• Risk-adapted therapy and broad use of HSCT resulted in a significant improvement in outcome.

  • AUTO- or ALLO-HSCT in high-risk patients resulted in a cumulative incidence of leukemia relapse superimposable to that of SR.

We evaluated the outcome of 482 children with acute myeloid leukemia (AML) enrolled in the Associazione Italiana di Ematologia e Oncologia Pediatrica AML 2002/01 trial. Treatment was stratified according to risk group; hematopoietic stem cell transplantation (HSCT) was used in high-risk (HR) children. Patients with core binding factor leukemia achieving complete remission (CR) after the first induction course were considered standard risk (SR; 99 patients), whereas the others (n = 383) were assigned to the HR group. Allogeneic (ALLO) or autologous (AUTO) HSCT was employed, respectively, in 141 and 102 HR patients after consolidation therapy. CR, early death, and induction failure rates were 87%, 3%, and 10%, respectively. Relapse occurred in 24% of patients achieving CR. The 8-year overall survival (OS), event-free survival (EFS), and disease-free survival (DFS) were 68%, 55%, and 63%, respectively. OS, EFS, and DFS for SR and HR patients were 83%, 63%, and 66% and 64%, 53%, and 62%. DFS was 63% and 73% for HR patients given AUTO-HSCT and ALLO-HSCT, respectively. In multivariate analysis, risk group, white blood cell >100 × 109/L at diagnosis, and monosomal karyotype predicted poorer EFS. Risk-oriented treatment and broad use of HSCT result in a long-term EFS comparing favorably with previously published studies on childhood AML.

The prognosis of childhood acute myeloid leukemia (AML) has significantly improved over the last 2 decades.1,2  In particular, the most recent and successful reports showed a probability of event-free survival (EFS) ranging between 40% and 60%.3-11  This improvement was largely due to (1) significant progress in stratification of patients, with a consequent risk-directed therapy; (2) optimization in induction and postremission treatment strategy, including the use of repeated courses of high-dose cytarabine (HD Ara-C); (3) better supportive therapy; and (4) broad use of allogeneic (ALLO) hematopoietic stem cell transplantation (HSCT) in high-risk (HR) patients.3  In this regard, in the past, randomized trials showed that ALLO-HSCT from an HLA-identical sibling is the postremission treatment able to offer the best chance of sustained remission.12-14  The efficacy of ALLO-HSCT from an HLA-matched sibling has been recently confirmed in the subsets of patients with intermediate/HR features.15  Despite these data, in the last years, and in view of an improved probability for relapsed patients to be rescued by salvage therapy, the use of ALLO-HSCT in children with AML in first complete remission (CR1) has been questioned by some groups.16,17  The role of autologous (AUTO) HSCT as postremission therapy for AML is even more controversial, and some studies have suggested an advantage over chemotherapy in terms of prevention of leukemia relapse,18  which, after the introduction of HD Ara-C in consolidation therapy, has not been confirmed12-14  or was offset by an increased risk of transplantation-related mortality (TRM).19 

Since December 2002, children without Down syndrome and with newly diagnosed, de novo AML other than promyelocytic leukemia (APL) were treated at Italian centers affiliated with Associazione Italiana di Ematologia e Oncologia Pediatrica (AIEOP) according to the AML 2002/01 protocol, the latest of 5 consecutive studies on childhood AML (LAM-82, LAM-87, LAM-87M, LAM-92, and AML 2002/01) conducted in Italy since 1982.4  The primary objective chosen when the study was designed was that of obtaining a 5-year EFS >50%. In order to reach this goal, in view of the evidence available at that time on factors influencing outcome, we stratified children according to cytogenetic/molecular findings and response to the first course of induction therapy. In particular, assigned to the standard risk (SR) treatment were children with either AML1-ETO fusion transcript or anomalies of core binding factor β (CBF-β) and in morphologic CR after the first of 2 induction courses. In all patients, consolidation therapy included courses containing HD Ara-C. It was also decided that patients allocated to the HR group were broadly offered either AUTO- or ALLO-HSCT, depending on the availability of an HLA-identical sibling.

In this report, we describe the results obtained in children enrolled in the AML 2002/01 protocol.

Eligibility

The entry criteria for the AIEOP AML 2002/01 study included (1) newly diagnosed de novo AML other than APL; (2) age ranging from 0 to 18 years; and (3) written informed consent from parents or legal guardians in accordance with the Declaration of Helsinki. Other eligibility criteria included serum bilirubin ≤3× upper limit of normal (ULN) for age, aspartate aminotransferase and alanine aminotransferase ≤5× ULN, and serum creatinine ≤2× ULN for age. Patients with a previous myelodysplastic phase or who had received previous treatment with either cytotoxic agents or steroids during the 2 weeks preceding diagnosis were excluded. The study was approved by the local ethics committees of each participating institution.

Diagnostic procedure

The initial diagnosis of AML with definition of subtype was established according to the French-American-British (FAB) and World Health Organization classification criteria. Bone marrow (BM) smears obtained at time of diagnosis were centrally reviewed at the Laboratory of Pediatric Hematology in Padua. All samples at diagnosis were analyzed for the presence of t(8;21), inv(16), t(16;16), t(15;17), and t(11q23)/mixed-lineage leukemia (MLL) and the related molecular transcripts, namely AML-ETO1, CBF-β abnormalities, promyelocytic leukemia-retinoic acid receptor α, and MLL rearrangements. Molecular analyses for internal tandem duplication of FLT-3 mutation (FLT3-ITD) and activating loop mutations of the same gene were also performed. The diagnosis of both FAB M0 and M7 subtypes was always confirmed by immunophenotype.

Morphologic CR status and diagnosis of relapse were centrally reviewed.

Definitions

CR was defined as <5% morphologically evident leukemic blasts in BM with normal hematopoiesis, no leukemia cells in peripheral blood or anywhere else, and signs of normal blood cell production (platelets >50 × 109/L without support, neutrophils > 1.0 × 109/L) after induction phase. Early death (ED) was defined as a fatal event occurring within the first 6 weeks from diagnosis.

Patients who did not achieve CR and who survived after the first course of treatment were divided in 2 groups: (1) partial responders (PR) were patients with percentage of BM leukemia blasts between 5% and 25% at the end of the first course of induction therapy; and (2) nonresponders (NR) were patients with percentage of BM leukemia blasts >25% at the end of the first course of induction therapy or patients with BM leukemia blasts >5% after the second induction course.

Central nervous system (CNS) involvement was defined as >5 leukocytes per μL of CSF and presence of leukemia cells on cytospin preparations or cranial nerve involvement.

Treatment design

Details on treatment are reported in Figure 1 and Table 1. Patients enrolled in the AIEOP AML 2002/01 study were assigned to either the SR or HR group. As mentioned above, patients belonging to the former group had isolated anomalies of CBF-β and were in morphologic CR after the first induction course; the remaining children were assigned to the HR group. All patients, irrespective of the risk group, were given 2 courses of induction chemotherapy, including idarubicin, cytarabine, and etoposide (ICE; see Table 1 for details). BM evaluation to document response to the first induction course was on day +21. The second induction course for patients achieving CR was scheduled to start at time of hematologic recovery and preferably no later than 28 days after the beginning of the first course, in the absence of complications precluding cytotoxic treatment. PR and NR patients started either the second cycle of induction or salvage therapy, respectively, at time of evidence of leukemia persistence. Children in CR after the second induction course received 2 consolidation courses, including HD Ara-C, combined with etoposide in the first course (AVE; Table 1) and mitoxantrone in the second course (HAM; Table 1). At the end of this treatment, SR patients still in CR1 were given a further course of HD Ara-C (Table 1), while HR patients had the indication to receive either ALLO- or AUTO-HSCT, depending on the availability of an HLA-identical family donor. Among HR patients, those <1 year of age, with AML-M7, or not in CR at the end of first ICE, or with a complex karyotype were considered at particularly HR of recurrence and, thus, eligible to be transplanted from unrelated donors (UD). After 2006, all patients with FLT3-ITD in CR1 were also offered an UD allograft. NR patients were eligible for salvage therapy.

Figure 1

Schematic representation of the induction and consolidation courses employed for treating patients enrolled in the AIEOP AML 2002/01 trial. AVE, HD Ara-C and etoposide; CONS, consolidation; HAM, HD Ara-C and mitoxantrone; MFD, matched family donor.

Figure 1

Schematic representation of the induction and consolidation courses employed for treating patients enrolled in the AIEOP AML 2002/01 trial. AVE, HD Ara-C and etoposide; CONS, consolidation; HAM, HD Ara-C and mitoxantrone; MFD, matched family donor.

Close modal
Table 1

AIOP LAM 2002 chemotherapy schedule

PhaseWeekAdministration routeDosesDays
Remission induction (ICE ×2)     
 Idarubicin 1, 4 IV 10 mg/m2 1, 2, 3 
 Etoposide 1, 4 IV 100 mg/m2 1, 2, 3, 4, 5 
 Cytarabine 1, 4 IV (24 h) 200 mg/m2 1, 2, 3, 4, 5, 6, 7 
IT therapy 1, 4    
  Cytarabine 1, 4 IT Depending on age* 
  Methotrexate (only for CNS disease) 1, 4 IT Depending on age* 1, 4, 7, 10, 14 of first cycle 
  Methylprednisolone (only for CNS disease) 1, 4 IT Depending on age* 1, 4, 7, 10, 14 of first cycle 
Consolidation 1 (AVE)     
 Etoposide IV 125 mg/m2 2, 3, 4, 5 
 Cytarabine IV 3 g/m2 bid 1, 2, 3 
IT therapy     
  Cytarabine IT Depending on age* 
Consolidation 2 (HAM)     
 Cytarabine 11 IV 3 g/m2 bid 1, 2, 3 
 Mitoxantrone 11 IV 10 mg/m2 3, 4 
IT therapy     
  Cytarabine 11 IT Depending on age* 
Consolidation 3 (HD Ara-C) (only for SR)     
 Cytarabine 14 IV 3 g/m2 bid 1, 2, 3 
IT therapy     
  Cytarabine 14 IT Depending on age* 
PhaseWeekAdministration routeDosesDays
Remission induction (ICE ×2)     
 Idarubicin 1, 4 IV 10 mg/m2 1, 2, 3 
 Etoposide 1, 4 IV 100 mg/m2 1, 2, 3, 4, 5 
 Cytarabine 1, 4 IV (24 h) 200 mg/m2 1, 2, 3, 4, 5, 6, 7 
IT therapy 1, 4    
  Cytarabine 1, 4 IT Depending on age* 
  Methotrexate (only for CNS disease) 1, 4 IT Depending on age* 1, 4, 7, 10, 14 of first cycle 
  Methylprednisolone (only for CNS disease) 1, 4 IT Depending on age* 1, 4, 7, 10, 14 of first cycle 
Consolidation 1 (AVE)     
 Etoposide IV 125 mg/m2 2, 3, 4, 5 
 Cytarabine IV 3 g/m2 bid 1, 2, 3 
IT therapy     
  Cytarabine IT Depending on age* 
Consolidation 2 (HAM)     
 Cytarabine 11 IV 3 g/m2 bid 1, 2, 3 
 Mitoxantrone 11 IV 10 mg/m2 3, 4 
IT therapy     
  Cytarabine 11 IT Depending on age* 
Consolidation 3 (HD Ara-C) (only for SR)     
 Cytarabine 14 IV 3 g/m2 bid 1, 2, 3 
IT therapy     
  Cytarabine 14 IT Depending on age* 

CNS disease was considered as presence of >5 leukemia blasts in the cerebrospinal fluid, or nerve palsy or CNS involvement, at either nuclear magnetic resonance or at computed tomography scan.

AVE, high-dose cytarabine; bid, twice daily; HAM, HD Ara-C and mitoxantrone; IT, intrathecal; IV, intravenous.

*

Children <1 year received 20 mg cytarabine, while those aged between 1 and 2, or 2 and 3, or >3 years were given 30, 50, and 70 mg, respectively.

Conditioning regimen combined busulfan (16 mg/kg), cyclophosphamide (120 mg/kg), and melphalan (L-PAM; 140 mg/m2) for both AUTO- and ALLO-HSCT.20  Busulfan dosage was adjusted based on the pharmacokinetic study performed following the first administration in order to maintain a steady-state concentration between 600 and 900 ng/mL. Patients given AUTO-HSCT were recommended to have in vitro marrow purging with mafosfamide.21 

Statistical analysis

The analysis used June 30, 2012, as the reference date.

Overall survival (OS) was calculated from date of diagnosis to time of death due to any cause or time of last contact. EFS was calculated from date of diagnosis to last follow-up or first event (failure to achieve CR, relapse, second malignancy, or death due to any cause, whichever occurred first). Patients who did not attain CR after 2 induction cycles were considered failures at time of remission evaluation. Disease-free survival (DFS) was calculated from the date of remission for both SR and HR patients reaching CR1 or, for the HR children given transplantation, from date of HSCT to last follow-up or first event (relapse, second malignancy, or death due to any cause, whichever occurred first).

Probabilities of OS, EFS, and DFS were estimated using the Kaplan-Meier method. Cumulative incidence (CI) of relapse and death in continuous CR (CCR) were constructed using the method of Kalbfleisch in order to adjust the analysis for competing risks. Death in remission was treated as a competing event to calculate CI of relapse, while relapse was considered the competing event for death in CCR. The significance of differences among the OS, EFS, and DFS curves was estimated by the log-rank test (Mantel-Cox), while Gray’s test was used to assess differences between CI of relapse and death in CCR. All variables having a P value < .05 in univariate analysis were included in a multivariate analysis on EFS using the Cox proportional regression model. Computations were performed using SAS (SAS Institute, Cary, NC).

Between December 2002 and June 2011, 504 children with de novo AML other than APL were enrolled in the study; 482 of them are evaluable. Twenty-two were excluded due to death before initiation of therapy (n = 3), previous treatment with steroids (n = 7) or with cytotoxic drugs (n = 5), and previous diagnosis of myelodysplastic syndrome (n = 7). Children were diagnosed and treated at 29 centers affiliated with the AIEOP network; the number of patients aged <14 years treated per year was consistent with the expected number of new cases in Italy.22  Each center treated a number of patients ranging from 2 to 69 (median value: 9). Patient characteristics are shown in Table 2, while patient flowchart is depicted in Figure 2.

Table 2

Patient characteristics of the AIEOP LAM 2002/01 protocol

Characteristicn%
Number of evaluable patients 482 100 
Gender   
 Male 262 52 
 Female 220 48 
Age   
 <1 y 63 13 
 1-2 y 52 10 
 2-10 y 181 38 
 >10 y 186 39 
WBC count (×109/L)   
 <10 171 35 
 10-99 236 49 
 >100 75 16 
CNS leukemia (yes) 40 
FAB subtype   
 M0 34 
 M1 88 18 
 M2 91 19 
 M4 40 
 M4 Eo 43 
 M5 117 24 
 M6 
 M7 44 
 Unclassifiable/not known 20 
Cytogenetics   
Patients with available cytogenetic data 418  
Favorable cytogenetics 99 24 
 t(8;21) 72 17 
 t(16;16) 
 inv 16 26 
Translocation t(9;11) 19 
Other 11q23 abnormalities 46 11 
Complex karyotype 27 
Normal 166 39 
Other abnormalities 61 15 
FLT3 aberrations   
Patients tested for FLT3 aberrations 384  
 ITD 42 11 
 Activating loop mutations (D835/I836) 10 
 Wild-type 332 86 
Other molecular aberrations   
Patients tested 244  
 NPM mutations 14 
 CEBPα mutations 18 
Risk groups   
 SR 99 20 
 HR 383 80 
Characteristicn%
Number of evaluable patients 482 100 
Gender   
 Male 262 52 
 Female 220 48 
Age   
 <1 y 63 13 
 1-2 y 52 10 
 2-10 y 181 38 
 >10 y 186 39 
WBC count (×109/L)   
 <10 171 35 
 10-99 236 49 
 >100 75 16 
CNS leukemia (yes) 40 
FAB subtype   
 M0 34 
 M1 88 18 
 M2 91 19 
 M4 40 
 M4 Eo 43 
 M5 117 24 
 M6 
 M7 44 
 Unclassifiable/not known 20 
Cytogenetics   
Patients with available cytogenetic data 418  
Favorable cytogenetics 99 24 
 t(8;21) 72 17 
 t(16;16) 
 inv 16 26 
Translocation t(9;11) 19 
Other 11q23 abnormalities 46 11 
Complex karyotype 27 
Normal 166 39 
Other abnormalities 61 15 
FLT3 aberrations   
Patients tested for FLT3 aberrations 384  
 ITD 42 11 
 Activating loop mutations (D835/I836) 10 
 Wild-type 332 86 
Other molecular aberrations   
Patients tested 244  
 NPM mutations 14 
 CEBPα mutations 18 
Risk groups   
 SR 99 20 
 HR 383 80 

CEBPα, CCAAT/enhancer binding protein; ITD, internal tandem duplication; NPM, nucleophosmin.

Figure 2

Flowchart of patients enrolled in the 2000/01 AML protocol.

Figure 2

Flowchart of patients enrolled in the 2000/01 AML protocol.

Close modal

Overall, 87% of 482 patients achieved CR after induction, the early death and induction failure rates being 3% and 10%, respectively. In detail, 13 children died during induction courses and 48 were NR at the end of induction therapy (see also Figure 2). Among these 48 patients, 16 died due to either disease progression or treatment-related complications, while 32 (67%) subsequently reached CR after salvage therapy.

After the first course of induction therapy, 99 children were assigned to SR (20% of the whole population) as they had anomalies of CBF-β and were in morphological CR (Table 3). Notably, none of the children with CBF-β anomalies were resistant to the first induction course; thus, the CR rate of these children was 100%. Three SR patients died between the first and second ICE course, and thus the number of SR patients at the end of the cycles of induction therapy was 96 (23% of those being in CR after induction therapy). The remaining 325 children (77%) belonged to the HR group (see also Figure 2). The CR rate of children other than those with CBF-β anomalies was 85%. The median time elapsing between the beginning of the first and second course of induction therapy was 28 days (range 21-66 days).

Table 3

Characteristics of SR and HR patients

SR patients n (%)HR patients n (%)P value
Number of evaluable patients 99 (100) 383 (100)  
Gender    
 Male 59 (60) 203 (53) .6 
 Female 40 (40) 180 (47) .6 
Age    
 <1 y 2 (2) 61 (16) .01 
 1-2 y 7 (7) 45 (12) .1 
 2-10 y 43 (43) 138 (36) .5 
 >10 y 47 (47) 139 (36) .3 
WBC count (×109/L)    
 <10 31 (31) 140 (36) .6 
 10-99 57 (58) 179 (47) .3 
 >100 11 (11) 64 (17) .3 
CNS leukemia (yes) 4 (4) 36 (9) .2 
FAB subtype    
 M0 0 (0) 34 (9) <.01 
 M1 5 (5) 83 (22) .02 
 M2 60 (61) 31 (8) <.01 
 M4 10 (10) 30 (8) .8 
 M4 Eo 17 (17) 26 (7) .08 
 M5 2 (2) 115 (30) <.01 
 M6 0 (0) 5 (1) 1.0 
 M7 0 (0) 44 (11) <.01 
 Unclassifiable/not known 5 (5) 15 (4) 1.0 
FLT3 aberrations    
 Patients tested for FLT3 aberrations 73 311  
 ITD 0 (0) 42 (13) <.01 
 ALM (D835/I836) 2 (3) 8 (3) 1.0 
 Wild-type 71 (96) 261 (84) .5 
Other molecular aberrations    
 Patients tested 59 185  
 NPM mutations 0 (0) 14 (8) <.01 
 CEBPα mutations 0 (0) 18 (10) <.01 
Outcome    
 Dead during induction 3 (3) 10 (3) 1.0 
 Dead in CR after induction 4 (4) 33 (7) .5 
 Relapsed 24 (24) 79 (21) .7 
 Salvaged after relapse 17 (17) 13 (3) <.01 
 Dead during/after salvage therapy 3 (3) 16 (4) 
 Dead after relapse 4 (4) 50 (13) .04 
SR patients n (%)HR patients n (%)P value
Number of evaluable patients 99 (100) 383 (100)  
Gender    
 Male 59 (60) 203 (53) .6 
 Female 40 (40) 180 (47) .6 
Age    
 <1 y 2 (2) 61 (16) .01 
 1-2 y 7 (7) 45 (12) .1 
 2-10 y 43 (43) 138 (36) .5 
 >10 y 47 (47) 139 (36) .3 
WBC count (×109/L)    
 <10 31 (31) 140 (36) .6 
 10-99 57 (58) 179 (47) .3 
 >100 11 (11) 64 (17) .3 
CNS leukemia (yes) 4 (4) 36 (9) .2 
FAB subtype    
 M0 0 (0) 34 (9) <.01 
 M1 5 (5) 83 (22) .02 
 M2 60 (61) 31 (8) <.01 
 M4 10 (10) 30 (8) .8 
 M4 Eo 17 (17) 26 (7) .08 
 M5 2 (2) 115 (30) <.01 
 M6 0 (0) 5 (1) 1.0 
 M7 0 (0) 44 (11) <.01 
 Unclassifiable/not known 5 (5) 15 (4) 1.0 
FLT3 aberrations    
 Patients tested for FLT3 aberrations 73 311  
 ITD 0 (0) 42 (13) <.01 
 ALM (D835/I836) 2 (3) 8 (3) 1.0 
 Wild-type 71 (96) 261 (84) .5 
Other molecular aberrations    
 Patients tested 59 185  
 NPM mutations 0 (0) 14 (8) <.01 
 CEBPα mutations 0 (0) 18 (10) <.01 
Outcome    
 Dead during induction 3 (3) 10 (3) 1.0 
 Dead in CR after induction 4 (4) 33 (7) .5 
 Relapsed 24 (24) 79 (21) .7 
 Salvaged after relapse 17 (17) 13 (3) <.01 
 Dead during/after salvage therapy 3 (3) 16 (4) 
 Dead after relapse 4 (4) 50 (13) .04 

ALM, activating loop mutations; CEBPα, CCAAT/enhancer binding protein; ITD, internal tandem duplication; NPM, nucleophosmin; Boldface P values denote statistically significant differences (<.05).

Thirty-seven patients died in CR after induction therapy; 33 and 4 out of these 37 children had been assigned to the HR and SR groups, respectively (P = .07). Of these 37 patients, 16 died after HSCT. The causes of death for the 21 patients who died in CR without or before having received HSCT were infections in 19 cases (6 of which were attributable to an invasive fungal infection) and cerebral hemorrhage in the remaining 2 children. The CI of death in CCR was 10% (standard error [SE] 1.7). Adolescents had a greater risk of death in CCR as compared with children aged <14 years (23% vs 6% in younger patients, P = .02). In detail, among the 14 adolescents who died in CR, 9 experienced fatal infections, 2 veno-occlusive disease after HSCT, 2 chronic graft-versus-host disease (GVHD), and 1 cerebral hemorrhage. Five adolescents died after HSCT.

Relapse occurred in 103 children (24%) who had achieved CR; 24 and 79 relapses occurred in SR and HR children, respectively. The median time between diagnosis and recurrence was 12.4 months (range 1.2-54.5 months). Four SR and 28 HR children relapsed while receiving consolidation therapy (see also Figure 2). Relapse involved BM only in 87 cases, BM and other extramedullary sites in 12 cases, and extramedullary sites in 4 cases only. The CI of relapse was 27% for both SR and HR patients (SE 4.7 and 2.8, respectively). Among the 24 SR patients who relapsed, 7 died of either disease progression (4 children) or toxicity during salvage treatment (3 children), while 17 patients are alive and disease-free in second CR. Of the 79 HR children experiencing leukemia recurrence, 66 died of either disease progression (50 children) or toxicity during salvage treatment (16 children), while 13 patients are alive and disease-free in second CR (see also Table 3).

With a median follow-up of 57 months (range 12-130 months), the 8-year probability of OS, EFS, and DFS for the whole cohort was 68% (SE 2.4), 55% (SE 2.6), and 63% (SE 2.7), respectively (Figure 3). OS, EFS, and DFS were 83% (SE 4.6), 63% (SE 5.6), and 66% (SE 5.7) in the SR group; these values were 64% (SE 2.8), 53% (SE 2.9), and 62% (SE 3.1) in the HR group (Figure 4). Both the probability of OS and EFS were better for SR patients, with P values of .0003 and .02, respectively; by contrast, the probability of DFS did not differ between the 2 groups of risk. All patients with nucleophosmin and CCAAT/enhancer binding protein α were assigned to the HR group; their probability of EFS was 64% and 60%, respectively (see also Table 4).

Figure 3

Eight-year probability of OS and EFS for the whole cohort of 482 children enrolled in the AIEOP AML 2002/01 trial. FUP, follow-up.

Figure 3

Eight-year probability of OS and EFS for the whole cohort of 482 children enrolled in the AIEOP AML 2002/01 trial. FUP, follow-up.

Close modal
Figure 4

Eight-year probability of OS, EFS, DFS, and CI of relapse for SR and HR children enrolled in the AIEOP AML 2002/01 trial. (A) OS. (B) EFS. (C) DFS. (D) CI of REL.

Figure 4

Eight-year probability of OS, EFS, DFS, and CI of relapse for SR and HR children enrolled in the AIEOP AML 2002/01 trial. (A) OS. (B) EFS. (C) DFS. (D) CI of REL.

Close modal
Table 4

Probability of EFS by patient subgroup

Global populationNo. of patientsEFS % (SE)P value
Risk stratification 482 55 (2.6)  
 SR 99 63 (5.6) .021 
 HR 383 53 (2.9) .021 
Age    
 <1 y 63 58 (6.3) .91 
 1-2 y 52 58 (7.8) .91 
 2-10 y 181 55 (4.6) .91 
 >10 y 186 57 (4.0) .91 
WBC count (×109/L)    
 <10 171 60 (4.4) .0001 
 10-99 236 57 (3.7) .0001 
 >100 75 37 (5.7) .0001 
FAB types    
 M0 34 49 (8.9) .12 
 M1 88 44 (6.5) .12 
 M2 91 64 (5.8) .12 
 M4 83 64 (5.7) .12 
 M5 117 54 (5.6) .12 
 M6 100 (/) .12 
 M7 44 56 (7.7) .12 
 Unclassifiable/not known 20 41(14.5) .12 
Subgroups    
 FLT3 Pos overall 52 47 (7,5)  
 FLT3-ITD Pos 42 47 (8.6)  
 FLT3-ALM (D835/I836) Pos 10 47 (6.7)  
 FLT3-ITD Pos vs FLT3-ITD Neg   .22 
 11q23 abnormalities other than t(9;11) 46 52 (10.1)  
 11q23 abnormalities other than t(9;11) vs other   .58 
 t(9;11) 19 61 (11.6)  
 t(9;11) vs other   .78 
 Monosomal karyotype 10 20 (12.6)  
 Monosomal karyotype vs other karyotype   .0002 
 Complex karyotype 27 40 (9.6)  
 Complex karyotype vs other karyotype   .048 
 NPM mutations Pos 14 64 (15.2)  
 NPM Pos vs NPM Neg   .24 
 CEBPα mutations Pos 18 60 (12.9)  
 CEBPα Pos vs Neg   .33 
Global populationNo. of patientsEFS % (SE)P value
Risk stratification 482 55 (2.6)  
 SR 99 63 (5.6) .021 
 HR 383 53 (2.9) .021 
Age    
 <1 y 63 58 (6.3) .91 
 1-2 y 52 58 (7.8) .91 
 2-10 y 181 55 (4.6) .91 
 >10 y 186 57 (4.0) .91 
WBC count (×109/L)    
 <10 171 60 (4.4) .0001 
 10-99 236 57 (3.7) .0001 
 >100 75 37 (5.7) .0001 
FAB types    
 M0 34 49 (8.9) .12 
 M1 88 44 (6.5) .12 
 M2 91 64 (5.8) .12 
 M4 83 64 (5.7) .12 
 M5 117 54 (5.6) .12 
 M6 100 (/) .12 
 M7 44 56 (7.7) .12 
 Unclassifiable/not known 20 41(14.5) .12 
Subgroups    
 FLT3 Pos overall 52 47 (7,5)  
 FLT3-ITD Pos 42 47 (8.6)  
 FLT3-ALM (D835/I836) Pos 10 47 (6.7)  
 FLT3-ITD Pos vs FLT3-ITD Neg   .22 
 11q23 abnormalities other than t(9;11) 46 52 (10.1)  
 11q23 abnormalities other than t(9;11) vs other   .58 
 t(9;11) 19 61 (11.6)  
 t(9;11) vs other   .78 
 Monosomal karyotype 10 20 (12.6)  
 Monosomal karyotype vs other karyotype   .0002 
 Complex karyotype 27 40 (9.6)  
 Complex karyotype vs other karyotype   .048 
 NPM mutations Pos 14 64 (15.2)  
 NPM Pos vs NPM Neg   .24 
 CEBPα mutations Pos 18 60 (12.9)  
 CEBPα Pos vs Neg   .33 

ALM, activating loop mutations; CEBPα, CCAAT/enhancer binding protein; ITD, internal tandem duplication; Neg, negative; NPM, nucleophosmin; Pos, positive; Boldface P values denote statistically significant differences (<.05).

Notably, the 8-year OS of the 48 NR patients was 30% (SE 7.6). Of them, 27 were given an allograft, either in CR or with persistent disease, from an HLA-identical sibling or an alternative donor.

Considering variables potentially influencing outcome (see Table 4 for details), we found that patients with white blood cell (WBC) count at diagnosis >100 × 109/L had a worse outcome as compared with the others, the EFS being 37% (SE 5.7) and 58% (SE 2.8), respectively (P < .05). The 8-year EFS of the 27 patients with complex karyotype (defined as ≥3 abnormalities, either numerical or structural) was 40% (SE 9.6) as compared with 56% (SE 2.6) in the other patients (P < .05). The EFS of patients carrying (n = 10) or not (n = 472) a monosomal karyotype, defined as the presence of isolated −7 and −5, was 20% (SE 12.6) and 53% (SE 2.9), respectively (P = .0002). Noteworthy, FLT3-ITD did not influence the probability of EFS (see also Table 3). In multivariate analysis for EFS, risk group (either SR or HR), WBC count at diagnosis >100 × 109/L, and monosomal karyotype predicted poor patient outcome (with P values of .001, .001, and .049 and hazard ratios of 1.95, 3.37, and 1.52, respectively).

As far as the outcome of the 243 patients given transplantation in CR1 is concerned, the 8-year DFS, calculated from the date of HSCT, for the 102 patients given AUTO-HSCT was 63% (SE 4.9); it was 73% (SE 4.0) for the 141 patients given ALLO-HSCT (P value not significant). Among this latter group, DFS was 71% (SE 6.3) and 79% (SE 6.2) for the 66 and 75 patients transplanted from a relative or an UD, respectively. Sixteen patients died after HSCT; causes of death were acute GVHD (n = 3), chronic GVHD (n = 4), veno-occlusive disease (n = 4), and infection (n = 5). The 8-year CI of TRM after either AUTO- or ALLO-HSCT from an HLA-compatible sibling or an UD was comparable (7% in both cohorts). The 8-year CI of relapse after AUTO- and ALLO-HSCT was 28% (SE 4.5) and 17% (SE 3.3), respectively (P = .043).

Thirty-seven HR patients (11% of the whole HR population) received neither AUTO- nor ALLO-HSCT at the end of consolidation courses (Figure 2) due to parent refusal (21 patients), physician decision (10 patients), or complications occurring during chemotherapy treatment and precluding transplantation (7 patients). These 37 HR patients did not differ from the other HR children and their OS and DFS probability, calculated from the end of consolidation chemotherapy, was 47% (SE 7.1) and 43% (SE 7.3), respectively. In comparison with HR patients given ALLO- and AUTO-HSCT, after adjusting for waiting time to transplantation (39 days, range 31-58 days), both DFS and OS of the 37 nontransplanted HR patients were lower (P = .033 and P = .041, respectively).

The 8-year OS and EFS probabilities of 68% and 55%, respectively, achieved in this multicenter study of risk-adapted therapy, based on genetic features and response to induction therapy, are better than those reported in previous AIEOP studies.4  This improvement can be attributed to a lower risk of both treatment-related death and disease recurrence. In particular, the ED rate (3%) of the AML 2002/01 protocol compares favorably with those reported in the LAM-92, LAM-87M, LAM-87, and LAM-82 studies (6%, 14%, 5%, and 9%, respectively).4  Our ED rate is similar to that reported by the BFM5  and MRC groups6,23  but worse than those reported by a monoinstitutional study at St. Jude7  and by the NOPHO24  and Japanese pediatric groups.8  Also, the incidence of death in CCR of our study (10%) was comparable to that recently reported by Rubnitz et al7  (9%). We found that both HR patients and adolescents were exposed to a greater risk of experiencing fatal toxicities. This observation suggests that special attention ought to be paid to the supportive therapy in these categories of children. Previously published studies showed that, also in acute lymphoblastic leukemia, adolescents are more fragile than children.25-27  The reasons behind the observation that adolescents have an increased risk for death in CCR are unclear, but they may reflect age-dependent differences in immunologic response to infections and tissue toxicity following chemotherapy.

The OS and EFS of our cohort of patients are comparable or better than those reported by several other international cooperative groups.5,6,24,28,29  Two recent reports have shown better results, since the Japanese AML99 study reported a 5-year EFS and OS of 61% and 75%,8  respectively, and the AML02 single-center study from St. Jude reported an EFS and OS at 3 years of 63% and 71%, respectively.7  However, it is noteworthy that the number of patients enrolled in the AIEOP AML 2002/01 multicenter study is, together with that reported by Gibson and coworkers,23  the highest of the published protocols on childhood AML and that we report results at 8 years, thus with a follow-up longer than that of St. Jude and Japanese studies.7,8  Moreover, in our trial, neither patients with APL (enrolled in the BFM trial)5  nor those with Down syndrome (enrolled in the MRC trial) were included,6,30  and the proportion of patients with CBF-β abnormalities is lower than that of the Japanese study (20% vs 37%, respectively).8 

The risk stratification based on genetic features and response to induction treatment, chosen in 2002 when the study was designed, has confirmed its prognostic value also in our cohort. Indeed, HR patients had both an OS and EFS probability worse than those of SR patients. However, the difference was less pronounced for EFS than for OS, since a higher proportion of SR patients than HR ones can be rescued by second-line therapies (Table 3); this finding is in line with a recent study reporting on the outcome of children with relapsed AML.31  The outcome of our SR patients was inferior to that reported by other groups.3,6-8,10,32  There is no obvious explanation for this result. Considering compliance with dose intensity as a possible reason for the outcome of our SR patients being worse than expected, the median treatment duration for SR patients was 170 days (range 135-210 days); the estimated median duration of treatment of SR patients was 140 days. This delay could have partly contributed to the unsatisfactory outcome of SR children. The new AIEOP study will address the question of whether the use of flow-cytometry–based minimal residual disease detection, which is certainly much more sensitive than morphologic evaluation for monitoring early leukemia clearance7  and/or early/repeated courses of mitoxantrone,32  can improve the EFS of SR children.

The worse outcome observed in patients with monosomal karyotype has been reported in adult patients,33  but it is a relatively novel observation for childhood AML, since only the negative impact of monosomy 7 has been previously described.10,11,34  In the study by Breems et al,33  out of 184 adults with monosomal karyotype, only 5 patients survived; similarly, in our study, only 2 out of 10 patients are alive and disease-free. Taken together, these results indicate that patients with monosomal karyotype should be considered at HR of treatment failure and should be offered the most effective treatments to prevent disease recurrence.

The outcome of children with WBC count at diagnosis >100 × 109/L was significantly worse than that of patients with lower counts, irrespective of their allocation to either the SR or the HR group (data not shown). This finding is in line with previously published reports,35  although more recent studies have documented that repeated courses of intensive chemotherapy may abrogate the impact on outcome of this variable.3,7 

FLT3-ITD has been reported to be an adverse prognostic factor in both adults and children with AML.36-38  The observation that our 42 patients with FLT3-ITD, 20 of them given ALLO-HSCT, had an outcome not statistically different from that of HR patients without this molecular abnormality can be explained considering that these children, after 2006, were offered, if in CR and with an available HLA-matched donor, ALLO-HSCT, a treatment associated with an immune-mediated graft-versus-leukemia effect. Indeed, the DFS of our children given ALLO-HSCT was 75% +/− 15%. Previously published studies have demonstrated that ALLO-HSCT can supersede the negative prognostic impact of FLT3-ITD in AML.39,40 

The broad use of either AUTO- or ALLO-HSCT in HR patients resulted into a CI of leukemia recurrence superimposable to that of SR children. This favorable effect of transplantation on the risk of recurrence was not offset by an unacceptable CI of TRM. In this regard, it is noteworthy that the outcome of ALLO-HSCT from UD was similar to that of patients given HLA-compatible sibling transplantation. This finding is backed by recent reports in patients with acute lymphoblastic leukemia41,42  or nonmalignant disorders43  and can be interpreted in view of optimization of HLA-typing techniques and strategies for preventing or treating GVHD. Children were given ALLO-HSCT in few experienced centers, while a larger number of centers performed AUTO-HSCT; this conceivably may have contributed to the comparable TRM observed in AUTO- and ALLO-HSCT recipients. Support to the role played by HSCT in preventing leukemia recurrence in our HR patients is also given by the worse OS and DFS calculated by the end of chemotherapy treatment observed in the 37 HR children who were not transplanted at the end of consolidation therapy.

The DFS observed in our HR children given ALLO-HSCT is comparable or better than that reported in other studies, which, however, sometimes also included SR children.8,16,23,44,45  In view of the remarkable chance of salvage recently reported in patients with AML who experience relapse,31,46  transplantation could have been spared in at least a proportion of our HR patients without jeopardizing the probability of OS. This consideration has particular value in view of the possible occurrence of late complications related to transplantation, namely loss of fertility, endocrine disturbances, growth impairment, and extensive chronic GVHD.47  The CI of this immune-mediated complication in our cohort was 9% (SE 2.5).

In conclusion, in a large cohort of children with a long follow-up, we show that risk-adapted therapy and broad use of HSCT resulted in a significant improvement in the outcome as compared with previous AIEOP protocols.4  The next AIEOP study will stratify patients into 3 risk groups, namely SR, HR, and very HR (VHR) children. The former will include children with CBF-β anomalies as well as cytogenetically normal, FLT3-ITD negative and nucleophosmin mutated children with early MRD clearance, measured through flow cytometry.7  In the VHR group, we will allocate children with (1) poor MRD clearance7 ; (2) WBC count at diagnosis >100 × 109/L in the absence of molecular lesions typical of SR; (3) AML FAB-M7 without t(1;22); (4) complex or monosomal karyotype; (5) FLT3-ITD; and (6) recently detected poor-prognosis molecular lesions.48,49  The remaining children will be assigned to the HR group. SR patients will be offered chemotherapy-only treatment, while HR children with an HLA-identical sibling available and all VHR children will be offered an allograft. We will explore whether this refined stratification permits us to further improve patient outcome and whether EFS of HR patients without an HLA-identical sibling can be reproduced by substituting AUTO-HSCT with chemotherapy courses including drugs such as fludarabine and amsacrine.

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 Dr Barbara Buldini, Dr Laura Sainati, Dr Anna Leszl, and Dr Gianni Cazzaniga for their valuable help in the conception and design of the study and for fruitful discussions during trial conduction.

This work was supported in part by a grant from AIRC (Associazione Italiana Ricerca sul Cancro, Special Grant “5x1000”) (F.L.) and by a grant from Italian Ministry of University and Scientific Research (PRIN-2009) (S.R.) (PRIN-2010) (F.L.).

Contribution: A.P. designed the study, treated patients, analyzed the data, and wrote the manuscript; R.M. collected and analyzed the data, treated patients, and wrote the manuscript; C.R., M.C.P., F.C., F.F., M.L., L.L.N., G.M., C.M., N.S., A.M.T., M.Z., and A.B. contributed to the design of the study, treated patients, and collected the data; M.P. performed polymerase chain reaction analyses for characterization of molecular lesions; S.R. analyzed the data and wrote the manuscript; R.R. checked and analyzed the data; G.B. contributed to the design of the study and was responsible for confirmation of diagnosis, flow cytometry analysis, and polymerase chain reaction analyses for characterization of molecular lesions; and F.L. designed the study, treated patients, analyzed the data, and wrote the manuscript.

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

Correspondence: Franco Locatelli, Department of Pediatric Hematology/Oncology, IRCCS Bambino Gesù Children’s Hospital, Piazza Sant’Onofrio, 4, 00165 Rome, Italy; e-mail: franco.locatelli@opbg.net.

1
Pui
 
CH
Carroll
 
WL
Meshinchi
 
S
Arceci
 
RJ
Biology, risk stratification, and therapy of pediatric acute leukemias: an update.
J Clin Oncol
2011
, vol. 
29
 
5
(pg. 
551
-
565
)
2
Kaspers
 
GJ
Zwaan
 
CM
Pediatric acute myeloid leukemia: towards high-quality cure of all patients.
Haematologica
2007
, vol. 
92
 
11
(pg. 
1519
-
1532
)
3
Creutzig
 
U
van den Heuvel-Eibrink
 
MM
Gibson
 
B
, et al. 
AML Committee of the International BFM Study Group
Diagnosis and management of acute myeloid leukemia in children and adolescents: recommendations from an international expert panel.
Blood
2012
, vol. 
120
 
16
(pg. 
3187
-
3205
)
4
Pession
 
A
Rondelli
 
R
Basso
 
G
, et al. 
AML Strategy & Study Committee of the Associazione Italiana di Ematologia e Oncologia Pediatrica (AIEOP)
Treatment and long-term results in children with acute myeloid leukaemia treated according to the AIEOP AML protocols.
Leukemia
2005
, vol. 
19
 
12
(pg. 
2043
-
2053
)
5
Creutzig
 
U
Zimmermann
 
M
Ritter
 
J
, et al. 
Treatment strategies and long-term results in paediatric patients treated in four consecutive AML-BFM trials.
Leukemia
2005
, vol. 
19
 
12
(pg. 
2030
-
2042
)
6
Gibson
 
BE
Wheatley
 
K
Hann
 
IM
, et al. 
Treatment strategy and long-term results in paediatric patients treated in consecutive UK AML trials.
Leukemia
2005
, vol. 
19
 
12
(pg. 
2130
-
2138
)
7
Rubnitz
 
JE
Inaba
 
H
Dahl
 
G
, et al. 
Minimal residual disease-directed therapy for childhood acute myeloid leukaemia: results of the AML02 multicentre trial.
Lancet Oncol
2010
, vol. 
11
 
6
(pg. 
543
-
552
)
8
Tsukimoto
 
I
Tawa
 
A
Horibe
 
K
, et al. 
Risk-stratified therapy and the intensive use of cytarabine improves the outcome in childhood acute myeloid leukemia: the AML99 trial from the Japanese Childhood AML Cooperative Study Group.
J Clin Oncol
2009
, vol. 
27
 
24
(pg. 
4007
-
4013
)
9
Hasle
 
H
Abrahamsson
 
J
Forestier
 
E
, et al. 
Nordic Society of Paediatric Haematology and Oncology (NOPHO)
Gemtuzumab ozogamicin as postconsolidation therapy does not prevent relapse in children with AML: results from NOPHO-AML 2004.
Blood
2012
, vol. 
120
 
5
(pg. 
978
-
984
)
10
von Neuhoff
 
C
Reinhardt
 
D
Sander
 
A
, et al. 
Prognostic impact of specific chromosomal aberrations in a large group of pediatric patients with acute myeloid leukemia treated uniformly according to trial AML-BFM 98.
J Clin Oncol
2010
, vol. 
28
 
16
(pg. 
2682
-
2689
)
11
Harrison
 
CJ
Hills
 
RK
Moorman
 
AV
, et al. 
Cytogenetics of childhood acute myeloid leukemia: United Kingdom Medical Research Council Treatment trials AML 10 and 12.
J Clin Oncol
2010
, vol. 
28
 
16
(pg. 
2674
-
2681
)
12
Amadori
 
S
Testi
 
AM
Aricò
 
M
, et al. 
Prospective comparative study of bone marrow transplantation and postremission chemotherapy for childhood acute myelogenous leukemia. The Associazione Italiana Ematologia ed Oncologia Pediatrica Cooperative Group.
J Clin Oncol
1993
, vol. 
11
 
6
(pg. 
1046
-
1054
)
13
Woods
 
WG
Neudorf
 
S
Gold
 
S
, et al. 
Children’s Cancer Group
A comparison of allogeneic bone marrow transplantation, autologous bone marrow transplantation, and aggressive chemotherapy in children with acute myeloid leukemia in remission.
Blood
2001
, vol. 
97
 
1
(pg. 
56
-
62
)
14
Burnett
 
AK
Goldstone
 
AH
Stevens
 
RM
Hann
 
IM
Rees
 
JK
Gray
 
RG
Wheatley
 
K
UK Medical Research Council Adult and Children’s Leukaemia Working Parties
Randomised comparison of addition of autologous bone-marrow transplantation to intensive chemotherapy for acute myeloid leukaemia in first remission: results of MRC AML 10 trial.
Lancet
1998
, vol. 
351
 
9104
(pg. 
700
-
708
)
15
Horan
 
JT
Alonzo
 
TA
Lyman
 
GH
, et al. 
Children’s Oncology Group
Impact of disease risk on efficacy of matched related bone marrow transplantation for pediatric acute myeloid leukemia: the Children’s Oncology Group.
J Clin Oncol
2008
, vol. 
26
 
35
(pg. 
5797
-
5801
)
16
Niewerth
 
D
Creutzig
 
U
Bierings
 
MB
Kaspers
 
GJ
A review on allogeneic stem cell transplantation for newly diagnosed pediatric acute myeloid leukemia.
Blood
2010
, vol. 
116
 
13
(pg. 
2205
-
2214
)
17
Klusmann
 
JH
Reinhardt
 
D
Zimmermann
 
M
, et al. 
The role of matched sibling donor allogeneic stem cell transplantation in pediatric high-risk acute myeloid leukemia: results from the AML-BFM 98 study.
Haematologica
2012
, vol. 
97
 
1
(pg. 
21
-
29
)
18
Zittoun
 
RA
Mandelli
 
F
Willemze
 
R
, et al. 
Autologous or allogeneic bone marrow transplantation compared with intensive chemotherapy in acute myelogenous leukemia. European Organization for Research and Treatment of Cancer (EORTC) and the Gruppo Italiano Malattie Ematologiche Maligne dell’Adulto (GIMEMA) Leukemia Cooperative Groups.
N Engl J Med
1995
, vol. 
332
 
4
(pg. 
217
-
223
)
19
Ravindranath
 
Y
Yeager
 
AM
Chang
 
MN
, et al. 
Pediatric Oncology Group
Autologous bone marrow transplantation versus intensive consolidation chemotherapy for acute myeloid leukemia in childhood.
N Engl J Med
1996
, vol. 
334
 
22
(pg. 
1428
-
1434
)
20
Locatelli
 
F
Pession
 
A
Bonetti
 
F
, et al. 
Busulfan, cyclophosphamide and melphalan as conditioning regimen for bone marrow transplantation in children with myelodysplastic syndromes.
Leukemia
1994
, vol. 
8
 
5
(pg. 
844
-
849
)
21
Bonetti
 
F
Zecca
 
M
Pession
 
A
, et al. 
The Italian Association for Pediatric Hematology and Oncology-Bone Marrow Transplantation Group
Total-body irradiation and melphalan is a safe and effective conditioning regimen for autologous bone marrow transplantation in children with acute myeloid leukemia in first remission.
J Clin Oncol
1999
, vol. 
17
 
12
(pg. 
3729
-
3735
)
22
Pession
 
A
Dama
 
E
Rondelli
 
R
, et al. 
Italian Association of Paediatric Haematology and Oncology
Survival of children with cancer in Italy, 1989-98. A report from the hospital based registry of the Italian Association of Paediatric Haematology and Oncology (AIEOP).
Eur J Cancer
2008
, vol. 
44
 
9
(pg. 
1282
-
1289
)
23
Gibson
 
BE
Webb
 
DK
Howman
 
AJ
De Graaf
 
SS
Harrison
 
CJ
Wheatley
 
K
United Kingdom Childhood Leukaemia Working Group and the Dutch Childhood Oncology Group
Results of a randomized trial in children with Acute Myeloid Leukaemia: medical research council AML12 trial.
Br J Haematol
2011
, vol. 
155
 
3
(pg. 
366
-
376
)
24
Abrahamsson
 
J
Forestier
 
E
Heldrup
 
J
, et al. 
Response-guided induction therapy in pediatric acute myeloid leukemia with excellent remission rate.
J Clin Oncol
2011
, vol. 
29
 
3
(pg. 
310
-
315
)
25
Rubnitz
 
JE
Lensing
 
S
Zhou
 
Y
Sandlund
 
JT
Razzouk
 
BI
Ribeiro
 
RC
Pui
 
CH
Death during induction therapy and first remission of acute leukemia in childhood: the St. Jude experience.
Cancer
2004
, vol. 
101
 
7
(pg. 
1677
-
1684
)
26
Pichler
 
H
Reismüller
 
B
Steiner
 
M
, et al. 
Austrian ALL-BFM (Berlin-Frankfurt-Münster) Study Group
The inferior prognosis of adolescents with acute lymphoblastic leukaemia (ALL) is caused by a higher rate of treatment-related mortality and not an increased relapse rate - a population-based analysis of 25 years of the Austrian ALL-BFM (Berlin-Frankfurt-Münster) Study Group.
Br J Haematol
2013
, vol. 
161
 
4
(pg. 
556
-
565
)
27
Pui
 
CH
Pei
 
D
Campana
 
D
, et al. 
Improved prognosis for older adolescents with acute lymphoblastic leukemia.
J Clin Oncol
2011
, vol. 
29
 
4
(pg. 
386
-
391
)
28
Burnett
 
AK
Hills
 
RK
Milligan
 
D
, et al. 
Identification of patients with acute myeloblastic leukemia who benefit from the addition of gemtuzumab ozogamicin: results of the MRC AML15 trial.
J Clin Oncol
2011
, vol. 
29
 
4
(pg. 
369
-
377
)
29
Franklin
 
J
Alonzo
 
T
Hurwitz
 
CA
, et al. 
 
COG AAML03P1: Efficacy and safety in a pilot study of intensive chemotherapy including gemtuzumab in children newly diagnosed with acute myeloid leukemia (AML) [abstract]. Blood. 2008;112(11). Abstract 136
30
Burnett
 
AK
Hills
 
RK
Milligan
 
DW
, et al. 
Attempts to optimize induction and consolidation treatment in acute myeloid leukemia: results of the MRC AML12 trial.
J Clin Oncol
2010
, vol. 
28
 
4
(pg. 
586
-
595
)
31
Kaspers
 
GJ
Zimmermann
 
M
Reinhardt
 
D
, et al. 
Improved outcome in pediatric relapsed acute myeloid leukemia: results of a randomized trial on liposomal daunorubicin by the International BFM Study Group.
J Clin Oncol
2013
, vol. 
31
 
5
(pg. 
599
-
607
)
32
Creutzig
 
U
Zimmermann
 
M
Bourquin
 
JP
, et al. 
Second induction with high-dose cytarabine and mitoxantrone: different impact on pediatric AML patients with t(8;21) and with inv(16).
Blood
2011
, vol. 
118
 
20
(pg. 
5409
-
5415
)
33
Breems
 
DA
Van Putten
 
WL
De Greef
 
GE
, et al. 
Monosomal karyotype in acute myeloid leukemia: a better indicator of poor prognosis than a complex karyotype.
J Clin Oncol
2008
, vol. 
26
 
29
(pg. 
4791
-
4797
)
34
Hasle
 
H
Alonzo
 
TA
Auvrignon
 
A
, et al. 
Monosomy 7 and deletion 7q in children and adolescents with acute myeloid leukemia: an international retrospective study.
Blood
2007
, vol. 
109
 
11
(pg. 
4641
-
4647
)
35
Inaba
 
H
Fan
 
Y
Pounds
 
S
, et al. 
Clinical and biologic features and treatment outcome of children with newly diagnosed acute myeloid leukemia and hyperleukocytosis.
Cancer
2008
, vol. 
113
 
3
(pg. 
522
-
529
)
36
Meshinchi
 
S
Alonzo
 
TA
Stirewalt
 
DL
, et al. 
Clinical implications of FLT3 mutations in pediatric AML.
Blood
2006
, vol. 
108
 
12
(pg. 
3654
-
3661
)
37
Staffas
 
A
Kanduri
 
M
Hovland
 
R
, et al. 
Nordic Society of Pediatric Hematology and Oncology (NOPHO)
Presence of FLT3-ITD and high BAALC expression are independent prognostic markers in childhood acute myeloid leukemia.
Blood
2011
, vol. 
118
 
22
(pg. 
5905
-
5913
)
38
Levis
 
M
Small
 
D
FLT3: ITDoes matter in leukemia.
Leukemia
2003
, vol. 
17
 
9
(pg. 
1738
-
1752
)
39
Meshinchi
 
S
Arceci
 
RJ
Sanders
 
JE
Smith
 
FO
Woods
 
WB
Radich
 
JP
Alonzo
 
TA
Role of allogeneic stem cell transplantation in FLT3/ITD-positive AML.
Blood
2006
, vol. 
108
 
1
pg. 
400
  
, author reply 400-401
40
DeZern
 
AE
Sung
 
A
Kim
 
S
, et al. 
Role of allogeneic transplantation for FLT3/ITD acute myeloid leukemia: outcomes from 133 consecutive newly diagnosed patients from a single institution.
Biol Blood Marrow Transplant
2011
, vol. 
17
 
9
(pg. 
1404
-
1409
)
41
Locatelli
 
F
Zecca
 
M
Messina
 
C
, et al. 
Improvement over time in outcome for children with acute lymphoblastic leukemia in second remission given hematopoietic stem cell transplantation from unrelated donors.
Leukemia
2002
, vol. 
16
 
11
(pg. 
2228
-
2237
)
42
Dini
 
G
Zecca
 
M
Balduzzi
 
A
, et al. 
Associazione Italiana Ematologia ed Oncologia Pediatrica–Hematopoietic Stem Cell Transplantation (AIEOP-HSCT) Group
No difference in outcome between children and adolescents transplanted for acute lymphoblastic leukemia in second remission.
Blood
2011
, vol. 
118
 
25
(pg. 
6683
-
6690
)
43
Bernardo
 
ME
Piras
 
E
Vacca
 
A
, et al. 
Allogeneic hematopoietic stem cell transplantation in thalassemia major: results of a reduced-toxicity conditioning regimen based on the use of treosulfan.
Blood
2012
, vol. 
120
 
2
(pg. 
473
-
476
)
44
Tomizawa
 
D
Tabuchi
 
K
Kinoshita
 
A
Hanada
 
R
Kigasawa
 
H
Tsukimoto
 
I
Tsuchida
 
M
Tokyo Children’s Cancer Study Group
Repetitive cycles of high-dose cytarabine are effective for childhood acute myeloid leukemia: long-term outcome of the children with AML treated on two consecutive trials of Tokyo Children’s Cancer Study Group.
Pediatr Blood Cancer
2007
, vol. 
49
 
2
(pg. 
127
-
132
)
45
Perel
 
Y
Auvrignon
 
A
Leblanc
 
T
, et al. 
French LAME (Leucémie Aiguë Myéloblastique Enfant) Cooperative Group
Treatment of childhood acute myeloblastic leukemia: dose intensification improves outcome and maintenance therapy is of no benefit—multicenter studies of the French LAME (Leucémie Aiguë Myéloblastique Enfant) Cooperative Group.
Leukemia
2005
, vol. 
19
 
12
(pg. 
2082
-
2089
)
46
Burnett
 
AK
Goldstone
 
A
Hills
 
RK
, et al. 
Curability of patients with acute myeloid leukemia who did not undergo transplantation in first remission.
J Clin Oncol
2013
, vol. 
31
 
10
(pg. 
1293
-
1301
)
47
Baker
 
KS
Bhatia
 
S
Bunin
 
N
, et al. 
NCI, NHLBI first international consensus conference on late effects after pediatric hematopoietic cell transplantation: state of the science, future directions.
Biol Blood Marrow Transplant
2011
, vol. 
17
 
10
(pg. 
1424
-
1427
)
48
Pigazzi
 
M
Manara
 
E
Bisio
 
V
, et al. 
Screening of novel genetic aberrations in pediatric acute myeloid leukemia: a report from the AIEOP AML-2002 study group.
Blood
2012
, vol. 
120
 
18
(pg. 
3860
-
3862
)
49
Masetti
 
R
Pigazzi
 
M
Togni
 
M
, et al. 
CBFA2T3-GLIS2 fusion transcript is a novel common feature in pediatric, cytogenetically normal AML, not restricted to FAB M7 subtype.
Blood
2013
, vol. 
121
 
17
(pg. 
3469
-
3472
)
Sign in via your Institution