survival (DFS) was adjusted for the waiting time to HSCT. This was in accordance with the largest retrospective study reported until then, which suggested that there was no clinical benefit associated with HSCT in infants with MLL+ ALL.11,12  Nevertheless, because of the unique opportunity to analyze a large cohort of homogeneously treated infant ALL cases (483 patients), we aimed to more precisely define a possible role of HSCT in newly defined prognostic subgroups of infants with MLL+ ALL.

Patients

Between May 1999 and January 2006, a total of 483 infants (up to 365 days of age) with previously untreated ALL were entered into the international study Interfant-99 by 17 collaborative study groups in 22 countries.6  ALL was diagnosed according to standard morphologic, cytochemical, and immunologic criteria.13  The detection of MLL rearrangements was preferentially based on reverse transcription polymerase chain reaction or MLL split-signal fluorescence in situ hybridization analysis.2,14  However, classic 11q23 translocations without reverse transcription polymerase chain reaction/fluorescence in situ hybridization confirmation also counted for MLL+ cases. Patients were treated with informed consent from the patient's parents or legal guardians. Studies were conducted in accordance with the Declaration of Helsinki, and approval was delivered by the ethic committees of all participating institutions.

Patient characteristics, including clinical and laboratory features at diagnosis, the MLL status, and early response to therapy as well as outcome of randomized chemotherapeutic treatment arms, overall outcome, and multivariate analysis of risk factors for treatment failure, have been previously published.6  Of the 483 patients, 376 (78%) were evaluable for the MLL status and other relevant prognostic factors, such as age and white blood cell (WBC) counts at diagnosis, immunophenotype, response to a 7-day pro-phase with predniso(lo)ne and 1 intrathecal dose of methotrexate at day 1, and remission status after completion of induction therapy.

Risk factors for a relapse gained from the multivariate analysis of the total study population are as follows (based on their hierarchy of statistical significance)6 : MLL rearrangement (MLL+ vs MLL), age at diagnosis (< 6 months vs ≥ 6 months), predniso(lo)ne response (good predniso(lo)ne response [GPR]: < 1.0 g/L blasts vs poor predniso(lo)ne response [PPR]: ≥ 1.0 g/L blasts at day 8), and WBC counts in peripheral blood at diagnosis (≥ 300 g/L vs < 300 g/L).6,15  Based on these findings, 3 new subgroups with a distinct relapse risk were defined to design a subsequent new study, Interfant-06. Therein, patients are assigned to 1 of 3 treatment branches: a low-risk group defined by the absence of MLL rearrangements (MLL), a high-risk group with MLL positivity (MLL+), age less than 6 months, and either WBC counts more than or equal to 300 g/L or PPR, and a medium-risk group, including all others. This risk stratification will be used throughout the paper to illustrate the results.

Treatment

Treatment stratification and protocols of the Interfant-99 study have been published previously, and response criteria as well as definition of relapse have been described in detail elsewhere.6  Briefly, patients were assigned to 1 of 2 treatment branches: a standard-risk group for the GPR and a high-risk group for the PPR patients. All patients received a 5-week induction therapy (prednisone for 7 and afterward dexamethasone for 21 days, vincristine [VCR], daunorubicin, L-asparaginase, low-dose cytarabine, and intrathecals) followed by a 4-week consolidation (high-dose methotrexate [MTX], 6-mercaptopurine [6-MP], high-dose cytarabine, L-asparaginase, and intrathecals) and a 7-week reinduction therapy (dexamethasone, 6-thioguanine, VCR, daunorubicin, low-dose cytarabine, cyclophosphamide, and intrathecals). GPR patients in complete remission (CR) randomly continued with a late 4-week reintensification element (similar to consolidation but with additional VCR) and maintenance therapy (6-MP, MTX, steroid/VCR pulses) or maintenance therapy only up to a total therapy duration of 24 months. PPR patients in CR who were not allocated to allogeneic HSCT randomly continued with a late 4-week reintensification (same protocol as for the GPR patients) and intensified maintenance therapy (6-MP, MTX, steroid/VCR, and etoposide/cytarabine pulses) or intensified maintenance therapy only up to a total treatment duration of 24 months. Doses of the antileukemic drugs used were adjusted according to patients' age at the start of each treatment phase: children younger than 6 months were given two-thirds of the full dose, those 6 to 12 months of age were given three-fourths of the dose, and those 12 months of age or older were given the full dose. Intrathecal drugs (methotrexate, cytarabine, and prednisolone) were given in age-dependent fixed dosages.

Indications for HSCT in study Interfant-99 were defined by the protocol and reserved for patients with PPR, being performed after completion of reinduction therapy. Of note, Interfant-99 was not designed to study a benefit or lack of benefit of HSCT in infant (MLL+) ALL. Donor selection and type of the conditioning regimen as well as of GVHD prophylaxis and treatment were at the discretion of the participating study groups and transplantation centers. The European Bone Marrow Transplantation and the international Berlin-Frankfurt-Münster study committees advised a preparative regimen with etoposide, busulfan, and cyclophosphamide. Total body irradiation (TBI) was strongly discouraged because of concerns about long-term side effects. Cyclosporin A was recommended for GVHD prophylaxis.

Statistical considerations

Event-free survival (EFS), DFS, and overall survival (OS) curves were estimated with the Kaplan-Meier method and their SE with the Greenwood formula. The starting point for the observation time was the date of diagnosis for EFS and OS, whereas the date of first complete remission (CR1) was used for DFS. Death in induction, resistance, relapse, secondary malignancy, and death in continuous CR (CCR) were considered events in the calculation of EFS rates. Relapse, secondary malignancy, and death in CCR were events in the DFS estimation, whereas death from any cause was the sole event in determining OS. The observation time was censored at the last follow-up date if no event was recorded. Follow-up was updated on December 31, 2007. The median follow-up was 5 years (range, 0-9 years). The log-rank test was used for univariate comparisons. The outcome of patients who did or did not undergo HSCT in CR1 was compared adjusting for waiting time to transplantation, both in the Cox model (using a time-dependent treatment indicator) and in the estimation of the survival curves (with a landmark at 5 months and delayed entry for patients transplanted after the landmark). A term for the interaction between time from CR1 and treatment was introduced to account for a nonproportional hazard in the overall analysis. The interaction between treatment and risk group was tested before performing a separate subgroup analysis based on the Cox model. The likelihood ratio test was used, and all tests were 2-sided (P ≤ .05 referred to as statistically significant).

Of the 376 evaluable patients, 297 (79%) had an MLL-rearranged leukemia and are described in Table 1 according to risk group stratification. Events and final outcome are shown in Table 2. A total of 37 of 297 (12%) MLL+ patients had HSCT in CR1 at a median time of 5 months from CR1 (range, 2-9 months). Twenty-one of 37 (57%) patients remained in CCR, compared with 91 of 240 (38%) patients treated with chemotherapy only. Notably, 10 of 37 (27%) underwent HSCT from a matched sibling donor, 19 of 37 (51%) from a matched unrelated donor, 5 of 37 (14%) from a mismatched unrelated donor, and in 3 of 37 (8%) cases type of stem cell donor was unknown. Stem cell sources were bone marrow (n = 22), peripheral blood stem cells (n = 3), cord blood (n = 9), and not available (n = 3). In 6 of 37 MLL+ patients, HSCT was performed despite a GPR, which was not an indication for this treatment procedure in the trial (WBC counts ≥ 300 g/L, n = 4; < 6 months of age, n = 3).

Table 1

Univariate analysis on prognostic factors in 297 MLL+ ALL patients according to risk group stratification

Medium riskHigh riskMedium risk + high riskEvents, no. (%)4-year EFS (SE)P*
Overall 200 97 297 185 36.8 (2.9)  
Age at diagnosis      < .001 
    < 3 mo 37 46 83 62 (75) 24.4 (5.5)  
    ≥ 3-6 mo or older 44 51 95 65 (68) 30.5 (5.1)  
    ≥ 6-9 mo or older 70 70 39 (56) 42.6 (6.1)  
    ≥ 9-12 mo or older 49 49 19 (39) 61.9 (7.1)  
WBC counts      < .001 
    < 100 g/L 97 8 105 51 (49) 49.9 (5.0)  
    ≥ 100-300 g/L 74 16 90 57 (63) 36.9 (5.3)  
    ≥ 300 g/L 29 73 102 77 (75) 23.2 (5.0)  
Prednisone response      < .001 
    GPR 169 31 200 111 (56) 43.2 (3.6)  
    PPR 31 66 97 74 (76) 23.5 (5.1)  
Immunophenotype      .51 
    B-cell, CD10 144 81 225 138 (61) 38.0 (3.3)  
    B-cell, CD10+ 37 10 47 33 (70) 27.8 (6.8)  
    B-cell, CD10 (unknown) 16 22 13 (59) 40.9 (10.6)  
    T-cell 1 (50) NA  
    Biphenotypic NA  
Medium riskHigh riskMedium risk + high riskEvents, no. (%)4-year EFS (SE)P*
Overall 200 97 297 185 36.8 (2.9)  
Age at diagnosis      < .001 
    < 3 mo 37 46 83 62 (75) 24.4 (5.5)  
    ≥ 3-6 mo or older 44 51 95 65 (68) 30.5 (5.1)  
    ≥ 6-9 mo or older 70 70 39 (56) 42.6 (6.1)  
    ≥ 9-12 mo or older 49 49 19 (39) 61.9 (7.1)  
WBC counts      < .001 
    < 100 g/L 97 8 105 51 (49) 49.9 (5.0)  
    ≥ 100-300 g/L 74 16 90 57 (63) 36.9 (5.3)  
    ≥ 300 g/L 29 73 102 77 (75) 23.2 (5.0)  
Prednisone response      < .001 
    GPR 169 31 200 111 (56) 43.2 (3.6)  
    PPR 31 66 97 74 (76) 23.5 (5.1)  
Immunophenotype      .51 
    B-cell, CD10 144 81 225 138 (61) 38.0 (3.3)  
    B-cell, CD10+ 37 10 47 33 (70) 27.8 (6.8)  
    B-cell, CD10 (unknown) 16 22 13 (59) 40.9 (10.6)  
    T-cell 1 (50) NA  
    Biphenotypic NA  

ALL indicates acute lymphoblastic leukemia; EFS, event-free survival; SE, standard error; WBC, white blood cell; GPR, good prednisone response; PPR, poor prednisone response; and NA, not applicable.

*

P values are based on log-rank test.

These patients had a PPR.

In B-cell only.

Table 2

Outcome of 297 MLL+ ALL patients according to the risk group stratification

Medium risk
High risk
ChemoHSCTChemoHSCT
No. of patients 200 (67%) 97 (33%) 
Deaths in induction 
Resistance/deaths 5/3 4/4 
CR1, total 190 (95%) 87 (90%) 
CR1 169 21 71 16 
Relapses 78 (46%) 8 (38%) 55 (77%) 6 (38%) 
    < 5 months from CR1/deaths 19/15 2/2 24/23 0/0 
    ≥ 5 months from CR1/deaths 59*/46 6/4 31*/28 6/4 
Deaths in CCR 10 (6%) 1 (5%) 6 (8%) 1 (6%) 
    < 5 months from CR1 
    ≥ 5 months from CR1 4 1 
Second malignancy 
CCR 81 (48%) 12 (57%) 10 (14%) 9 (56%) 
4-year EFS (SE) 45.6 (3.6) 18.6 (5.1) 
Medium risk
High risk
ChemoHSCTChemoHSCT
No. of patients 200 (67%) 97 (33%) 
Deaths in induction 
Resistance/deaths 5/3 4/4 
CR1, total 190 (95%) 87 (90%) 
CR1 169 21 71 16 
Relapses 78 (46%) 8 (38%) 55 (77%) 6 (38%) 
    < 5 months from CR1/deaths 19/15 2/2 24/23 0/0 
    ≥ 5 months from CR1/deaths 59*/46 6/4 31*/28 6/4 
Deaths in CCR 10 (6%) 1 (5%) 6 (8%) 1 (6%) 
    < 5 months from CR1 
    ≥ 5 months from CR1 4 1 
Second malignancy 
CCR 81 (48%) 12 (57%) 10 (14%) 9 (56%) 
4-year EFS (SE) 45.6 (3.6) 18.6 (5.1) 

ALL indicates acute lymphoblastic leukemia; HSCT, hematopoietic stem cell transplantation; CR1, first complete remission; Chemo, chemotherapy only; CCR, continuous complete remission; and EFS, event-free survival.

*

Nineteen of 59 relapses in the medium risk group and 13 of 31 relapses in the high risk group occurred within 9 months from CR1 (maximum waiting time to HSCT).

Two of 4 deaths in the medium risk group and none in the high risk group occurred within 9 months from CR1 (maximum waiting time to HSCT).

DFS and OS curves for the MLL+ patients having received chemotherapy only or HSCT in CR1 adjusted for waiting time to transplantation are shown in Figure 1. Results of the Cox regression model (Table 3) reveal that HSCT was associated with a significantly superior DFS (P = .03). The advantage of HSCT over chemotherapy becomes more evident with every successive year of follow-up, suggesting that HSCT provides greater protection against late relapses in children who do not fail initially (from relapse or toxic deaths; Table 3). These estimates, which account for waiting time to transplantation, are similar whether they are adjusted by known prognostic factors in Model I or by the risk stratification adopted in study Interfant-06 (Model II; Table 3). There was a significant interaction between risk group and treatment (P = .03), indicating that HSCT had a different effect in patients classified to the medium-risk or high-risk groups, as described in Figures 2 and 3, respectively. When analyzing DFS for both subgroups separately, the advantage of HSCT over chemotherapy was especially evident in the high-risk group (P = .01) but not in the medium-risk group (P = .09). In the high-risk group, HSCT was associated with a 64% reduction in the risk of failure resulting from relapse or death in CR (hazard ratio = 0.36, 95% confidence interval, 0.15-0.86). In the medium-risk group, there was an initial disadvantage of transplantation, which was overcome after 1 year from CR. Comparable results were found when OS instead of DFS was analyzed in the medium-risk (Figure 2) and high-risk (Figure 3) groups.

Figure 1

DFS and OS of 274 MLL+ infant ALL patients by treatment performed, adjusted by waiting time to HSCT. P value is from Cox Model. CHEMO indicates chemotherapy only; and HSCT, hematopoietic stem cell transplantation.

Figure 1

DFS and OS of 274 MLL+ infant ALL patients by treatment performed, adjusted by waiting time to HSCT. P value is from Cox Model. CHEMO indicates chemotherapy only; and HSCT, hematopoietic stem cell transplantation.

Close modal
Table 3

Effect of HSCT compared with chemotherapy only on outcome adjusted by known prognostic factors (Model I) and by risk stratification (Model II)

Disease-free survival
Overall survival
Hazard ratio95% CIPHazard ratio95% CIP
Model I       
    Chemo — — — — 
    HSCT   .027   .023 
        6 mo 0.96 0.50-1.82 — 0.74 0.28-1.95 — 
        1 y 0.52 0.27-0.99 — 0.51 0.27-0.96 — 
        2 y 0.28 0.10-0.80 — 0.35 0.15-0.84 — 
        3 y 0.19 0.05-0.75 — 0.28 0.09-0.92 — 
    Age at diagnosis       
        < 6 mo 1.89 1.32-2.69 .001 1.86 1.26-2.75 .002 
        ≥ 6-12 mo — — — — 
    WBC counts       
        < 100 g/L — — — — 
        ≥ 100-300 g/L 1.38 0.91-2.09 .127 1.50 0.94-2.38 .088 
        ≥ 300 g/L 1.63 1.06-2.50 .027 2.09 1.29-3.36 .003 
    Immunophenotype       
        B-cell, CD10 — — — — 
        B-cell, CD10+ 1.51 0.98-2.32 .064 1.24 0.76-2.00 .387 
        Other 0.96 0.53-1.72 .885 0.69 0.35-1.38 .295 
    Prednisone response       
        GPR — — — — 
        PPR 1.69 1.14-2.52 .010 1.60 1.05-2.45 .029 
Model II       
    Chemo — — — — 
    HSCT   .032   .014 
        6 mo 0.92 0.50-1.71 — 0.72 0.28-1.86 — 
        1 y 0.53 0.28-0.98 — 0.51 0.28-0.93 — 
        2 y 0.30 0.11-0.84 — 0.36 0.15-0.83 — 
        3 y 0.22 0.06-0.81 — 0.29 0.09-0.92 — 
    Risk group       
        Medium — — — — 
        High 2.56 1.85-3.53 < .001 2.82 2.00-3.98 < .001 
Disease-free survival
Overall survival
Hazard ratio95% CIPHazard ratio95% CIP
Model I       
    Chemo — — — — 
    HSCT   .027   .023 
        6 mo 0.96 0.50-1.82 — 0.74 0.28-1.95 — 
        1 y 0.52 0.27-0.99 — 0.51 0.27-0.96 — 
        2 y 0.28 0.10-0.80 — 0.35 0.15-0.84 — 
        3 y 0.19 0.05-0.75 — 0.28 0.09-0.92 — 
    Age at diagnosis       
        < 6 mo 1.89 1.32-2.69 .001 1.86 1.26-2.75 .002 
        ≥ 6-12 mo — — — — 
    WBC counts       
        < 100 g/L — — — — 
        ≥ 100-300 g/L 1.38 0.91-2.09 .127 1.50 0.94-2.38 .088 
        ≥ 300 g/L 1.63 1.06-2.50 .027 2.09 1.29-3.36 .003 
    Immunophenotype       
        B-cell, CD10 — — — — 
        B-cell, CD10+ 1.51 0.98-2.32 .064 1.24 0.76-2.00 .387 
        Other 0.96 0.53-1.72 .885 0.69 0.35-1.38 .295 
    Prednisone response       
        GPR — — — — 
        PPR 1.69 1.14-2.52 .010 1.60 1.05-2.45 .029 
Model II       
    Chemo — — — — 
    HSCT   .032   .014 
        6 mo 0.92 0.50-1.71 — 0.72 0.28-1.86 — 
        1 y 0.53 0.28-0.98 — 0.51 0.28-0.93 — 
        2 y 0.30 0.11-0.84 — 0.36 0.15-0.83 — 
        3 y 0.22 0.06-0.81 — 0.29 0.09-0.92 — 
    Risk group       
        Medium — — — — 
        High 2.56 1.85-3.53 < .001 2.82 2.00-3.98 < .001 

HSCT indicates hematopoietic stem cell transplantation; CI, confidence interval; WBC, white blood cell; GPR, good prednisone response; PPR, poor prednisone response; Chemo, chemotherapy only; and —, not applicable.

Figure 2

DFS and OS of 188 medium-risk patients with MLL+ infant ALL by treatment performed, adjusted by waiting time to HSCT. P value is from Cox Model. CHEMO indicates chemotherapy only; and HSCT, hematopoietic stem cell transplantation.

Figure 2

DFS and OS of 188 medium-risk patients with MLL+ infant ALL by treatment performed, adjusted by waiting time to HSCT. P value is from Cox Model. CHEMO indicates chemotherapy only; and HSCT, hematopoietic stem cell transplantation.

Close modal
Figure 3

DFS and OS of 97 high-risk patients with MLL+ infant ALL by treatment performed, adjusted by waiting time to HSCT. P value is from Cox Model. CHEMO indicates chemotherapy only; and HSCT, hematopoietic stem cell transplantation.

Figure 3

DFS and OS of 97 high-risk patients with MLL+ infant ALL by treatment performed, adjusted by waiting time to HSCT. P value is from Cox Model. CHEMO indicates chemotherapy only; and HSCT, hematopoietic stem cell transplantation.

Close modal

One of the principal aims of the multinational trial Interfant-99 was to establish robust and independent prognostic markers in infant ALL to implement a new risk stratification system for the subsequent study, Interfant-06.6 MLL status, age, and WBC counts at initial diagnosis and response to steroids at day 8 appeared to have independent prognostic value.6  Of note, there was no difference in outcome with respect to the various translocation partners of the MLL gene. The overall 5-year EFS rate was 46.1% (SE 2.3%), and survival was 55.2% (SE 2.4%) for the whole study cohort of 483 patients. Of the 376 of 483 patients (78%) with evaluable risk factors, 297 (79%) were MLL+. Five-year EFS was 36.3% (SE 2.9%) for the 297 MLL+ patients and only 18.6% (SE 5.1%) for the subgroup of 97 patients with additional poor prognostic factors (ie, age < 6 months and either WBC counts ≥ 300 g/L or poor response to steroids at day 8).6  Consequently, this patient subset was defined as the high-risk group of the new trial Interfant-06 and, more importantly, as the target group for allogeneic HSCT in CR1.

Our present results on a large cohort of infants with MLL+ ALL actually demonstrated that only such high-risk patients (one-third of all MLL+ cases) appeared to benefit from allogeneic HSCT in CR1. Thus, the analysis may have helped to define a possible role for HSCT in the setting of infant ALL. Of note, there was a high early failure rate, with one-third of the high-risk patients having an event before the median time to transplantation, suggesting that early disease control in trial Interfant-99 was insufficient. Therefore, we are also attempting to improve remission quality in the Interfant-06 trial by an early randomized treatment intensification being delivered immediately after completion of induction therapy.

The main limitation of the present study is that the HSCT versus chemotherapy comparison was not supported by a donor-no donor design (the so-called “genetic randomization”).16,17  Thus, patients for whom a transplantation was planned but who relapsed before can only be counted in the chemotherapy group. The statistical analyses attempted to limit the related biases by adjusting for waiting time to transplantation and also by accounting for prognostic characteristics in a regression model.

Conflicting findings in several, mostly retrospective, studies dealing with the role of HSCT in infant (MLL+) ALL have suggested either no clinical advantage at all or a benefit at relatively low EFS rates.12,18-20  These (mostly single-center) studies had low patient numbers, and it was often unclear whether their results were truly representative of the whole population of patients with infant (MLL+) ALL or not.18-20  In addition, even some large retrospective multi-institutional series in infant MLL+ ALL saw no clear advantage for HSCT in CR1 compared with chemotherapy only.11,12 

In a recent report from the Japan Infant Leukemia Group (MLL96 and MLL98 trials), the efficacy of an MLL-based stratifying treatment strategy involving HSCT for all infants with MLL+ ALL was evaluated.10,21  The 5-year EFS rate was only 38.6% for MLL+ ALL, which, however, was an improvement compared with historical controls. Notably, of 80 MLL+ infant cases, 27 of 49 events occurred before HSCT (mostly early bone marrow relapses); and of 22 of 49 events that occurred after HSCT, 8 were the result of toxic deaths. This report suggests that half of the events could not have been prevented by HSCT because they occurred too early for this final treatment procedure to be performed at all. In addition, the toxic death rate after HSCT was considerably high, so both findings suggest that further stratification within the MLL+ group is needed before the full potential of HSCT can be realized.

However, the results of another study using TBI during the conditioning regimen showed that, of 14 infants with MLL+ ALL transplanted in CR1, 11 (78.5%) are surviving in CCR.19  Moreover, the authors19  reported that outcome was associated with only moderate late-term effects, including growth and other neuroendocrine deficiencies, which were not different from that in older children given TBI and HSCT. Nevertheless, as the report did not specify the number of infant ALL patients with initial treatment failure and early relapses who accordingly failed before the time point of potential HSCT, studies of this type are not actually representative but encouraging to pursue HSCT for this group of poor prognostic infant ALL patients. This trial's caveat is also evident in another report using TBI within the conditioning regimen and not reporting either on the nontransplanted subgroup or on long-term side effects.18  Not surprisingly, most published studies available also indicated that disease status (CR1 vs CR2 or relapse) at HSCT and a short interval (without previous complications) between CR1 and HSCT were associated with a better prognosis.19-22 

Recently, Dreyer et al23  reported on data from infant ALL trials of the Children's Cancer Group (CCG 1953) and Pediatric Oncology Group (POG 9407) that used a similar chemotherapy backbone and advised allogeneic HSCT to be performed in MLL+ cases. The 5-year EFS rates were 50.9% in the transplantation group and 48.7% in the chemotherapy only group, suggesting no clinical benefit for HSCT in CR1 for infants with MLL+ ALL. In addition, the United Kingdom infant ALL trial 92 found no difference in EFS comparing 11q23-positive patients who received HSCT (n = 12) and chemotherapy only.24  Moreover, a large retrospective collaborative study (1983-1995) that showed a marked clinical heterogeneity among MLL+ ALL requiring distinct treatment approaches for different age groups and different 11q23/MLL translocations also reported no significant benefit of HSCT in infant MLL+ ALL.12  A more recent update of this report still suggested that HSCT did not improve prognosis for t(4;11)-positive infant ALL.11  However, as the former large analysis might have been biased by patients' selection, the heterogeneous composition and intensity of the treatment approaches applied about more than 10 years earlier, the possible heterogeneous subtype distributions, and differing follow-up times, it cannot serve as the cornerstone on which the role of HSCT in infant MLL+ ALL can be founded.

A major concern in transplantation of infants is the risk of late effects because of the conditioning regimen before and occurrence and treatment of chronic GVHD after HSCT.8-10,25  There are no data available yet for the patients analyzed in the present study because of the relatively short observation time. However, late sequelae are reported to be common in transplanted long-term survivors of infant leukemia.8-10,25  Growth hormone deficiency, hypothyroidism, abnormal pubertal development, cardiac and hearing abnormalities, learning and academic difficulties, neuropsychologic deficiencies, osteopenia and osteoporosis, cataracts, hypertension, hyperinsulinemia, and dyslipidemias have all been reported to occur in young survivors after HSCT and have mostly been attributed to irradiation.8-10,25  In comparison, data on long-term side effects of transplantations not using TBI for infant ALL are scarce so that no definite conclusions can be drawn to date, although the European Bone Marrow Transplantation and international Berlin-Frankfurt-Münster study groups recommended a non–TBI-containing preparative regimen in the Interfant-99 trial.

Recently, we have shown that, in the Interfant-99 study, cases with high minimal residual disease (MRD) levels, as assessed at the end of consolidation therapy by immunoglobulin/T-cell receptor and/or MLL gene rearrangements, had an inferior DFS rate.26,27  The most striking finding was that all patients (n = 18) with MRD levels more than or equal to 10−4 after consolidation therapy relapsed, including 8 patients who would have been classified as medium risk in the Interfant-06 study and thus would have had no indication for allogeneic HSCT. Currently, the role of HSCT in improving the outcome for this patient subset awaits future studies. In addition, it is also unknown whether intensification of chemotherapy or innovative therapies (ie, FLT3 inhibitors, demethylating agents), either in conjunction with or instead of HSCT, will be able to permanently eradicate the disease.28-31  Nevertheless, given the extremely poor prognosis with standard chemotherapy only, HSCT in CR1 appears to be a possible treatment strategy for this intermediate risk group of infant MLL+ ALL with high end-consolidation MRD levels.

In conclusion, our present analysis of a large cohort of unselected infants with MLL+ ALL included in the international Interfant-99 study demonstrated that the presence of an MLL rearrangement itself is not sufficient to warrant HSCT in CR1. However, because of the superior outcome of HSCT in CR1 compared with chemotherapy only in infant MLL+ ALL carrying further poor prognostic factors (ie, age < 6 months and either WBC counts ≥ 300 g/L at diagnosis or poor response to steroids at day 8), HSCT seems to be a valuable and recommendable option for this high-risk group of infants with ALL.

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 all participating institutions, physicians, and nurses for their support of the study.

The Associazione Italiana Ematologia Oncologia Pediatrica was supported by the Associazione Italiana Ricerca sul Cancro (Regional Grant Code 1105; A.B., M.G.V.) and the international trial center by the Fondazione Tettamanti and Comitato ML Verga (M.G.V., P.D.L.). The Berlin-Frankfurt-Münster Study Group-Austria was supported by the St Anna Kinderkrebsforschung, Berlin-Frankfurt-Münster Study Group-Germany by the Deutsche Krebshilfe, the Czech Pediatric Haematology by the Czech Ministry of Education (grant MSM0021620813), and the Polish Pediatric Leukemia and Lymphoma Study Group (grant 2 P054 095 30) by the Polish Ministry of Science and Higher Education (T.S.).

Contribution: R.P., M.S., G.M., and M.G.V. designed and planned the study; G.M., A.A., M.G.V., P.D.L., and R.P. wrote the manuscript; P.D.L. was in charge of data pooling and checking and statistical analysis; M.G.V. revised the data and also performed statistical analyses; C.P., I.H., G.D.R., M.F., B.L., T.L., T.S., A.F., G.J.-S., J.R., L.B.S., J.S., M.C., C.K.L., R.S., A.B., A.V., R.P., M.S., G.M., and A.A. were principal or coinvestigators in their study groups and institutions, coordinated the study in their countries, and recruited patients; and all authors read and approved the final version of the manuscript.

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

Correspondence: Georg Mann, St Anna Children's Hospital, Department of Pediatric Oncology and Hematology, Kinderspitalgasse 6, 1090 Vienna, Austria; e-mail: georg.mann@stanna.at; and Rob Pieters, Erasmus MC-Sophia Children's Hospital, Department of Pediatric Oncology and Hematology, Dr Molewaterplein 60, 3015GJ Rotterdam, The Netherlands; e-mail: rob.pieters@erasmusmc.nl.

1
Biondi
 
A
Cimino
 
G
Pieters
 
R
Pui
 
CH
Biological and therapeutic aspects of infant leukemia.
Blood
2000
, vol. 
96
 
1
(pg. 
24
-
33
)
2
Borkhardt
 
A
Wuchter
 
C
Viehmann
 
S
, et al. 
Infant acute lymphoblastic leukemia: combined cytogenetic, immunophenotypical and molecular analysis of 77 cases.
Leukemia
2002
, vol. 
16
 
9
(pg. 
1685
-
1690
)
3
Mann
 
G
Cazzaniga
 
G
van der Velden
 
VH
, et al. 
Acute lymphoblastic leukemia with t(4;11) in children 1 year and older: the ‘big sister’ of the infant disease?
Leukemia
2007
, vol. 
21
 
4
(pg. 
642
-
646
)
4
Dördelmann
 
M
Reiter
 
A
Borkhardt
 
A
, et al. 
Prednisone response is the strongest predictor of treatment outcome in infant acute lymphoblastic leukemia.
Blood
1999
, vol. 
94
 
4
(pg. 
1209
-
1217
)
5
Silverman
 
LB
Acute lymphoblastic leukemia in infancy.
Pediatr Blood Cancer
2007
, vol. 
49
 
7 suppl
(pg. 
1070
-
1073
)
6
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 randomized trial.
Lancet
2007
, vol. 
370
 
9583
(pg. 
240
-
250
)
7
Meyer
 
C
Schneider
 
B
Jakob
 
S
, et al. 
The MLL recombinome of acute leukemias.
Leukemia
2006
, vol. 
20
 
5
(pg. 
777
-
784
)
8
Hunger
 
SP
Loh
 
KM
Baker
 
KS
Schultz
 
KR
Controversies of and unique issues in hematopoietic cell transplantation for infant leukemia.
Biol Blood Marrow Transplant
2009
, vol. 
15
 
1 suppl
(pg. 
79
-
83
)
9
Leung
 
W
Hudson
 
M
Zhu
 
Y
, et al. 
Late effects in survivors of infant leukemia.
Leukemia
2000
, vol. 
14
 
7
(pg. 
1185
-
1190
)
10
Tomizawa
 
D
Koh
 
K
Sato
 
T
, et al. 
Outcome of risk-based therapy for infant acute lymphoblastic leukemia with or without an MLL gene rearrangement, with emphasis on late effects: a final report of two consecutive studies, MLL96 and MLL98, of the Japan Infant Leukemia Study Group.
Leukemia
2007
, vol. 
21
 
11
(pg. 
2258
-
2263
)
11
Pui
 
CH
Chessells
 
JM
Camitta
 
B
, et al. 
Clinical heterogeneity in childhood acute lymphoblastic leukemia with 11q23 rearrangements.
Leukemia
2003
, vol. 
17
 
4
(pg. 
700
-
706
)
12
Pui
 
CH
Gaynon
 
PS
Boyett
 
JM
, et al. 
Outcome of treatment in childhood acute lymphoblastic leukaemia with rearrangements of the 11q23 chromosomal region.
Lancet
2002
, vol. 
359
 
9321
(pg. 
1909
-
1915
)
13
van der Does-van den Berg
 
A
Bartram
 
CR
Basso
 
G
, et al. 
Minimal requirements for the diagnosis, classification, and evaluation of the treatment of childhood acute lymphoblastic leukemia (ALL) in the ”BFM Family” Cooperative Group.
Med Pediatr Oncol
1992
, vol. 
20
 
6
(pg. 
497
-
505
)
14
Attarbaschi
 
A
Mann
 
G
König
 
M
, et al. 
Mixed lineage leukemia-rearranged childhood pro-B and CD10-negative pre-B acute lymphoblastic leukemia constitute a distinct clinical entity.
Clin Cancer Res
2006
, vol. 
12
 
10
(pg. 
2988
-
2994
)
15
De Lorenzo
 
P
Antolini
 
L
Valsecchi
 
MG
Evaluation of alternative prognostic stratifications by prediction accuracy measures on individual survival with application to childhood leukaemia.
Eur J Cancer
2009
, vol. 
45
 
8
(pg. 
1432
-
1437
)
16
Balduzzi
 
A
Valsecchi
 
MG
Uderzo
 
C
, et al. 
Chemotherapy versus allogeneic transplantation for very-high-risk childhood acute lymphoblastic leukaemia in first complete remission: comparison by genetic randomization in an international prospective study.
Lancet
2005
, vol. 
366
 
9486
(pg. 
635
-
642
)
17
Schrauder
 
A
Reiter
 
A
Gadner
 
H
, et al. 
Superiority of allogeneic hematopoietic stem-cell transplantation compared with chemotherapy alone in high-risk childhood T-cell acute lymphoblastic leukemia: results from ALL-BFM 90 and 95.
J Clin Oncol
2006
, vol. 
24
 
36
(pg. 
5742
-
5749
)
18
Jacobsohn
 
DA
Hewlett
 
B
Morgan
 
E
Tse
 
W
Duerst
 
RE
Kletzel
 
M
Favorable outcome for infant acute lymphoblastic leukemia after hematopoietic stem cell transplantation.
Biol Blood Marrow Transplant
2005
, vol. 
11
 
12
(pg. 
999
-
1005
)
19
Sanders
 
JE
Im
 
HJ
Hoffmeister
 
PA
, et al. 
Allogeneic hematopoietic cell transplantation for infants with acute lymphoblastic leukemia.
Blood
2005
, vol. 
105
 
9
(pg. 
3749
-
3756
)
20
Marco
 
F
Bureo
 
E
Ortega
 
JJ
, et al. 
High survival rate in infant acute leukemia treated with early high-dose chemotherapy and stem-cell support: Groupo Espanol de Trasplante de Medula Osea en Ninos.
J Clin Oncol
2000
, vol. 
18
 
18
(pg. 
3256
-
3261
)
21
Kosaka
 
Y
Koh
 
K
Kinukawa
 
N
, et al. 
Infant acute lymphoblastic leukemia with MLL gene rearrangements: outcome following intensive chemotherapy and hematopoietic stem cell transplantation.
Blood
2004
, vol. 
104
 
12
(pg. 
3527
-
3534
)
22
Eapen
 
M
Rubinstein
 
P
Zhang
 
MJ
, et al. 
Comparable long-term survival after unrelated and HLA-matched sibling donor hematopoietic stem cell transplantations for acute leukemia in children younger than 18 months.
J Clin Oncol
2006
, vol. 
24
 
1
(pg. 
145
-
151
)
23
Dreyer
 
ZE
Dinndorf
 
P
Sather
 
H
, et al. 
Hematopoietic stem cell transplant (HSCT) versus intensive chemotherapy in infant acute lymphoblastic leukemia [abstract].
J Clin Oncol
2007
, vol. 
25
 pg. 
9514
  
suppl ASCO Meeting Abstracts
24
Chessells
 
JM
Harrison
 
CJ
Watson
 
SL
Vora
 
AJ
Richards
 
SM
Treatment of infants with lymphoblastic leukaemia: results of the UK Infant Protocols 1987-1999.
Br J Haematol
2002
, vol. 
117
 
2
(pg. 
306
-
314
)
25
Perkins
 
JL
Kunin-Batson
 
AS
Youngren
 
NM
, et al. 
Long-term follow-up of children who underwent hematopoeitic cell transplant (HCT) for AML or ALL at less than 3 years of age.
Pediatr Blood Cancer
2007
, vol. 
49
 
7
(pg. 
958
-
963
)
26
Van der Velden
 
VH
Corral
 
L
Valsecchi
 
MG
, et al. 
Prognostic significance of minimal residual disease in infants with acute lymphoblastic leukemia treated within the Interfant-99 protocol.
Leukemia
2009
, vol. 
23
 
6
(pg. 
1073
-
1079
)
27
van der Velden
 
VH
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
)
28
Stam
 
RW
den Boer
 
ML
Schneider
 
P
, et al. 
Targeting FLT3 in primary MLL-gene-rearranged infant acute lymphoblastic leukemia.
Blood
2005
, vol. 
106
 
7
(pg. 
2484
-
2490
)
29
Kreyenberg
 
H
Eckert
 
C
Yarkin
 
Y
, et al. 
Immunoglobulin and T-cell receptor gene rearrangements as PCR-based targets are stable markers for monitoring minimal residual disease in acute lymphoblastic leukemia after stem cell transplantation.
Leukemia
2009
, vol. 
23
 
7
(pg. 
1355
-
1358
)
30
Pulsipher
 
MA
Bader
 
P
Klingebiel
 
T
Cooper
 
LJ
Allogeneic transplantation for pediatric acute lymphoblastic leukemia: the emerging role of peritransplantation minimal residual disease/chimerism monitoring and novel chemotherapeutic, molecular, and immune approaches aimed at preventing relapse.
Biol Blood Marrow Transplant
2009
, vol. 
15
 
1 suppl
(pg. 
62
-
71
)
31
Stumpel
 
DJ
Schneider
 
P
van Roon
 
EH
, et al. 
Specific promoter methylation identifies different subgroups of MLL-rearranged infant acute lymphoblastic leukemia, influences clinical outcome, and provides therapeutic options.
Blood
2009
, vol. 
114
 
27
(pg. 
5490
-
5498
)

Author notes

*

G.M. and A.A. contributed equally to this study.

Sign in via your Institution