Leukemia (2005) 19, 2030–2042 & 2005 Nature Publishing Group All rights reserved 0887-6924/05 $30.00 www.nature.com/leu Treatment strategies and long-term results in paediatric patients treated in four consecutive AML-BFM trials U Creutzig1, M Zimmermann2, J Ritter1, D Reinhardt1, J Hermann3, G Henze4, H Jürgens1, H Kabisch5, A Reiter6, H Riehm2, H Gadner7 and G Schellong1, for the AML-BFM Study Group 1 Department of Haematology, Oncology, University Children’s Hospital, Münster, Germany; 2Department of Haematology, Oncology, University Children’s Hospital, Hannover, Germany; 3Department of Haematology, Oncology, University Children’s Hospital, Jena, Germany; 4Department of Haematology, Oncology, University Children’s Hospital, Berlin, Germany; 5 Department of Haematology, Oncology, University Children’s Hospital, Hamburg, Germany; 6Department of Haematology, Oncology, University Children’s Hospital, Giessen, Germany; and 7Children’s Cancer Research Institute and St Anna Kinderspital, Vienna, Austria A total of 1111 children with acute myeloblastic leukaemia (AML) were treated in four consecutive Berlin–Frankfurt– Münster (BFM) studies from 1978 to 1998. The first cooperative trial AML-BFM 78 established intensive chemotherapy with seven drugs, CNS irradiation and 2-year maintenance, achieving a long-term survival (overall survival (OS)) of 40%. Induction intensification in AML-BFM 83 resulted in significant improvement of disease-free survival (DFS). The risk of haemorrhage, especially in children with hyperleukocytosis, proved the high relevance of supportive care. In AML-BFM 87, the benefit of CNS irradiation in preventing CNS/systemic relapses was demonstrated. In AML-BFM 93, the introduction of idarubicin during first induction followed by intensification with HAM increased the 5-year EFS, DFS and OS to 5072, 6173 and 5772%, respectively. Stem cell transplantation (SCT), as applied in high-risk patients with a matched related donor, did not significantly improve the outcome compared to chemotherapy alone. In spite of treatment intensification, the therapyrelated death rate decreased from trial to trial, mainly during induction. The future aim is to reduce long-term sequelae, especially cardiotoxicity, by administration of less cardiotoxic drugs, and toxicity of SCT by risk-adapted indications. The AML-BFM studies performed in three European countries with 470 cooperating centres have significantly improved the outcome in AML children; nevertheless, increasing experience with these intensive treatment regimens is of fundamental importance to reduce fatal complications. Leukemia (2005) 19, 2030–2042. doi:10.1038/sj.leu.2403920 Keywords: AML therapy; children; long-term results Introduction The Berlin–Frankfurt–Münster (BFM) group started their cooperation in 1976 for paediatric acute lymphoblastic leukaemia (ALL) and extended the cooperation in 1978 to acute myeloblastic leukaemia (AML). Since that time, four consecutive AML trials have been performed within the AML-BFM study group. Table 1 presents some details of the four trials, including the number of centres involved, their average number of patients and the number of patients per trial. Correspondence: Professor U Creutzig, Klinik und Poliklinik für Kinderheilkunde, Pädiatrische Hämatologie/Onkologie, Albert-Schweitzer-Str. 33, D-48129 Münster, Germany; Fax: þ 49 251 83 56489; E-mail: [email protected] This paper is dedicated to Christa Lausch, our valuable coworker in the AML-BFM Trial Centre from 1982-2003. Received 14 December 2004; accepted 10 May 2005 Background and treatment strategy of the AML-BFM trials The treatment regimens of the four trials are shown schematically in Figure 1, and the treatment elements of the AML-BFM protocols including drug dosages are summarised in Table 2. Each trial was based on the experience of the previous studies and of other AML trials in children or adults. The intensity of treatment was increased from trial to trial, with the exception of AML-BFM 87. Before 1978, children with AML had been treated in Germany either according to ALL strategies or adult AML regimens, and the outcome was accordingly poor. Between 7/1973 and 10/1978, 23 children with AML were treated at the University Children’s Hospitals in Münster and Berlin with a similar drug combination as used in the ‘West-Berlin Pilot Study’ for children with ALL,1 however, including more AML specific drugs like cytarabine and anthracyclines, according to a schedule which was also introduced by Riehm et al. Based on the fact that the outcome of this group was better than expected, with a 4-year survival probability of 44%,2 the first cooperative AML-BFM trial was initiated in 1978. AML-BFM 78: The AML-BFM 78 treatment protocol used a seven-drug, 8-week induction/consolidation (see Consolidation, Table 2), which was similar to the initial ‘West-Berlin’ ALL therapy regimen,1 combined in its second phase with preventive CNS irradiation (standard dose ¼ age-dependent: 0–o1 year ¼ no irradiation, 1–o3 years ¼ 15 Gy, X3 years ¼ 18 Gy) and followed by 2 years of maintenance with daily thioguanine 40 mg/m2 orally and cytarabine 40 mg/m2 s.c. 4 days monthly for a total duration of 24 months. Doxorubicin was given every 8 weeks during the first year of maintenance. The probability of event-free survival (EFS) at 5 years was 3874%,3 which was a good result at that time. Therefore, this therapy strategy was used as backbone for the following trials. AML-BFM 83: At the beginning of the 80s, AML studies for adults had demonstrated that cytarabine administration for 7 days was superior to a 5-day treatment with the same agent (Cancer and Leukemia Group B (CALGB) Study in adults4), and that high remission rates could be achieved with thioguanine, cytarabine, daunorubicin (TAD) induction in the AMLCG Study.5 Consequently, in AML-BFM 83, therapy started with an 8-day induction cycle consisting of cytarabine and daunorubicin (3 60 mg/m2) as in the adult AMLCG study, however, as thioguanine was already included as daily medication during maintenance, it was replaced by etoposide. This ADE Improved survival in paediatric AML, BFM strategy U Creutzig et al 2031 Table 1 Patient accrual and follow-up Trial AML-BFM AML-BFM AML-BFM AML-BFM 78 83 87 93 Accrual of patients: time period Total number of patients (n) Centres (n) 12/78–09/82 12/82–09/86 12/86–09/92 1/93–06/98 151 182 307 471 30 30 37 68 Figure 1 Flow diagram of trials AML-BFM 78–93. RT ¼ CNS radiotherapy. ( ¼ cytarabine, daunorubicin and etoposide) induction was followed by an 8-week induction/consolidation therapy as administered in AML-BFM 78. After this treatment element, maintenance was given as in the previous trial. Compared to the previous trial, AML-BFM 83 resulted in a major improvement in long-term results, with a 5-year EFS of 4774%.6 AML-BFM 87: The main question of this trial was the importance of CNS irradiation for CNS prophylaxis. Irradiation was not generally included in therapy protocols for AML in adults and children. Most investigators agreed that cranial irradiation could prevent CNS relapses, but its effect on overall remission duration was unknown.7 Results of ALL trials in children8 and of the AML study 72–75 reported by Dahl et al indicated that cranial irradiation had an impact on the number of CNS relapses, but not on overall survival (OS).9 Most AML studies in children and adults used intrathecal methotrexate or cytarabine or a combination of these agents with hydrocortisone as CNS-directed therapy. In the AML-BFM studies, CNS irradiation was generally included. The AML-BFM 87 study tested prospectively if CNS irradiation could be replaced by late intensification therapy with high-dose cytarabine and etoposide. The premature decision to stop irradiation for all children, however, resulted in a significant increase of CNS and systemic relapses. Detailed analyses of the study cohort and the randomised and nonrandomised patients clearly demonstrated the need of prophylactic irradiation within the AML-BFM setting. As this conclusion was based on nonrandomised patients too, these findings still have to be confirmed by other paediatric or adult AML trials. ADE induction therapy remained the same as in the previous study. Duration of consolidation was reduced to 6 weeks, because considerable variations with many interruptions due to aplasia or toxicity had occurred in AML-BFM 83 during the Number of patients/centre (median/range) 4 5 6 5 (1–24) (1–22) (1–33) (1–41) Number of patients/year 41 48 53 86 Follow-up (median, range, years) 15 11 9 5 (2–19) (4–17) (3–14) (1–9) second phase of the 8-week consolidation.10 After consolidation, two cycles of late intensification with high-dose cytarabine (3 g/m2) and etoposide (HAE) were added. CNS irradiation was scheduled at the beginning of maintenance, and patients without CNS involvement were randomly assigned to receive an irradiation of 18 Gy or no irradiation. Duration of maintenance was reduced to approximately 12 months (total therapy duration: 18 months), and doxorubicin was removed from maintenance therapy since AML-BFM 87. Allogeneic stem cell transplantation (SCT) in first CR (after the second treatment course) was recommended in children of the high-risk group only if a sibling donor was available. Based on the results of studies AML-BFM 83 and 87, two different risk groups (standard and high) could be defined, see ‘risk group definition’ page 8. AML-BFM 93: Patients were randomly assigned to receive the 8-day induction with either daunorubicin (ADE) or idarubicin (AIE) in order to compare the efficacy and toxicity of these two drugs (Figure 2). The rationale for using idarubicin was that in vitro and preclinical studies had suggested a possibly higher clinical benefit of idarubicin, showing a faster cellular uptake, increased retention and lower susceptibility to multidrug resistance.12,13 Additional advantages seemed to arise from the long plasma half-life of 54 h of its main metabolite idarubicinol, which also showed antileukaemic activity in the cerebral spinal fluid.14 Furthermore, idarubicin showed less cardiotoxic side-effects in several animal models12 – an important finding regarding the risk of an increased anthracycline-induced cardiomyopathy in children.15 After induction, patients were treated depending on risk groups (see Figure 3).11 High-risk patients were randomised to receive either high-dose cytarabine and mitoxantrone (HAM) followed by consolidation ( ¼ arm early HAM) or 6-week consolidation followed by HAM ( ¼ arm late HAM). Standardrisk patients received consolidation only, without HAM. Subsequently, all patients were treated with one intensification cycle of HAE instead of two HAE cycles after consolidation as in AML-BFM 87. Recommendations for SCT in first CR were the same as in the previous trial. The rationale for this schedule with HAM was the intention to improve even more the unsatisfactory outcome in children with high-risk AML. Another aim of AML-BFM 93 was to evaluate whether placing HAM as the first or second post-induction treatment cycle had a different impact and prognosis. This question was based on the experience in adults with AML showing that the dose intensity during the first two treatment courses was of prognostic significance.16 In view of reports showing that this intensification regimen was associated with increased toxicity and consequently required rigorous supportive care, HAM was restricted to high-risk patients. Leukemia 2032 Leukemia Table 2 Treatment elements in the four trials AML-BFM 78, 83, 87 and 93 AML-BFM 78 AML-BFM 83 AML-BFM 87 ADE induction introduced in trial 83 Not given ADE: cytarabine and etoposide as before ADE: cytarabine, daunorubicina and etoposide as before daunorubicin 30 mg/m2 30 min infusion every 12 h on days 3–5 or with idarubicin (AIE): idarubicin 12 mg/m2 30 min infusiona every 24 h, days 3–5 (instead of daunorubicin), cytarabine and etoposide as before Consolidation introduced in trial 78 8-week induction/consolidation: Phase 1: 6-thioguanine 60 mg/m2/day, days 1–43 orally; prednisone 60 mg/m2/day, days 1–28 orally; vincristine 1.5 mg/m2/day, days 1, 8, 15, 22; doxorubicin 25 mg/m2/day, days 1, 8, 15, 22; cytarabine 75 mg/m2/day, days 3–6, 10–13, 17–20, 24–27, Phase 2: cytarabine 75 mg/m2/day, days 31–34, 38–41, 45–48, 52–55; doxorubicin 25 mg/m2/day, days 36, 50; cyclophosphamide 500 mg/m2/day, days 29, 43, 57 Not given ADE: cytarabine 100 mg/m2/day continuous infusion on days 1 and 2 followed by 30 min infusion every 12 h on days 3–8, daunorubicin 60 mg/m2 30 min infusion per day on days 3, 4, 5 etoposide 150 mg/m2 60 min infusion on days 6–8 As in trial 78, except: Not given Not given Not given HAE introduced in trial 87 HAM introduced in trial 93 Doxorubicin 30 mg/m2/day, days 1, 8, 15, 22; Phase 2: no doxorubicin cyclophosphamide 500 mg/m2/day, days 29, 43 18 Gy ( ¼ standard dose in children As in trial 78 X3 years), o1 year ¼ 12 Gy, 1–2 years ¼ 15 Gy (during the second phase of consolidation) Intrathecal therapy Methotrexate 12.5 mg days 31; 38, 45; 52 Cytarabine days 31; 38, 45; 52 (during (during consolidation) consolidation) adjusted to age, children o1 year ¼ 20 mg, 1–2 years ¼ 26 mg, 2–3 years 34 mg, X3 years 40 mg 2 As in trial 78 Maintenance Daily thioguanine 40 mg/m orally, cytarabine 40 mg/m2 s.c. 4 days introduced in trial 78 monthly for a total duration of 24 months; doxorubicin 25 mg/m2 every 8 weeks during the first year Stem cell transplantation Not given Not recommended CNS irradiation introduced in trial 78 a Since October 1996, 4 h infusion of daunorubicin and idarubicin. 6-week consolidation: 6-thioguanine 60 mg/m2/day, days 1–43 orally; prednisone 40 mg/m2/day, days 1–28 orally; vincristine 1.5 mg/m2/day, days 1, 8, 15, 22; doxorubicin 30 mg/m2/day, days 1, 8, 15, 22; cytarabine 75 mg/m2/day, days 3–6, 10–13, 17–20, 24–27, 31–34, 38–41; cyclophosphamide 500 mg/m2/day, days 29, 43 AML-BFM 93 As in trial 87 Two cycles of high-dose cytarabine 3 g/m2 One cycle HAE 3-h infusion every 12 h for 3 days and etopside 125 mg/m2 on days 2–5 Not given High-dose cytarabine 3 g/m2 3-h infusion every 12 h for 3 days, mitoxantrone 10 mg/m2 on days 4 and 5 High-risk patients only As in trial 78 As in trial 78 except, compared with no irradiation o1 year ¼ no irradiation (at the beginning (at the beginning of maintenance) of maintenance) Cytarabine days 1, 15, 29, 43 (during consolidation and concomitantly with CNS irradiation days 1, 8, 15, 21) adjusted to age Without doxorubicin total duration 18 months (maintenance 12 months) As in trial 87 in addition at day 1 during induction Matched related in high-risk patients As in trial 87 As in trial 87 Improved survival in paediatric AML, BFM strategy U Creutzig et al New element Improved survival in paediatric AML, BFM strategy U Creutzig et al 2033 SR Consolidation ADE Cumulative doses HAM R AIE Consolidation HAE Maintenance1 year HR Consolidation HAM alloSCT, if MSD available 200 mg 180 mg/m2 10 g 120 mg/m2 100 mg/m2 18 g/m2 Cytarabine 41.1 g/m2 Anthracyclines 400 mg/m2 18 g/m2 1.9 g/m2 1.4 g/m2 1.8 g/m2 Figure 2 Overview of the AML-BFM 93 regimen and cumulative doses of cytarabine and anthracyclines. HAM ¼ high-dose cytarabine/ mitoxantrone; SR ¼ standard risk; HR ¼ high-risk; RT ¼ CNS radiotherapy; ADE ¼ cytarabine/daunorubicin/etoposide; AIE ¼ cytarabine/idarubicin/ etoposide; HAE ¼ high-dose cytarabine/etoposide. M0 and M7 subtypes always required immunological confirmation.18,19 Day 15 bone marrow aspirates were also reviewed centrally. Definitions and statistics Figure 3 Risk classification by morphology, cytogenetics and response on day 15 – according to the current AML-BFM trials (until trial 93 information about cytogenetics were not included). Patients and methods Eligibility (see editorial) The entry criteria for studies AML-BFM 78–93 were: Newly diagnosed AML (diagnosis confirmed by the reference laboratory), patient aged between 0 and 17 years, and written informed consent signed by the patient or parent. Patients with myelosarcoma, secondary AML, myelodysplastic syndrome or Down’s syndrome as well as patients with a pretreatment for more than 14 days were registered and treated with the standard study therapy – partially with reduced dosages – but analysed separately from the study patients (see paragraph ‘Down syndrome’). Diagnosis The initial diagnosis of AML and its subtypes was established according to the FAB classification.17–19 Initial smears were centrally reviewed at the University Children’s Hospital in Münster and in addition reviewed by a regular panel, including an external investigator (H Löffler). The diagnoses of Complete remission (CR) was defined according to the CALGB criteria20 with minor deviations as described below, and had to be achieved at the end of intensification treatment. Definition of CR: p5% leukaemic blasts in the bone marrow with signs of normal haematopoiesis in the bone marrow and with clear signs of regeneration of normal blood cell production in the peripheral blood (platelets 480 109/l without transfusions, neutrophils 41.0 109/l), and no leukaemic cells in the peripheral blood or anywhere else (the required platelet and neutrophil number is lower than originally requested, because many patients have slow regeneration especially of platelets after the intensive therapy used today). Early death (ED) patients were those dying before or within the first 6 weeks of treatment. Response after induction was evaluated on day 15 by the blast cell count in the bone marrow p/45%. Since the main cause of death is different in each specific phase of initial treatment, ED was subclassified into (a) ED before starting treatment, (b) ED during and after the first therapy course (p14 days of treatment), (c) ED in aplasia between day 15–42 before remission with bone marrow regeneration is generally seen (after the second induction course). This classification reflects the ED rate due to initial problems (hyperleukocytosis, leukostasis) or due to aplasia after induction therapy. Nonresponders (NR): All patients not achieving remission (CR) and surviving the first 6 weeks of treatment were classified as NR. Patients with CR criteria lasting less than 4 weeks were also reported as NR (type V failure according to CALGB criteria). Toxicity was assessed according to the NCI common toxicity criteria.21 Leukemia Improved survival in paediatric AML, BFM strategy U Creutzig et al 2034 EFS was calculated from the date of diagnosis to the last follow-up or first event (failure to achieve remission, resistant leukaemia, relapse, second malignancy or death of any cause). Patients who did not attain a complete remission were considered failures at time zero. Survival was calculated from the date of diagnosis to death of any cause or last follow-up, disease-free survival (DFS) from the date of remission to the first event (relapse, second malignancy, death of any cause). Univariate analysis was conducted by the Wilcoxon test for quantitative variables and Fisher’s exact test for qualitative variables. When frequencies were sufficiently large, w2 statistic was used. Probabilities of survival were estimated using the Kaplan–Meier method with standard errors according to Greenwood, and were compared with the log-rank test. Cumulative incidence functions of relapse and death in CCR were constructed by the method of Kalbfleisch and Prentice. Functions were compared with Gray’s test. Analysis of efficacy data was performed according to the intent-to-treat principle. Toxicity data were evaluated for treatment groups. Computations were performed using Statistical Analysis System (SAS Version 6.12, SAS Institute Inc, Cary, NC, USA). The database for all analyses was ‘frozen’ on March 1, 2002. Patient characteristics From January 1978 to June 1998, 1111 patients were enrolled in studies AML-BFM 78–93. A comparison of initial patient data (see Table 3) of AML-BFM 78, 83, 87 and 93 did not reveal clinically important differences, except for the shift from FAB type M1 to M2 seen from studies 78/83 to 87/93 due to more precise definitions of these types and the introduction of the FAB-types M0 (1991)19 and M7 (1985).18 Results Overall outcome in AML-BFM 78, 83 and 87, 93 The results of the four trials are given in Table 4. In addition, the results in patients o15 years of age are presented separately in order to facilitate comparisons with other studies (Table 4a). Overall, the probabilities of survival, EFS and DFS have improved in the four consecutive studies – with the exception of AML-BFM 87 (see Figures 4 and 5). The ED rate (before therapy or during the first 42 days) decreased from 12.6% in AML-BFM 78 to 7.4% in AML-BFM 93 (P-trend ¼ 0.03). The decrease was seen mainly during the first phase of treatment (before day 15), see Table 4b. Death rates between day 15 and 150 remained similar in all studies (Table 4b). The percentage of nonresponse did not change significantly (12.1% in AML-BFM 83, 10.4% in AML-BFM 93, Table 4). The main reason for a better outcome was a reduction in relapse rates. Results of AML-BFM 78: Results of the first AML-BFM trial were considerable, with a 5-year survival of 4274%. This was achieved by an 8-week induction consolidation therapy, including CNS irradiation, followed by continuous maintenance treatment over 2 years. These treatment elements were the basis for the following trials. Results of AML-BFM 83: The main difference between the first two AML-BFM studies, 78 and 83, was the addition of an Leukemia 8-day ADE induction treatment in the second trial. Due to this intensification, the relapse rate was reduced but not the rate of induction failures. The probability of a 5-year EFS increased from 3874% in AML-BFM 78 to 4774% in AML-BFM 83, Plogrank 0.13 (P-survival to 5274%, Plogrank 0.07). The improvement of the 5-year DFS was significant in the second trial (from 4875 to 6274%, Plogrank 0.03). According to the results in AML-BFM 83, two different risk groups could be identified by combinations of predominantly pretherapeutic parameters (see the section ‘Risk groups’). Results of AML-BFM 87: AML-BFM 87 compared prospectively whether CNS irradiation could be abandoned by adding two cycles of intensification with high-dose cytarabine and etoposide after consolidation, and furthermore whether this could improve prognosis compared to AML-BMF 83. Out of 307, 230 (75%) children achieved complete remission. Kaplan–Meier estimates for survival, EFS and DFS of 5 years were: 4973, 4173 and 5573%. Preventive cranial irradiation was either given or not in children without initial CNS involvement after achieving remission. It remained mandatory for patients with leukocyte count 470 000/ml and/or initial CNS involvement. Results excluding the latter group as well as patients with events before the scheduled time of irradiation showed that the cumulative incidence (CI) of relapses was significantly lower in irradiated than in nonirradiated patients (5-year CI 2975% vs 5076%, P(Gray) ¼ 0.001; Figure 6). This comparison included randomised and non-randomised patients; similar incidences were found in randomised patients only (5-year CI 33714 vs 56712%, P(Gray) ¼ 0.12). Relapses in nonirradiated children occurred mainly in the bone marrow and less often in the CNS. Results of AML-BFM 93: In AML-BFM 93, 387 of 471 (82%) patients achieved remission. The estimated probabilities for 5-year survival, EFS and DFS were 5772, 5072 and 6173%, respectively. Idarubicin induction resulted in a significantly better blast cell reduction in the bone marrow on day 15 (17% of patients with 45% blasts compared to 31% of patients in the daunorubicin arm, Pw2 ¼ 0.01).22 However, probabilities of 5-year EFS (IDA 5174% vs ADE 5074%, Plogrank 0.72) and survival were similar (IDA 6074% vs ADE 5774%, Plogrank 0.55).22,23 Estimated survival and pEFS were higher in AML-BFM 93 than in AML-BFM 87 (Plogrank 0.01; Table 4, Figures 4 and 5). This improvement, however, concerned only the 310 high-risk patients (remission rate in AML-BFM 93 vs AML-BFM 87: 78 vs 68%, P ¼ 0.007, and 5-year pEFS 42 vs 31%, Plogrank 0.03; Figure 7). There was also a significant difference for high-risk patients in terms of survival (5073 vs 3973%, Plogrank 0.03), but not of DFS (5473 vs 4674%, Plogrank 0.46). As to the 161 standard-risk patients, 5-year survival, pEFS and DFS were similar to those of AML-BFM 87: 7174 vs 7175%, Plogrank 0.88; 6574 vs 6375%, Plogrank 0.5; 7374 vs 7075%, Plogrank 0.42, respectively. Outcome of the 28 high-risk patients receiving an allogeneic matched related donor SCT in first CR was in the same range as of the non-SCT high-risk patients (DFS 6479%). Figure 8 shows the estimated survival for high-risk patients in AML-BFM 87 and 93 with and without SCT. Results by randomisation of early HAM vs late HAM in high-risk patients: In all, 196 patients were randomised to either early HAM (n ¼ 98) or late HAM (n ¼ 98). Overall results regarding response and relapse rates were similar in both arms Improved survival in paediatric AML, BFM strategy U Creutzig et al 2035 Table 3 Initial patient data of trials AML-BFM 78, 83, 87 and 93 AML-BFM 78 AML-BFM 83 AML-BFM 87 AML-BFM 93 n % n % n % n % Number of eligible patients Gender (male) 151 81 53.6 182 99 54.4 307 166 54.1 471 255 54.1 Age o2 years 2–9 years X10 years 23 55 73 15.2 36.4 48.3 33 69 80 18.1 37.9 44.0 67 123 117 21.8 40.1 38.1 112 174 185 23.8 36.9 39.3 Leukocytes ( 103/mm3) o20 20–o100 X100 71 47 33 47.0 31.1 21.9 84 60 38 46.2 33.0 20.9 131 118 58 42.7 38.4 18.9 241 143 87 51.2 30.4 18.5 CNS leukemia (yes) (ND or questionable) 13 (3) 8.8a 10 (13) 5.9a 36 (13) 12.2a 45 (36) 10.3a 42 (39) 29.4a 85 (45) 32.4a 96 (89) 25.1a 0b 37b (18) 37 (23) 5 45 (17) 48 7 3b 0 0 20.3 (9.9) 20.3 (12.6) 2.7 24.7 (9.3) 26.4 3.8 1.6 0 17b 30b (16) 84 (57) 15 66 (38) 69 8 17b 1 5.5 9.8 (5.2) 27.3 (18.6) 4.9 21.5 (12.4) 22.5 2.6 5.5 0.3 25 55 (35) 125 (94) 23 91 (36) 101 16 31 4 5.3 11.7 (7.4) 26.5 (20.0) 4.9 19.3 (7.6) 21.4 3.4 6.6 0.8 123 12 8 1 3 20.3a 13.6 1.7 5.1 161 42 25 12 5 28.8a 17.1 8.2 3.4 185 82 41 25 16 28.7a 14.3 8.7 5.6 21 26 4 35.6 44.1 6.7 29 75 17 19.9 51.4 11.6 58 146 31 20.3 51 10.8 Blasts day 15 45% (ND) FAB types M0 M1 (M1 with Auer)a M2 (M2 with Auer)a M3 M4 (M4eo) M5 M6 M7 Other (basophilic leukaemia) Karyotypes ND Cytogenetic favourablec t(8;21) inv(16) t(15;17) Normal Other t(9;11) 0 36 (17) 34 (26) 6 40 (12) 32 3 0 151 0 24 (11) 23 (17) 4 27 (8) 21 2 0 ND ¼ no data. Percentage of patients with data, no information about Auer rods in eight patients of study 83 and four patients of study 87. b P(w2) o0.05. c Definition of favourable cytogenetics: t(8;21), t(15;17), inv(16). a (5-year survival, EFS and DFS in early HAM compared with late HAM patients were: 5775 vs 5475%; 4975 vs 4175%; and 5676 vs 4976%). The pEFS was slightly higher in patients initially treated with daunorubicin who received early HAM instead of late HAM, whereas results with early or late HAM were similar in patients initially treated with idarubicin.24 Toxicity in AML-BFM 93 during intensive therapy elements and according to randomisation: Nonhaematological toxicities of CTC-grade 3/4 were mainly infections occurring in 17–18% of patients during induction and HAM, and in 11–12% of patients during consolidation and late intensification HAE. Other nonhaematological toxicities were rarely seen (o3% of patients) during the intensive therapy elements of AML-BFM 93. The infection rate in the AIE group was slightly higher than in the ADE group (P-trend ¼ 0.016), and duration of aplasia (until neutrophil recovery to 500/ml) was also 2 days longer in the AIE patients. Toxicities were similar in the arms of early HAM and late HAM. Toxicity in trials AML-BFM 78–93 To compare acute toxicities in different studies, fatal events are listed in Table 4b according to time periods. EDs, mainly due to haemorrhage or leukostasis, occurring before or during the first 15 days of treatment, could be reduced from trial to trial. The number of deaths in aplasia or early after achieving CR (day 15–150) did not change significantly during the studies. Prophylactic CNS irradiation was used generally in the BFM trials. Only in AML-BFM 87 a significant group of children did not receive irradiation. In order to evaluate late sequelae, longterm survivors were tested for neuropsychological functions Leukemia Improved survival in paediatric AML, BFM strategy U Creutzig et al 2036 Table 4 Results in trials AML-BFM 78, 83, 87 and 93 (total groups): (a) results in trials AML-BFM 78, 83, 87 and 93 (only patients o 15 years at diagnosis); (b) death before or during induction and intensification in trials AML-BFM 78, 83, 87 and 93 (total groups) AML-BFM 78 n Number of patients Median follow-up of pts. in CCR (years, range) a AML-BFM 83 % (s.e.) 151 15.2 n 182 (2.1–19.4) 11.1 AML-BFM 87 AML-BFM 93 % (s.e.) n % (s.e.) n % (s.e.) 100 307 100 471 100 (4.4–16.6) 9.4 (2.6–13.8) 5.1 (1.1–9.0) 19 13 119 12.6 8.6 78.8 21 22 139 11.5 12.1 76.4 28 49 230 9.1 16.0 74.9 35 49 387 7.4 10.4 82.2 Death in CCR (cumulative incidence) Relapse (cumulative incidence) (with CNS involvement) 10 57 9 5.3 (3) 35.2 (6) 5.3 (3.2) 7 50 7 3.3 (2) 25.8 (5) 3.8 (2.3) 9 98 15 2.9 (2) 30.6 (4) 4.9 (2.2) 18 131 23 3.8 (1) 28.1 (3) 4.9 (1.3) Lfu in CCRb 12 11 6.0 18 5.9 Early deaths (total) Nonresponse CR achieved 7.9 9 1.9 p survival At 5 years At 10 years 42 (4) 39 (4) 52 (4) 50 (4) 49 (3) 48 (3) 57 (2) pEFS At 5 years At 10 years 38 (4) 36 (4) 47 (4) 46 (4) 41 (3) 40 (3) 50 (2) pDFS At 5 years At 10 years 48 (5) 44 (5) 62 (4) 60 (4) 55 (3) 53 (3) 61 (3) Allogeneic BMT in first CR (a) only patients o15 years at diagnosis Number of patients Median follow-up of pts. in CCR (years, range) Early deathsa Nonresponse CR achieved 4 2.2 17 5.5 42 8.9 146 15.0 18 12 116 100.0 (2.1–15.0) 12.3 8.2 79.5 161 11.3 18 19 124 100.0 (4.4–15.0) 11.2 11.8 77.0 283 9.4 25 47 211 100.0 (3.7–13.8) 8.8 16.6 74.6 427 7.5 31 42 354 100.0 (1.1–9.0) 7.3 9.8 82.9 Death in CCR (cumulative incidence) Relapse (cumulative incidence) (with CNS involvement) Lfu in CCRb 9 56 9 12 4.8 (3) 35.7 (6) 5.5 (3.3) 8.2 6 44 6 10 3.1 (2) 26.1 (5) 3.7 (2.4) 6.2 6 89 14 18 2.1 (1) 30.0 (4) 4.9 (2.3) 6.4 15 118 21 7 3.5 (1) 27.8 (3) 4.9 (1.4) 1.6 p survival At 5 years At 10 years 61 54 42 (4) 40 (4) 82 56 52 (4) 51 (4) 137 49 50 (3) 48 (3) 137 58 (2) pEFS At 5 years At 10 years 56 49 39 (4) 36 (4) 75 51 48 (4) 47 (4) 117 38 42 (3) 41 (3) 124 51 (3) pDFS At 5 years At 10 years 55 49 48 (5) 45 (5) 75 51 62 (4) 60 (4) 117 36 57 (3) 54 (3) 104 62 (3) 100 471 4 21 35 100 (b) death before or during induction and intensification in trials AML-BFM 78, 83, 87 and 93 (total groups) Patients No. 151 100 182 100 307 ED before therapy 2 9 4 9.3 9.8 ED oday 15 12 9 14 15 9.9 13 7.1 26 Death day 15 –o150 (during intensification courses)c 5.9 8.5 5.3 7.5 CR, complete remission; CCR, continuous complete remission; s.e., standard error; lfu, lost to follow-up. a Early deaths are defined as death until day 42 and reported in detail (a) before starting therapy (b) during the first 14 days of treatment in Table 4b. b Lfu after 0.7–11.5, median 8.7 years (all patients). c Irrespective of response or nonresponse. such as concentration, intelligence quotient and academic performance. Although no statistically different results regarding intelligence quotient and concentration or academic perforLeukemia mance could be shown, questionnaires to patients and parents revealed some problems in social behaviour and school attendance in children who had been irradiated prior to 5 years Improved survival in paediatric AML, BFM strategy U Creutzig et al 2037 Figure 4 Estimated probability of survival in patients of trials AML-BFM 78–93, 5-year data given. Slash indicates the patient alive with the shortest follow-up. Figure 7 Comparison between the estimated probability of eventfree survival in standard and high-risk patients in trial AML-BFM 93 and trial AML-BFM 87, 5-year data given (slash: see Figure 4). Figure 5 Estimated probability of event-free survival in patients of trials AML-BFM 78–93, 5-year data given (slash: see Figure 4). Figure 8 Estimated probability of survival for high-risk patients with or without matched related (MRD) SCT in first CR (survival 443 years, median time to SCT) in trials AML-BFM 87 and 93, 5-year data given (slash: see Figure 4). in the four AML-BFM trials so far, including two brain tumors and two thyroid carcinomas). Risk factors Table 5 details the results for different risk parameters, showing an improvement especially for children with a lower risk (first standard-risk definition was slightly different from the currently used definition, see next paragraph) in AML-BFM 83 compared to AML-BFM 78 and in high-risk patients in AML-BFM 93 compared to the previous studies. The better outcome concerned patients with a blast count 45% on day 15, as well as patients with hyperleukocytosis. Figure 6 Cumulative incidence of relapses in trial AML-BFM 87 in patients with and without CNS irradiation, 5-year data given (slash: see Figure 4). of age.25 An analysis of growth and endocrine system did not show differences between irradiated and nonirradiated children. Finally, the overall risk of secondary malignancies was low (16 Risk group definition: The main risk factors had been defined according to the results of AML-BFM 836 and were reanalysed, including the data of AML-BFM 87,11 leading to the current risk definition (Figure 3). In AML-BFM 83, among the different FAB types, the difference in prognosis was significant in patients with (a) M1 Leukemia Improved survival in paediatric AML, BFM strategy U Creutzig et al 2038 Table 5 Results according to different risk parameters in trials AML-BFM 78–93 (5-year pEFS (%), only for subgroups nX10) Presenting feature AML-BFM 78 AML-BFM 83 AML-BFM 87 AML-BFM 93 Total number of patients pEFS (s.e.) Total number of patients pEFS (s.e.) Total number of patients EFS (s.e.) Total number of patients EFS (s.e.) SR HR 61a 90a 43 (6) 36 (5) 51 131 78 (6) 35 (4) 99 208 63 (5) 31 (3) 161 310 65 42 (3) Cytogenetics favourableb t(8;21) inv(16) t(15;17) Cytogenetics normal Cytogenetics other 11q23 (excl. t(9;11)) t(9;11) ND 12 8 1 3 21 26 ND 4 83 (11) ND ND ND 43 (11) 39 (10) ND ND 42 25 12 5 29 75 ND 17 55 (8) 64 (10) 33 (14) 82 41 25 16 58 146 20 31 70 63 80 69 51 41 50 48 (5) (8) (8) (12) (7) (4) (11) (9) ND 37 37 5 45 17 48 7 3 62 (8) 54 (8) ND 49 (8) 77 (10) 27 (6) ND ND 17 30 84 15 66 38 69 8 17 18 (9) 25 55 125 23 91 36 101 16 31 40 48 60 65 55 80 37 31 52 (10) (7) (5) (10) (5) (7) (5) (12) (9) 101 42 60 (5) 36 (7) 177 85 54 (4) 27 (5) 286 96 57 (3) 44 (5) FAB FAB FAB FAB FAB FAB FAB FAB FAB M0 M1 M2 M3 M4 (excl. M4eo) M4eo M5 M6 M7 ND ND ND 36 34 6 40 12 32 3 ND 44 (8) 41 (8) ND 32 (7) 42 (14) 41 (9) 45 (9) 28 (5) ND 35 (12) 29 47 52 53 42 53 32 (11) (9) (5) (13) (6) (8) (6) Blasts day 15 p5% Blasts day 15 45% ND ND Leukocytes o 100 000/ml Leukocytes 4100 000/ml 118 33 44 (5) 18 (7) 144 38 54 (4) 21 (7) 249 58 45 (3) 24 (6) 384 87 52 (3) 39 (5) Age o2 years Age X2 years 23 128 44 (10) 37 (4) 33 149 42 (9) 48 (4) 67 240 22 (5) 47 (3) 112 359 41 (5) 53 (3) CNS positive CNS neg. 13 135 23 (12) 41 (4) 10 159 30 (15) 49 (4) 36 260 36 (8) 42 (3) 45 390 42 (8) 53 (3) ND ¼ no data. a Risk definition in study 78 according to morphological parameters only. b Definition of favorable cytogenetics: t(8;21), t(15;17), inv(16). with or without Auer rods, (b) in patients with M2 with a white blood cell count lower or higher than 20 000/ml and (c) in M4 patients with or without atypical eosinophils (P each o0.07).6 According to these results, two different risk groups could be identified by combinations of predominantly pretherapeutic parameters: The low-risk group, comprising 32% of the patients, included the FAB types with granulocytic differentiation and specific additional features: FAB M1 with Auer rods, FAB M2 with WBC o20 000 mm3, FAB M3 (all patients) and FAB M4 with eosinophils. The 5-year Kaplan–Meier estimation of DFS for this group was 9074%, compared to 4775% in the highrisk group (all other patients).6 Based on a higher number of patients and the results of AMLBFM 83 and 87, the risk group definition (standard and high-risk) was slightly modified and could be improved mainly by including the blast cell reduction in bone marrow on day 1511 in addition to the initial morphological parameters (Figure 3). The standard-risk group included: FAB M1/M2 with Auer rods, FAB M4eo (if these patients presented with X5% blasts in the bone marrow on day 15, they were shifted to the high-risk group) and FAB M3 (all patients regardless of the blast count on day 15); high-risk group: ) all others. The risk group definition was prospectively used in AML-BFM 93. Leukemia FAB M3 With the introduction of all-trans retinoid acid (ATRA) in the year 1994, less EDs due to haemorrhage and/or severe infections occurred in FAB M3 patients in AML-BFM 93. Seven of 22 patients within the patient group treated without ATRA (AMLBFM 87 and during the year 1993) died early, but only one of 22 patients treated with ATRA.26 The 5-year OS and EFS of the children receiving ATRA and chemotherapy were significantly better than those in conventionally treated children (survival: 8779 vs 45711%, Plogrank ¼ 0.003; EFS: 76711 vs 43711%, Plogrank ¼ 0.02). Down syndrome Children with Down syndrome were not included in the patient group with de novo AML presented above.27 Compared to the non-Down children, these patients had been treated in the 80s mostly with a less intensive therapy, and up to 48% of them had not been treated with specific AML therapy because their therapy tolerance was thought to be low and high toxicity was expected. Improved survival in paediatric AML, BFM strategy U Creutzig et al Since AML-BFM 93, treatment recommendations were given (ie less intensive treatment like in non-Down children of the standard-risk group and no CNS irradiation). In AML-BFM 93, only 14% of children with Down syndrome got no treatment.27 Before 1993, in the total Down syndrome group (including nontreated patients), outcome was poor (5-year survival 2577%, in patients treated with chemotherapy 55714%). In AML-BFM 93, the survival probability increased to 5377% (in patients treated with chemotherapy 6778%). Discussion General results The results of the four AML-BFM trials presented above indicate that a stepwise increase in long-term survival could be achieved by more intensive chemotherapy applied during induction and early intensification. Compared to the previous study AML-BFM 78, the major improvement was seen after the introduction of the ADE induction in AML-BFM 83. The relapse rate was reduced mainly for the group of children with low-risk factors. Later on, in AML-BFM 93, also the relapse rate in high-risk patients decreased as a result of a more intensive induction and intensification therapy. Relapse remains the main reason for failure in AML therapy. However, prevention of any other event is also of great importance in AML considering the relatively high number of deaths in aplasia and fatalities in CCR. In AML-BFM 83, the main risk factors for ED by leukostasis and/or haemorrhage were identified (initial hyperleukocytosis and especially FAB M5 type and concomitant hyperleukocytosis and/or extramedullary organ involvement).28 Consequently, in the next trials, recommendations for early exchange transfusion and slow blast cell reduction before starting induction treatment were made to avoid these complications. Death in aplasia during or after intensive treatment occurred in about 4–6% of patients. Some of them were also categorised as NR because they died during the period when achievement of CR would have been possible. Some of these patients were NR with resistant disease (remaining blasts 45 to 410%). However, most patients were aplastic or hypoplastic, with insufficient regeneration of haematopoieses; this is why these cases should be assigned to the category ‘aplastic death’ and not to the group of NR. It might be possible to reduce the number of patients dying in aplasia by improving supportive care and treatment conditions like in stem cell transplant units. Under these circumstances, these patients may have a chance to achieve a late remission even after 42 up to 150 days. The number of treatment-related deaths in CCR has remained constant at a range of 3–4% over all studies since 1978, which is remarkable when keeping in mind that the intensity of treatment has increased a lot. Nearly all of these patients – referred to as failures in CCR – died of severe infections during intensive treatment or shortly after the end of intensification.29,30 Transplant-related deaths in first CR occurred in 10% of this particular patient group. In conclusion, the possibility of improving results by further intensification of conventional chemotherapy may be limited because acute and late toxicity is already on threshold value. However, there may be options to further reduce fatalities which are not caused by progression of leukaemia. AML-BFM-specific therapy elements 2039 Compared to other AML studies in children and adults, there are some particular therapy elements in the AML-BFM studies. Maintenance therapy was included since AML-BFM 78: The results of this first trial using 2 years of maintenance after an 8-week induction/consolidation regimen combined with CNS irradiation showed a long-term survival of 3974% after 10 years, which was much better than that expected in the 70s. The duration of maintenance therapy was reduced to 1.5 years in AML-BFM 87 without inferior results for patients surviving at least 1.5 years. Studies in adults showed that maintenance benefited patients treated with less intensive induction consolidation regimens.15 Paediatric studies, by contrast, showed that maintenance was not beneficial.31,32 As outcome with or without maintenance depends on initial therapy, its significance has to be clarified in the BFM setting by randomisation in a future trial. A specific characteristic of the BFM trials is the general use of prophylactic CNS irradiation. Only AML-BFM 87 compared patients with and without CNS irradiation. Results indicated that CNS irradiation is an essential therapy element of the BFM protocols. Outcome was inferior in the nonirradiated groups, especially due to bone marrow recurrences, but also due to higher incidence of isolated or combined CNS relapses. These results suggested that chemotherapy alone could destroy leukaemic blasts in the bone marrow, but with less success in the CNS. Residual blasts from the CNS may reseed the bone marrow and lead to bone marrow relapses. Therefore, CNS irradiation has remained a therapy element in the AML-BFM studies. However, in the ongoing AML-BFM trial 98, a dose reduction is tested prospectively (randomisation of CNS irradiation with 12 vs 18 Gy) in order to reduce late sequelae. SCT in first remission from a matched related donor has been recommended in high-risk patients since AML-BFM 87. However, an advantage of SCT could not be proven in our patients so far.33 SCT in first CR has remained an option in our trials, because further improvement is expected. Results by intensification The main difference between AML-BFM 93 and the previous trial 87 consisted rather in the introduction of the HAM combination and its scheduling than in the application of high-dose cytarabine, which in the 87 protocol had been given as late intensification after consolidation in combination with etoposide. Furthermore, all patients of AML-BFM 87 received daunorubicin for induction. The randomisation of idarubicin and daunorubicin during induction showed a significantly better blast cell reduction in the bone marrow on day 15 in the idarubicin group, whereas long-term outcome was similar in both treatment arms22 and was also comparable to that of AML-BFM 87 in standard-risk patients. The second randomisation comparing early and late HAM was restricted to high-risk patients only. By the early application of HAM, an attempt was made to enhance cytotoxic activity and consequently to achieve higher efficacy. There was evidence to suggest that this approach – as compared to a treatment course with lower dose intensity – might overcome resistance by more rapid blast cell clearance and cause a reduction in the rate of minimal residual disease. The randomisation of early vs late HAM was carried out to evaluate whether or not a possibly improved blast cell reduction Leukemia Improved survival in paediatric AML, BFM strategy U Creutzig et al 2040 in consequence of the early application might be counterbalanced by more toxicity after the course. HAM treatment, however, was not scheduled for standard-risk patients because of the expected higher rate of acute adverse events and possible late cardiotoxicity associated with additional cardiotoxic drugs. The cumulative dose of anthracyclines, including the anthracycline analogue mitoxantrone (assuming a dose ratio of 5:1 for daunorubicin:mitoxantrone), was 300 mg/m2 in standard-risk and 400 mg/m2 in high-risk patients. Treatment with HAM has been shown to be an effective, though toxic, therapy element in adults and children with AML.16,34,35 The dose effect of cytarabine given either at a standard (100 mg/m2), intermediate (400 mg/m2) or high dose (3 g/m2) during post-remission treatment was first shown by the CALGB Study.36 Arlin et al 34 reported a higher CR rate after a single induction course of the mitoxantrone-based regimen (3 12 mg/m2) than after the standard regimen using daunorubicin (3 45 mg/m2) in adult patients with de novo AML. Büchner et al16,37 demonstrated in adults that HAM given as second induction course benefited poor-risk patients. With a 5-year estimated survival of 5772% and an EFS of 5072%, the results of AML-BFM 93 were significantly better for the total group of patients than for those of the previous trial AML-BFM 87 and similar to those of the Medical Research Council (MRC) 10 trial in children38 and the Nordic Society for Paediatric Haematology and Oncology (NOPHO) AML trial.39 This improvement can most probably be attributed to the intensification with HAM in high-risk patients (two-thirds of our patients). The effect of dose scheduling and dose intensity during postremission treatment was demonstrated by the Children’s Cancer Group (CCG) 213P Study. Two courses of high-dose cytarabine/ asparaginase administered at 7-day intervals resulted in better survival rates compared to 28-day intervals.40 Furthermore, in the CCG Study 2861, patients on intensive timing during induction (second cycle 10 days after the first cycle) had a significantly better DFS than patients on standard timing (14 days or later depending on bone marrow status).41 Another study in adults, reported by Estey et al,42 demonstrated that time to achieve remission was an important predictor of survival and DFS. This supports the hypothesis that rapid blast clearance may avoid the development of resistance. Results of the randomised scheduling of HAM as the second or third treatment course after induction did not reveal major differences in outcome. However, the treatment given for induction had to be considered as well. Induction with idarubicin, as opposed to daunorubicin, was more effective in reducing the blast cell count in the bone marrow on day 15.22 Patients who received the less-intensive daunorubicin treatment during induction had a benefit from early HAM.24 Moreover, when comparing high-risk patients of the historical control group of AML-BFM 87 to high-risk patients treated initially with daunorubicin followed by late HAM, it became obvious that results were similar (5-year pEFS: high-risk, AMLBFM 87 ¼ 3173% vs AML-BFM 93 late HAM after induction with daunorubicin ¼ 3677%, P ¼ 0.54). This finding suggests that there might be a cumulative effect of induction with idarubicin and HAM in high-risk patients. These results are in line with the German AMLCG trial in adults, showing that mainly poor-risk patients benefit more from a two-course induction combining TAD with HAM as the second course than from two TAD courses.16 The incidence of therapy-related deaths and infections was similar in early HAM and late HAM, indicating that this adult therapy was feasible also in children with AML. Leukemia As improved treatment results in AML-BFM 93 in children with high-risk AML were attributed mainly to the introduction of HAM and as the rate of toxicity was tolerable, HAM was introduced in AML-BFM 98 as second therapy course for the vast majority of patients (excluding M3 and Down syndrome children), aiming at a better survival rate for standard-risk patients, too. Risk groups The risk definition used in the AML-BFM trials is based on results of AML-BFM 83 and 87.11 It allows (a) a risk-adapted treatment for standard-risk patients, reducing the risk of treatment mortality and severe late toxicity. In AML-BFM 93, these patients received a lower cumulative anthracycline dose and no allogeneic SCT in first CR. (b) For high-risk patients, more investigational therapy elements are justified during induction and consolidation to increase the rate and quality of remission, and in addition, allogeneic SCT in first remission from a matched related donor is recommended. (c) In case of hyperleukocytosis or extramedullary organ involvement, the definition of a third, very-high-risk group is possible. However, at present there are no curative treatment options available, and the risk of ED, including death at diagnosis, is high for this group. In other AML studies in children and adults, mostly cytogenetic results are used for defining risk groups. Most investigators agree that the favourable karyotypes are t(8;21), t(15;17) and inv16.31,38,43,44 These karyotypes are closely related to favourable morphology.11 We could demonstrate that morphology in combination with blast cell reduction on day 15 can be used as a prognostic parameter identifying a standard-risk group. This group includes the known favourable cytogenetic subtypes, but, in addition, 38% more patients with normal or other karyotypes who show the same good prognosis (Figure 9). In children with high-risk criteria and favourable karyotypes prognosis is excellent, indicating that these children can be shifted to the standardrisk group. Using this feature additionally in our studies, we can show that only high-risk patients with unfavourable cytogenetics do Figure 9 Estimated probability of event-free survival in patients with standard and high risk combined with favourable (fav.) or unfavourable (unfav.) cytogenetics, 5-year data given (slash: see Figure 4). Improved survival in paediatric AML, BFM strategy U Creutzig et al 2041 worse compared to others (standard-risk patients with either favourable or unfavourable cytogenetics and high-risk patients with favourable cytogenetics; Figure 9). This led to the current risk group definition, which includes cytogenetics (Figure 3). However, it has been reported in recently published studies including patients of AML-BFM 93 and 98 that FLT3 internal tandem duplications (FLT3-ITDs) predict poor clinical outcome.45,46 This was especially noticeable in the group of standard risk patients (as defined above); therefore, in the future, FLT3-ITDs-positive patients from the standard risk group will be reclassified to the high-risk group (FAB M3 excluded). Promyelocytic leukaemia – FAB M3 Outcome could be improved especially in children with M3 due to treatment with the new agent all-trans-retinoid acid (ATRA), which was introduced for these patients in our trials in 1994. It could be shown that ATRA combined with intensive chemotherapy can not only induce a stable, continuous remission but also avoid ED in these children with considerable but manageable toxic side effects.26 However, further improvements are necessary; one example in this direction may be the use of arsenic trioxide in combination with chemotherapy, which recently showed similar remission rates and minimal toxicity.47 Perspective of further improvements In the 80s, our data indicated that it was possible to improve prognosis in standard-risk patients by intensification of initial chemotherapy. In the last 10 years, prognosis could be increased in high-risk patients by more intensive therapy, for example, by high-dose cytarabine courses. This may indicate that these patients might even benefit from further treatment intensification. However, this has to be done without risking therapy-related mortality. Before starting AML-BFM 98, this was tried to increase in a pilot study with a dosage of 3 14 mg/m2 idarubicin during induction; however, results showed a relatively high toxicity.48 Reduction of the treatment-related mortality which averages out at more than 10% in high-risk patients may be of the same importance. For all patients, therapy schedules with less toxic drugs (eg less cardiotoxic drugs) and less toxic regimens (by limiting the indications for SCT in first remission to the very high-risk patients) will spare more patients late toxicities. Acknowledgements This work is supported by Deutsche Krebshilfe e.v. Principal investigators of the AML-BFM trials in different countries are given in Supplementary Information. Supplementary Information Supplementary Information accompanies the paper on the Leukemia website (http://www.nature.com/leu). References 1 Riehm H, Gadner H, Welte K. Die West-Berliner Studie zur Behandlung der akuten lymphoblastischen Leukämie des Kindes – Erfahrungsbericht nach 6 Jahren. Klin Pädiatr 1977; 189: 89–102. 2 Scheer U, Schellong G, Riehm H. Verbesserte Prognose der akuten myeloischen Leukämien bei Kindern nach intensivierter Anfangstherapie. Klin Pädiatr 1979; 191: 210–216. 3 Creutzig U, Ritter J, Riehm H, Langermann H-J, Henze G, Kabisch H et al. Improved treatment results in childhood acute myelogenous leukemia: a report of the German cooperative study AML-BFM-78. Blood 1985; 65: 298–304. 4 Rai KR, Holland JF, Glidewell OJ, Weinberg V, Brunner K, Obrecht JP et al. Treatment of acute myelocytic leukemia: a study by Cancer and Leukemia Group B. Blood 1981; 58: 1203–1212. 5 Büchner T, Urbanitz D, Hiddemann W, Rühl H, Ludwig WD, Fischer J et al. Intensified induction and consolidation with or without maintenance chemotherapy for acute myeloid leukemia (AML): two multicenter studies of the German AML Cooperative Group. J Clin Oncol 1985; 3: 1583–1589. 6 Creutzig U, Ritter J, Schellong G. Identification of two risk groups in childhood acute myelogenous leukemia after therapy intensification in the study AML-BFM-83 as compared with study AMLBFM-78. Blood 1990; 75: 1932–1940. 7 Baehner RL, Bernstein ID, Sather H, Higgins G, McCreadie S, Chard RL et al. Contrasting benefits of two maintenance programs following identical induction in children with acute nonlymphocytic leukemia: a report from the Childrens Cancer Study Group. Cancer Treat Rep 1984; 68: 1269–1272. 8 Pinkel D, Simone J, Hustu HO, Aur RJ. Nine years’ experience with ‘total therapy’ of childhood acute lymphocytic leukemia. Pediatrics 1972; 50: 246–251. 9 Dahl GV, Simone JV, Hustu HO, Mason C. Preventive central nervous system irradiation in children with acute nonlymphocytic leukemia. Cancer 1978; 42: 2187–2192. 10 Creutzig U, Hofmann J, Ritter J, Schellong G. Therapierealisierung und Komplikationen in der Therapiestudie BFM-83 für die akute myeloische Leukämie. Klin Pädiatr 1988; 200: 190–199. 11 Creutzig U, Zimmermann M, Ritter J, Henze G, Graf N, Löffler H et al. Definition of a standard-risk group in children with AML. Br J Haematol 1999; 104: 630–639. 12 Carella AM, Berman E, Maraone MP, Ganzina F. Idarubicin in the treatment of acute leukemias. An overview of preclinical and clinical studies. Haematologica 1990; 75: 159–169. 13 Berman E, McBride M. Comparative cellular pharmacology of daunorubicin and idarubicin in human multidrug-resistant leukemia cells. Blood 1992; 79: 3267–3273. 14 Hollingshead LM, Faulds D. Idarubicin. A review of its pharmacodynamic and pharmacokinetic properties, and therapeutic potential in the chemotherapy of cancer. Drugs 1991; 42: 690–719. 15 Kesavan S, Lincoff AM, Young JB. Anthracycline-induced cardiotoxicity. Ann Intern Med 1996; 125: 47–58. 16 Büchner T, Hiddemann W, Wörmann B, Löffler H, Gassmann W, Haferlach T et al. Double induction strategy for acute myeloid leukemia: the effect of high-dose cytarabine with mitoxantrone instead of standard-dose cytarabine with daunorubicin and 6-thioguanine: a randomized trial by the German AML Cooperative Group. Blood 1999; 93: 4116–4124. 17 Bennett JM, Catovsky D, Daniel MT, Flandrin G, Galton DAG, Gralnick HR et al. Proposed revised criteria for the classification of acute myeloid leukemia. Ann Intern Med 1985; 103: 626–629. 18 Bennett JM, Catovsky D, Daniel MT, Flandrin G, Galton DAG, Gralnick HR et al. Criteria for the diagnosis of acute leukemia of megakaryocyte lineage (M7). A report of the French–American– British Cooperative Group. Ann Intern Med 1985; 103: 460–462. 19 Bennett JM, Catovsky D, Daniel MT, Flandrin G, Galton DA, Gralnick HR et al. Proposal for the recognition of minimally differentiated acute myeloid leukaemia (AML-M0). Br J Haematol 1991; 78: 325–329. 20 Cheson BD, Cassileth PA, Head DR, Schiffer CA, Bennett JM, Bloomfield CD et al. Report of the National Cancer Institutesponsored workshop on definitions and response in acute myeloid leukemia. J Clin Oncol 1990; 8: 813–819. 21 The National Cancer Institute. Investigators Handbook, A Manual for Participants in Clinical Trials of Investigational Agents Sponsored by the Division of Cancer Treatment. The National Cancer Institute, Washington, 1986. 22 Creutzig U, Ritter J, Zimmermann M, Hermann J, Gadner H, Blütters-Sawatzki R, et al, for the AML-BFM Study Group. Idarubicin improves blast cell clearance during induction therapy Leukemia Improved survival in paediatric AML, BFM strategy U Creutzig et al 2042 23 24 25 26 27 28 29 30 31 32 33 34 Leukemia in children with AML: results of study AML-BFM 93. Leukemia 2001; 15: 348–354. Creutzig U, Berthold F, Boos J, Fleischhack G, Gadner H, Gnekow A et al. Improved treatment results in children with AML: results of study AML-BFM 93]. Klin Pädiatr 2001; 213: 175–185. Creutzig U, Ritter J, Zimmermann M, Reinhardt D, Hermann J, Berthold F, et al, for the AML-BFM Study Group. Improved treatment results in high risk pediatric AML patients after intensification with high dose Ara-C and mitoxantrone: results of study AML-BFM 93. J Clin Oncol 2001; 19: 2705–2713. Reinhardt D, Thiele C, Creutzig U. Neuropsychological sequelae in children with AML treated with or without prophylactic CNS-irradiation. Klin Pädiatr 2002; 214: 22–29. Mann G, Reinhardt D, Ritter J, Hermann J, Schmitt K, Gadner H et al. Treatment with all-trans retinoid acid in acute promyelocytic leukemia reduces early deaths in children. Ann Hematol 2001; 80: 417–422. Creutzig U, Reinhardt D, Diekamp S, Dworzak M, Stary J, Zimmermann M. AML patients with Down syndrome have a high cure rate with AML-BFM therapy with reduced dose intensity. Leukemia 2005; 19: 1355–1360. Creutzig U, Ritter J, Budde M, Sutor A, Schellong G. Early deaths due to hemorrhage and leukostasis in childhood acute myelogenous leukemia: associations with hyperleukocytosis and acute monocytic leukemia. Cancer 1987; 60: 3071–3079. Creutzig U, Zimmermann M, Reinhardt D, Dworzak M, Stary J, Lehrnbecher T. Early deaths and treatment-related mortality in children undergoing therapy for acute myeloid leukemia: analysis of the multicenter clinical trials AML-BFM 93 and AML-BFM 98. J Clin Oncol 2004; 22: 4384–4393. Lehrnbecher T, Varwig D, Kaiser J, Reinhardt D, Klingebiel T, Creutzig U. Infectious complications in pediatric acute myeloid leukemia: analysis of the prospective multi-institutional clinical trial AML-BFM 93. Leukemia 2004; 18: 72–77. Perel Y, Auvrignon A, Leblanc T, Vannier JP, Michel G, Nelken B, et al., for the Group LAME of the French Society of Pediatric Hematology and Immunology. Impact of addition of maintenance therapy to intensive induction and consolidation chemotherapy for childhood acute myeloblastic leukemia: results of a prospective randomized trial, LAME 89/91. J Clin Oncol 2002; 20: 2774–2782. Steuber CP, Civin C, Krischer J, Culbert S, Ragab A, Ruymann FB et al. A comparison of induction and maintenance therapy for acute nonlymphocytic leukemia in childhood: Results of a Pediatric Oncology Group Study. J Clin Oncol 1991; 9: 247–258. Creutzig U, Reinhardt D. Current controversies: which patients with acute myeloid leukaemia should receive a bone marrow transplantation? A European view. Br J Haematol 2002; 118: 365–377. Arlin Z, Case-DC J, Moore J, Wiernik P, Feldman E, Saletan S et al. Randomized multicenter trial of cytosine arabinoside with mitoxantrone or daunorubicin in previously untreated adult patients with acute nonlymphocytic leukemia (ANLL). Lederle Cooperative Group. Leukemia 1990; 4: 177–183. 35 Bishop JF, Matthews JP, Young GA, Szer J, Gillett A, Joshua D et al. A randomized study of high-dose cytarabine in induction in acute myeloid leukemia. Blood 1996; 87: 1710–1717. 36 Mayer RJ, Davis RB, Schifffer CA, Berg DT, Powell BL, Schulman P et al. Intensive postremission chemotherapy in adults with acute myeloid leukemia. N Engl J Med 1994; 331: 896–903. 37 Büchner T, Hiddemann W, Löffler G, Gassmann W, Maschmeyer G, Heit W et al. Improved cure rate by very early intensification combined with prolonged maintenance chemotherapy in patients with acute myeloid leukemia: data from the AML Cooperative Group. Semin Hematol 1991; 28: 76–79. 38 Stevens RF, Hann IM, Wheatley K, Gray RG, on behalf of the MRC Childhood Leukaemia Working Party. Marked improvements in outcome with chemotherapy alone in paediatric acute myeloid leukaemia: results of the United Kingdom Medical Research Council’s 10th AML trial. Br J Haematol 1998; 101: 130–140. 39 Lie SO, Abrahamsson J, Clausen N, Forestier E, Hasle H, Hovi L et al. Treatment stratification based on initial in vivo response in acute myeloid leukaemia in children without Down’s syndrome: results of NOPHO-AML trials. Br J Haematol 2003; 122: 217–225. 40 Wells RJ, Woods WG, Lampkin BC, Nesbit ME, Lee JW, Buckley JD et al. Impact of high-dose cytarabine and asparaginase intensification on childhood acute myeloid leukemia: a report from the Children’s Cancer Group. J Clin Oncol 1993; 11: 538–545. 41 Woods WG, Kobrinsky N, Buckley J, Neudorf S, Sanders J, Miller L et al. Intensively timed induction therapy followed by autologous or allogeneic bone marrow transplantation for children with acute myeloid leukemia or myelodysplastic syndrome: a Children’s Cancer Group pilot study. J Clin Oncol 1993; 11: 1448–1457. 42 Estey EH, Shen Y, Thall PF. Effect of time to complete remission on subsequent survival and disease-free survival time in AML, RAEB-t, and RAEB. Blood 2000; 95: 72–77. 43 Dastugue N, Payen C, Lafage Pochitaloff M, Bernard P, Leroux D, Huguet Rigal F et al. Prognostic significance of karyotype in de novo adult acute myeloid leukemia. The BGMT group. Leukemia 1995; 9: 1491–1498. 44 Haferlach T. More individual markers are necessary for patients with acute myeloid leukemia (AML). Leukemia 1996; 10 (Suppl 3): S5–S9. 45 Liang DC, Shih LY, Hung IJ, Yang CP, Chen SH, Jaing TH et al. FLT3-TKD mutation in childhood acute myeloid leukemia. Leukemia 2003; 17: 883–886. 46 Zwaan CM, Meshinchi S, Radich JP, Veerman AJ, Huismans DR, Munske L et al. FLT3 internal tandem duplication in 234 children with acute myeloid leukemia: prognostic significance and relation to cellular drug resistance. Blood 2003; 102: 2387–2394. 47 George B, Mathews V, Poonkuzhali B, Shaji RV, Srivastava A, Chandy M. Treatment of children with newly diagnosed acute promyelocytic leukemia with arsenic trioxide: a single center experience. Leukemia 2004; 18: 1587–1590. 48 Creutzig U, Körholz D, Niemeyer CM, Kabisch H, Graf N, Reiter A et al. 3 14 mg idarubicin during induction. Results of a pilot study in children with AML. Leukemia 2000; 14: 340–342.
© Copyright 2025 Paperzz