• 沒有找到結果。

Improved treatment results for childhood acute myeloid leukemia in Taiwan 

N/A
N/A
Protected

Academic year: 2021

Share "Improved treatment results for childhood acute myeloid leukemia in Taiwan "

Copied!
6
0
0

加載中.... (立即查看全文)

全文

(1)

ORIGINAL ARTICLE

Improved treatment results for childhood acute myeloid leukemia in Taiwan

D-C Liang1, T-T Chang2, K-H Lin3, D-T Lin3, M-Y Lu3, S-H Chen1, H-C Liu1, M-T Lin4, M-T Lee4, S-G Shu5, T-K Chang5, J-S Chen6, C-C Hsiao7, I-J Hung8, Y-L Hsieh9, R-L Chen10, S-N Cheng11, W-H Chang12, C-H Lee12and K-S Lin3,12

1Pediatric Hematology-Oncology Division, Mackay Memorial Hospital, Taipei, Taiwan;2Pediatric Hematology-Oncology Division, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan;3Pediatric Hematology-Oncology Division, National Taiwan University Hospital, Taipei, Taiwan;4Pediatric Hematology-Oncology Division, Changhua Christian Hospital, Changhua, Taiwan;5Pediatric Hematology-Oncology Division, Veterans General Hospital at Taichung, Taichung, Taiwan;6Pediatric Hematology-Oncology Division, National Cheng Kung University Hospital, Tainan, Taiwan;7Pediatric Hematology-Oncology Division, Chang Gung Children’s Hospital at Kaohsiung, Kaohsiung, Taiwan;8Pediatric Hematology-Oncology Division, Chang Gung Children’s Hospital at Linkou, Linkou, Taiwan;9Pediatric Hematology-Oncology Division, Veterans General Hospital at Taipei, Taipei, Taiwan;10Pediatric Hematology-Oncology Division, Buddhist Tzu Chi General Hospital, Hualien, Taiwan; 11

Pediatric Hematology-Oncology Division, Tri-Service General Hospital, Taipei, Taiwan and12Taiwan Childhood Cancer Foundation, Taipei, Taiwan

To improve treatment results for children with de novo acute myeloid leukemia (AML), we introduced a novel protocol, Taiwan Pediatric Oncology Group-AML-97A, for AML other than acute promyelocytic leukemia (APL), for which modified conventional protocols were used. From January 1, 1997, to December 31, 2002, 141 children younger than 17 years old with de novo AML were enrolled. In total, 117 patients with non-APL AML were treated with induction therapy of idarubicin and cytarabine (Ara-C), postremission therapy with high-dose Ara-C – containing regimens for four monthly courses, and moderate-dose therapy with idarubicin and Ara-C for four monthly courses. The first 19 patients with APL were treated with all-trans retinoic acid, idarubicin and Ara-C, with the remaining five patients receiving all-trans retinoic acid and idarubicin, followed by maintenance therapy for 2 years. Stem cell trans-plantation was performed in 29 patients in first remission with a similar outcome as chemotherapy alone. The remission rate in the AML-97A study was 90%, the 5-year survival 5175.3% (s.e.) and the 5-year event-free survival 5074.8%; for APL, these were 100%, 8677.0, and 7579.8%. For the whole group, the 5-year survival was 5774.7% and the 5-year event-free survival 5474.4%. The AML-97A regimen was well tolerated.

Leukemia (2006) 20, 136–141. doi:10.1038/sj.leu.2403979; published online 10 November 2005

Keywords: childhood acute myeloid leukemia; treatment; outcome

Introduction

Several treatment components have contributed to the recent improvement in treatment for de novo childhood acute myeloid leukemia (AML). A combination of cytarabine (Ara-C) and anthracycline remains the most frequently used remission induction therapy. Mitoxantrone has shown to be an active agent against AML.1The addition of etoposide to some regimens has also improved the treatment of AML.2 Another crucial development is the introduction of high-dose Ara-C (HDAra-C) in either the postremission or induction phase of therapy, for both adults3,4and children5with AML. The beneficial effect of HDAra-C during postremission therapy was first demonstrated by the Cancer and Leukemia Group B,4while Hiddemann et al.6

reported the effectiveness of HDAra-C in combination with mixtoxantrone (HAM) in adults with refractory AML. Other innovations contributing to an improved outcome in AML were described in numerous recent reports.7–15 Besides agents that increase the antileukemic efficacy of modern AML treatments, advances in molecular biology have led to the recognition of prognostically and therapeutically relevant subtypes of AML, while improved supportive care, consisting of more effective antimicrobials (especially the antifungal agents), blood compo-nent therapy, hematopoietic stem cell transplantation, and antiemetics, has prevented excessive toxic deaths due to intensive chemotherapy.

Here we report the improved results of recent Taiwan Pediatric Oncology Group (TPOG) protocols for children and adolescents with de novo AML, which incorporated many of the treatment components deemed to be important in other successful studies.

Patients and methods

Eligibility

Studies TPOG AML-97A, TPOG APL-97, and TPOG-APL-2001 enrolled children under 18 years of age with newly diagnosed de novo AML, after written informed consent from the guardians. While patients with Down’s syndrome or systemic disease with chloroma were included, those with secondary AML or myelodysplastic syndrome were excluded. In our nationwide TPOG studies from January 1, 1997, to December 31, 2002, all patients with APL were enrolled in the TPOG-APL-97 study and then the TPOG-APL-2001 study. AML patients without APL were treated with either the TPOG-97A study according to the participating institutions, or the TPOG-97B study; the latter study was modified from MRC AML109and will be reported separately. Hematopoietic stem cell transplantation, when feasible, was performed in 29 patients. The study results were censored on December 31, 2004.

Diagnosis and classification

The initial diagnosis of AML, including the subtyping, was established according to the French–American–British (FAB) classification.16–18Diagnosis of the M0 and M7 subtypes was confirmed by immunologic methods.17,18The karyotypes were

Received 24 August 2005; accepted 30 August 2005; published online 10 November 2005

Correspondence: Dr K-S Lin, Taiwan Blood Services Foundation, 3F, No. 3, Nan-Hai Road, Taipei 100, Taiwan.

E-mail: kslin@blood.org.tw

(2)

interpreted according to the International System for Human Cytogenetics Nomenclature.19Common fusion transcripts were detected by reverse transcriptase (RT) PCR assays, followed by Southern blot analysis as described previously.20The diagnosis and classification of each case were also evaluated according to the new classification system of the World Health Organiza-tion.21

Treatment

TPOG-AML-97A protocol. The TPOG-AML-97A protocol consisted of induction therapy with idarubicin (9 mg/m2/ day  3) and Ara-C (100 mg/m2/day  7) every 3 weeks. If a remission could not be induced after two courses, second-line treatment including mitoxantrone (8 mg/m2/day  5) and etopo-side (100 mg/m2/day  5) every 4 weeks was given. Cranial irradiation was not used for either prophylaxis or treatment of central nervous system leukemia (CNSL) at diagnosis. Instead, intrathecal methotrexate (IT MTX) was administered to all patients (including those with APL) on day 1 of each course. The doses were 6, 8, 10, 12, and 15 mg for patients agedo1, 1–2, 2–3, 3–9, and 49 years, respectively. Once complete remission was achieved, postremission therapy was begun, consisting of four monthly courses of Ara-C (1 g/m2/12 h on days 1–4) and etoposide (100 mg/m2/day  5) alternating with Ara-C (1 g/m2/12 h on days 1–4) and mitoxantrone (10 mg/m2/day on days 2–5) if absolute neutrophils exceeded 1.5  109/l and platelets 100  109/l. Four monthly courses of idarubicin (9 mg/ m2/day  1) and Ara-C (200 mg/m2/day  5) followed. IT MTX was administered on day 1 of the four courses at the dose schedule described above. The median duration of treatment was 10 months.

TPOG-APL-97 protocol. All-trans retinoic acid (ATRA, 30 mg/m2/day) was started on day 1. Idarubicin and Ara-C (administered as described for TPOG-AML-97A) were given if the leukocyte count exceeded 5  109/l before treatment, 6  109/l on day 5, 10  109/l on day10, or 15  109/l on day 15 of induction therapy. The postremission therapy consisted of six monthly courses of idarubicin and Ara-C (as described above). IT MTX was administered on day 1 of each course at the same dose schedule described above. In all, 19 patients were enrolled.

TPOG-APL-2001 protocol. This treatment plan was mod-ified from the PETHEMA LPA 96 protocol,22and was activated in 2001. It started with ATRA (25 mg/m2/day) and idarubicin (9 mg/m2/day  3). After remission induction, consolidation therapy with three monthly courses of idarubicin (9 mg/m2/ day  3) was instigated. IT MTX was given on day 1 of each idarubicin course at a dose schedule described before. This phase of therapy was followed by 2 years of maintenance therapy comprising 6-mercaptopurine (90 mg/m2/day PO), MTX (15 mg/m2/week PO), and ATRA (25 mg/m2/day  15 every 3 months). Five patients were enrolled.

Hematopoietic stem cell transplantation

Hematopoietic stem cell transplantation, including allogeneic bone marrow transplantation, allogeneic peripheral blood stem cell transplantation, autologous bone marrow transplantation, or autologous peripheral blood stem cell transplantation, when feasible, was performed during first remission.

Supportive care

Granulocyte-colony-stimulating factor (G-CSF, 200 mg/m2/day SC) was administered each day during marrow aplasia induced by induction chemotherapy and prophylactically over 36–48 h after the end of each HDAra-C-containing course. Bactrim (trimethoprim-sulfamethoxazole) for prophylaxis of Pneumocys-tis carinii pneumoniPneumocys-tis was given 3 days a week beginning 2 weeks after the initiation of induction therapy and continuing to 6 months after the completion of chemotherapy. The strategy in preventing and treating infectious complications applied the same principles as used in BFM group.23 Rasburicase,24 a recombinant urate oxidase, has been available for effective prevention and treatment of leukemia lysis syndrome for the recent 2 years.

Statistical methods

The primary end points of this study were overall survival (OS), event-free survival (EFS), and disease-free survival (DFS). OS was measured from the start of treatment to death from any causes, and EFS from the start of treatment to first progression, relapse or death from any cause. Patients not achieving a first remission were assigned an EFS of zero. DFS extended from the time of transplantation to the time of failure. Patients who did not fail were censored at the time of their last follow-up. Survival curves were estimated by the Kaplan–Meier method. Prognostic factor analysis was performed by univariate analysis using the two-sided log-rank test, and multivariate analysis using the Cox regression model. A statistically significant difference was classified as a P-value o0.05. Survival rates were represented as the mean percent (7s.e.) probability estimates.

Results

A total of 243 children were enrolled in the TPOG-AML studies, of whom 12 girls and 12 boys had APL and 219 non-APL AML. In the latter group, 117 patients (56 girls, 61 boys) were enrolled in study TPOG-AML-97A and 102 in study TPOG-AML-97B (not included in this report). Overall, the ages of the patients ranged from 0 to 17 years old, with 32 patients older than 13 years. In this subgroup, 17 patients were older than 15 years. In all, 68 of the patients were girls and 73 were boys. In the study TPOG-AML-97A, 61 patients presented with a leukocyte count o20  109/l, 56 had counts 20–100  109/l, while 21 had counts X100  109/l. In the TPOG-APL studies, there were 23 patients with a leukocyte count o50  109/l, and only one patient in APL-97 had a count 450  109/l. The distribution of phenotype is reported in Table 1. The most frequent subtypes in decreasing order were M2, M3, M5, M1, and M4. Genetic classification showed after the t(15;17)/PML-RARa, the leading abnormalities were t(8;21)/AML1-ETO, MLL rearrangements, and inv(16)/CBFb-MYH11. Three patients had Down’s syn-drome. Two patients had monosomy 7, and five patients had complex chromosomal abnormalities. In all, 56 patients had a normal karyotype or other cytogenetic changes.

In TPOG-AML-97A, 90% attained complete remission, compared with 100% of those with APL. The remission rate was 91% overall. Of 29 patients who underwent hematopoietic stem cell transplantation in first remission, 17 had allogeneic bone marrow donor and six an allogeneic peripheral blood stem cell donor. Other procedures used were two autologous bone marrow and four autologous peripheral blood stem cells. In the AML-97A study, 31 patients relapsed: 26 in the bone marrow, two in the isolated CNS, two in both bone marrow and CNS, and one in bone marrow and breast. All three relapses in APL

(3)

patients were in the bone marrow: two in APL-97 and one in APL-2001. In all, 56 patients in 97A have died, 30 with refractory or relapsed leukemia, 25 of infection, and one of hemorrhage. Four patients in APL-97 study died, three of infection, one of leukemia, and none of hemorrhage.

Figure 1 shows the EFS and OS rates for all 141 patients regardless of disease subtype. The 5-year EFS estimate was 5474.4%, with a 5-year OS of 5774.7%. One patient relapsed very late, at 25 months after the initiation of treatment, and died 29 months later. The 117 patients enrolled in TPOG-AML-97A had a 5-year EFS rate of 5074.8%, and a 5-year OS of 5175.3% (Figure 2). Overall, the 5-year OS and EFS rates for patients with APL were 8677.6 and 7579.8%, respectively. The DFS rate for patients undergoing hematopoietic stem cell transplantation were 6079.5%, compared with 6875.4% for those receiving chemotherapy alone (P ¼ 0.63). Table 1 shows the distribution of patients and their survival by FAB subtype. Outcome was clearly better among patients with an M2, M3 or M6 subtype, and clearly worse among those with an M7 or M0 subtype. Of the genetic features examined (Table 1), trisomy 21, t(15;17)/PML-RARa, inv(16)/CBFb-MYH11, t(8;21)/AML1-ETO, and t(9;11)/MLL-AF9 were associated with a better outcome, while MLL rearrangements other than t(9;11)/MLL-AF9 con-ferred a poor prognosis. The 56 patients with a normal karyotype or other cytogenetic changes had a 5-year OS rate of 48%. Table 2 lists the complications that were encountered during these studies. Sepsis, other infections, and febrile neutropenia were common, especially in study AML-97A. Notably, there were no episodes of cardiotoxicity of the three studies. None of the treatments produced excessive toxicity.

Among the factors analyzed, only high leukocyte count X100  109/l and poor early response (attaining complete

1.0 0.8 0.6 0.4 0.2 0.0 Probability 0 12 24 36 48 60 72 84 96 Months EFS: 54 ± 4.4% OS: 57 ± 4.7%

Figure 1 5-year event-free survival (EFS) and overall survival (OS) rates for all 141 patients with AML.

1.0 0.8 0.6 0.4 0.2 0.0 Probability 0 12 24 36 48 60 72 84 96 Months EFS: 50 ± 4.8% OS: 51 ± 5.3%

Figure 2 5-year event-free survival (EFS) and overall survival (OS) rates for 117 children with non-APL, treated with AML-97A protocol.

Table 1 Patient characteristics and treatment outcome

Category No. of patients 5-years EFS (s.e.) P-value 5-years OS (s.e.) P-value Overall 138 54 (4.4) 57 (4.7) Gender 0.683 0.988 Male 71 56 (6.2) 58 (6.2) Female 67 52 (6.2) 55 (7.0) Age 0.418 0.197 o2 years 29 54 (9.4) 54 (9.4) 2–9 years 61 61 (6.6) 68 (6.3) 10–14 years 34 47 (8.9) 51 (9.2) 15–17 years 14 38 (14.8) 42 (13.4) WBC(109/l) 0.011 0.003 o20 61 53 (6.8) 58 (7.2) 20–100 56 63 (6.5) 66 (6.6) X100 21 30 (10.3) 30 (10.3) FAB 0.194 0.076 M0 8 29 (17.3) 29 (17.1) M1 14 52 (14.6) 48 (15.7) M2 43 54 (7.6) 59 (7.7) M3 23 75 (9.8) 86 (7.6) M4 13 40 (15.6) 51 (14.7) M5 21 51 (11.1) 51 (11.1) M6 6 83 (15.2) 83 (15.2) M7 10 33 (15.7) 22 (17.8) Genetic 0.470 0.322 inv(16) 4 50 (4.9) 75 (9.2) t(15;17) 21 67 (11.2) 79 (9.2) t(8;21) 19 63 (11.2) 67 (11.2) t(9;11) 2 100 100 Other MLL 7 29 (17.1) 29 (8.9) Trisomy 21 3 100 100 Monosomy 7 2 50 (35.4) 50 (35.4) Complex 5 60 (21.9) 60 (22.0) Normal and others 56 48 (7.1) 48 (7.9) Cyclea 0.001 0.002 1 42 75 (7.0) 80 (6.3) 41 96 45 (5.3) 47 (5.7) CNSL 0.710 0.821 Absent 122 55 (4.7) 57 (5.1) Present 16 46 (13.1) 52 (13.2) APL APL97 18 75 (11.0) 81 (9.7) APL2001 5 67 (27.2) 100 Overall 23 75 (9.8) 86 (7.6) a

To achieve complete remission.

Table 2 Complications in studies TPOG-97A and TPOG-APL

Complication No. of episodes

97A (n¼ 117) APL-97 (n¼ 19) APL-2001 (n¼ 5) Total (n¼ 141) Sepsis 152 15 2 169 Other infection 481 62 19 562 Febrile neutropenia 386 47 11 444 Hemorrhage 41 15 4 60 Cardiotoxicity 0 0 0 0 138

(4)

remission after two or more courses of therapy) were associated with poor OS and EFS (Table 1).

Discussion

The 5-year EFS of 5474.4% achieved in studies TPOG-AML-97A, TPOG-APL-97, and TPOG-APL-2001 were at least comparable to the 31–54% attained in studies conducted in other developed countries during the same era,25–37 and the treatment was well tolerated. However, like in the Dutch Childhood Oncology Group experience, a more effective, but less toxic, therapy and better supportive care guidelines for childhood AML are still needed.38For remission induction, the use of idarubicin with Ara-C on a ‘3 þ 7’ schedule was based on a report from Cancer and Leukemia Group B, which demon-strated that this regimen is better than a ‘2 þ 5’ regimen.39 Idarubicin was selected for remission induction therapy because of its superiority over daunorubicin in treatment. In study AML-BFM93, induction with idarubicin was more effective than daunorubicin in reducing the blast cells in bone marrow on day 15.10Moreover, meta-analysis of results from randomized trials comparing idarubicin, mitoxantrone, and daunorubicin re-vealed a statistically significant improvement in remission rate with idarubicin, and a trend toward a better rate with mitoxantrone compared to daunorubicin (Wheatley K. Blood, 1995;86:43; abstract). When combined with Ara-C (100 mg/m2/ day  7), idarubicin was more effective than daunorubicin in remission induction.40,41A 9 mg/m2/day schedule of idarubicin was selected because of prior experience in Taiwan, indicating that higher doses of idarubicin resulted in unacceptable morbidity and mortality in pediatric patients.

Ara-C at 100 mg/m2/day  7, rather than 200 mg/m2/day  7 or HDAra-C, was chosen for induction therapy because increases in Ara-C doses have not consistently resulted in a higher remission rate, but have contributed to increased toxicity, particularly in older patients.42 For example, randomized studies in adults with AML found no significant differences in the induction rate at doses raising from 100 to 200 mg/m2.43 Further dose increases, by even 20–30 times (i.e., to 3 g/m2 every 12 h for 8–12 doses), have not significantly improved the remission rates.44,45

Cardiotoxicity due to intensive chemotherapy is of great concern in the treatment of AML patients. Idarubicin, for example, has a reduced cardiotoxic index by comparing with either daunorubicin or doxorubicin.46,47 In animal studies, the therapeutic index related to chronic cardiotoxicity favors idarubicin over daunorubicin by a factor of about 3.46In a retro-spective study, a cumulative dose of idarubicin at 150 mg/m2 was estimated to be safe, while 290 mg/m2 was considered probably safe.48 The therapeutic index of idarubicin versus doxorubicin with regard to chronic cardiotoxicity is estimated to be 1.8–1.9. In study TPOG-AML-97A, patients generally received a cumulative dose of idarubicin 63 mg/m2 and mitoxantrone dose of 80 mg/m2, which is equivalent to a dose of 340 mg/m2 of doxorubicin. The lack of cardiotoxicity in the current TPOG studies supports these calculations of a ‘safe’ cumulative dose.

A third or fourth drug was not added to our induction regimen because the benefits of this modification are uncertain. Some trials have demonstrated an increased remission duration with the addition of etoposide to the ‘3 þ 7’ regimen.2 However, another study by the Cancer and Leukemia Group B did not observe an increased remission rate or duration of remission with the addition of 6-thioguanine to ‘3 þ 7’.49 Since the

combination of mitoxantrone and etoposide in refractory AML is both effective and well tolerated,50we reserved this regimen for second-line induction therapy.

Some studies have achieved dose intensification, in part, by extending the period of induction therapy as a part of consecutive daily treatments. This practice has resulted in a trend toward slightly higher remission induction rates.9 An alternative strategy is based on the concept that leukemic blasts could be effectively recruited into S phase of the cell cycle by following the initial course of chemotherapy with a 6- to 8-day gap before reinstitution of therapy. This type of timed, sequential, or intensively timed therapy originated from work on leukemic cell cycle kinetic studies.51For example, the CCG 2891 trial tested a ‘standard’ versus ‘intensive’ timing induction using the five-drug dexamethasone, Ara-C, thioguanine, etopo-side, and rubidomycin (DCTER) regimen.8,52 Standard timing specified that the second course of chemotherapy be delivered after bone marrow recovery, usually at about 30 days unless residual leukemia was present on day 14, in which case therapy was instigated at that time. Intensively timed induction therapy specified the second course of DCTER on days 10–13, regardless of bone marrow status. Complete remission rate for the standard and intensive timing arms were not significantly different, 74 and 78%, respectively.8,53 However, induction mortality was appreciably greater with the intensively timed induction, although there were few cases of refractory leukemia. Recently, the addition of cladribine to daunorubicin and Ara-C was reported to increase the remission induction rate in adults with AML.53

During postremission treatment, the effect of dose scheduling and dose intensification was demonstrated in the Children’s Cancer Group 213P study. Two courses of HDAra-C and asparaginase administered at a 7-day interval resulted in superior survival rates compared with arms administered at a 28-day interval.5Furthermore, in the Children’s Cancer Group study 2861 patients receiving intensive-timing induction chemo-therapy (second cycle 10 days after the first cycle) had a significantly better DFS than did patients receiving standard-timing induction therapy (second cycle 14 days or more after the first cycle, depending on bone marrow status).52In postremis-sion therapy, however, several studies in adults have strongly suggested that increasing the intensity of immediate postremis-sion therapy, especially with HDAra-C, results in a lower relapse rate, with relapse occurring later than in patients receiving less intense maintenance therapy.54 This was espe-cially true for patients with favorable cytogenetics, for example, t(8;21) or inv(16).55

HDAra-C cannot increase the remission rate because primary refractoriness to conventional-dose Ara-C does not arise until the residual leukemic infiltrate has decreased to fewer than 106 cells,56 and the probability that subpopulations of cells will become resistant to Ara-C increases with time.56Thus, it seems more reasonable to use HDAra-C for postremission treatment rather than remission induction.57 In consolidation therapy, HDAra-C has a beneficial effect on long-term outcome that may be similar to the results achieved with bone marrow transplan-tation.57 In adult patients younger than 60 years, induction therapy with mitoxantrone and Ara-C, in randomized doses of 1 or 3 g/m2/dose, resulted in similar remission rates, although CNS toxicity was more frequent in the Ara-C 3 g/m2group than in the 1 g/m2group (79 versus 28%, Po0.10).58This finding promotes the rationale for the use of the Ara-C 1 g/m2/dose rather than 3 g/m2in the present study.

These results show remarkable improvement over a previous TPOG AML study (1990–1996), in which the 5-year OS was

(5)

only 21%, and compare favorably with outcomes during the same era reported from successful treatment centers world-wide.25–37 The challenge now is to extend the benefits of contemporary AML therapy to larger numbers of patients in developing countries, where fewer than 10% children with acute leukemia are receiving protocol-based therapy.59

Acknowledgements

We thank Ching-Hon Pui, MD, St Jude Children’s Research Hospital, Memphis, USA, for his critical review and continuous support, and also Ching-Tien Peng, MD, and Kang-Hsi Wu, MD, China Medical University Hospital, Taichung, Taiwan; Shiann-Tarng Jou, MD, National Taiwan University Hospital, Taipei, Taiwan; Chao-Neng Cheng, MD, National Cheng Kung University Hospital, Tainan, Taiwan; Jiunn-Ming Sheen, MD, Chang Gung Children’s Hospital at Kaohsiung, Kaohsiung, Taiwan; Chao-Ping Yang, MD, and Tang-Her Jaing, MD, Chang Gung Children’s Hospital at Linkou, Linkou, Taiwan; Lin-Yen Wang, MD, Mackay Memorial Hospital, Taipei, Taiwan; Min-Chang Liu, MD, Chi Mei Hospital, Liouying, Taiwan; and Giun-Yi Hung, MD, Veterans General Hospital at Taipei, for Provision of study material. We are very grateful to the Data Managers of the Taiwan Childhood Cancer Foundation: Ms Xiu-Wen Liao, Bee-Rou Wu, Hsiu-Chuan Lin, and Hsiu-E Hsu for data collection. This work was supported by the Taiwan Childhood Cancer Foundation, Taipei, Taiwan.

References

1 Estey EH, Keating MJ, McCredie KB, Bodey GP, Freireich EJ. Phase II trial of mitoxantrone in refractory acute leukemia. Cancer Treat Rep 1983; 67: 389–390.

2 Bishop JF, Lowenthal RM, Joshua D, Matthews JP, Todd D, Cobcroft R, et al., for the Australian Leukemia Study Group. Etoposide in acute nonlymphocytic leukemia: Australian Leukemia Study Group. Blood 1990; 75: 27–32.

3 Bu¨chner T, Hiddemann W, Lo¨ffler 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.

4 Mayer RJ, Davis RB, Schiffer 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.

5 Wells RJ, Woods WG, Lampkin BC, Nesbit ME, Lee JW, Buckley JD et al. Impact of high-dose cytarabine and asparaginase intensifica-tion on childhood acute myeloid leukemia: a report from the Childrens Cancer Group. J Clin Oncol 1993; 11: 538–545. 6 Hiddemann W, Kreutzmann H, Straif K, Ludwig WD, Mertelsmann

R, Donhuijsen-Ant R et al. High-dose cytosine arabinoside and mitoxantrone: a highly effective regimen in refractory acute myeloid leukemia. Blood 1987; 69: 744–749.

7 Ravindranath Y, Yeager AM, Chang MN, Steuber CP, Krischer J, Graham-Pole J et al. Autologous bone marrow transplantation versus intensive consolidation chemotherapy for acute myeloid leukemia in childhood. Pediatric Oncology Group. N Engl J Med 1996; 334: 1428–1434.

8 Woods WG, Kobrinsky N, Buckley JD, Lee JW, Sanders J, Neudorf S et al. Timed-sequential induction therapy improves postremis-sion outcome in acute myeloid leukemia: a report from the Children’s Cancer Group. Blood 1996; 87: 4979–4989.

9 Hann IM, Stevens RF, Goldstone AH, Rees JK, Wheatley K, Gray RG et al. Randomized comparison of DAT versus ADE as induction chemotherapy in children and younger adults with acute myeloid leukemia. Results of the Medical Research Council’s 10th AML trial (MRC AML10). Adult and Childhood Leukaemia Working Parties of the Medical Research Council. Blood 1997; 89: 2311–2318.

10 Creutzig U, Ritter J, Zimmermann M, Reinhardt D, Hermann J, Berthold F et al. Improved treatment results in high-risk pediatric

acute myeloid leukemia patients after intensification with high-dose cytarabine and mitoxantrone: results of Study Acute Myeloid Leukemia – Berlin–Frankfurt–Munster 93. J Clin Oncol 2001; 19: 2705–2713.

11 Krance RA, Hurwitz CA, Head DR, Raimondi SC, Behm FG, Crews KR et al. Experience with 2-chlorodeoxyadenosine in previously untreated children with newly diagnosed acute myeloid leukemia and myelodysplastic diseases. J Clin Oncol 2001; 19: 2804–2811.

12 Perel Y, Auvrignon A, Leblanc T, Vannier JP, Michel G, Nelken B, et al., 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. Leucamie Aique Myeloide Enfant. J Clin Oncol 2002; 20: 2774–2782.

13 O’Brien TA, Russell SJ, Vowels MR, Oswald CM, Tiedemann K, Shaw PJ, et al., Australian and New Zealand Children’s Cancer Study Group. Results of consecutive trials for children newly diagnosed with acute myeloid leukemia from the Australian and New Zealand Children’s Cancer Study Group. Blood 2002; 100: 2708–2716.

14 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 leukemia in children without Down’s syndrome: results of NOPHO-AML trials. Br J Haematol 2003; 122: 217–225. 15 Hann IM, Webb DK, Gibson BE, Harrison CJ. MRC trials in childhood acute myeloid leukemia. Ann Hematol 2004; 83 (Suppl 1): S108–S112.

16 Bennett JM, Catovsky D, Daniel MT, Flandrin G, Galton DA, Gralnick HR et al. Proposed revised criteria for the classification of acute myeloid leukemia. Ann Intern Med 1985; 103: 620–625. 17 Bennett JM, Catovsky D, Daniel MT, Flandrin G, Galton DA,

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. 18 Bennett JM, Catovsky D, Daniel MT, Flandrin G, Galton DA, Gralnick HR et al. Proposal for the recognition of minimally differentiated acute myeloid leukemia (AML-M0). Br J Haematol 1991; 78: 325–329.

19 Mitelman F. An International System for Human Cytogenetic Nomenclature. Karger: Basel, Switzerland, 1995.

20 Liang DC, Shih LY, Yang CP, Hung IJ, Chen SH, Liu HC. Molecular analysis of fusion transcripts in childhood acute myeloid leukemia in Taiwan. Med Pediatr Oncol 2001; 37: 555–556.

21 Vardiman JW, Harris NL, Brunning RD. The World Health Organization (WHO) classification of the myeloid neoplasms. Blood 2002; 100: 2292–2302.

22 Sanz MA, Martin G, Rayon C, Esteve J, Gonzalez M, Diaz-Mediavilla J et al. A modified AIDA protocol with anthracycline-based consolidation results in high antileukemic efficacy and reduced toxicity in newly diagnosed PML/RARalpha-positive acute promyelocytic leukemia. PETHEMA group. Blood 1999; 94: 3015–3021.

23 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.

24 Jeha S, Kantarjian H, Irwin D, Shen V, Shenoy S, Blaney S et al. Efficacy and safety of rasburicase, a recombinant urate oxidase (Elitekt), in the management of malignancy-associated hyperur-emia in pediatric and adult patients: final results of a multicenter compassionate use trial. Leukemia 2005; 19: 34–38.

25 Pession A, Rondelli R, Basso G, Rizzari C, Testi AM, Fagioli F, et al., on behalf of the AML Strategy & Study Committee of the Associazione ltaliana Ematologia Oncologia Pediatrica (AIEOP). Treatment and long-term results in children with acute myeloid leukaemia (AML) treated according to the AIEOP AML protocols. Leukemia 2005; 19: 2043–2053.

26 Creutzig U, Zimmermann M, Ritter J, Reinhardt D, Hermann J, Henze G, et al., for the AML-BFM Study Group. Treatment strategies and long-term results in paediatric patients treated in four consecutive AML-BFM trials. Leukemia 2005; 19: 2030–2042. 27 Smith FO, Alonzo TA, Gerbing RB, Woods WG, Arceci RJ, for the Childre n’ s Cancer Group. Long-term results of children with

(6)

acute myeloid leukemia: a report of three consecutive phase III trials by the Children’s Cancer Group: CCG 251, CCG213 and CCG 2891. Leukemia 2005; 19: 2054–2062.

28 Quintana J, Advis P, Becher A, Beresi V, Campbell M, Vine´s EF, et al., for the Programa Infantil de Drogas Antineopla´sicas de Chile (PINDA). Acute myelogenous leukemia in Chile PINDA protocols 87 and 92 results. Leukemia 2005; 19: 2143–2146.

29 Kardos G, Zwaan CM, Kaspers GJL, de Graaf SSN, de Bont ESJM, Postma A et al. Treatment strategy and results in children treated on three Dutch Childhood Oncology Group acute myeloid leukemia (AML) trials. Leukemia 2005; 19: 2063–2071.

30 Entz-Werle N, Suciu S, Van Der Werff Ten Bosch J, Vilmer E, Bertrand Y, Benoit Y, et al., on behalf of EORTC Children Leukemia Group (CLG). Results of 58872 and 58921 trials in acute myeloblastic leukemia (AML) and relative value of chemotherapy vs allogeneic bone marrow transplantation (alloBMT) in 1st complete remission: the EORTC Children Leukemia Group report. Leukemia 2005; 19: 2072–2081.

31 Armendariz H, Fernandez Barbieri MA, Freigeiro D, Lastiri F, Felice MS, Dibar E, on behalf of the GATLA Group, Buenos Aires, Argentina. Treatment strategy and long-term results in pediatric patients treated in two consecutive AML-GATLA trials. Leukemia 2005; 19: 2139–2142.

32 Perel Y, Auvrignon A, Leblanc T, Michel G, Reguerre Y, Vannier J-P, et al., for the Group LAME of the Socie´te´ Franc¸aise des Cancers de l ’ Enfant (SFCE), France. Treatment of childhood acute myeloblastic leukemia. Dose intensification improves outcome and maintenance therapy is of no benefit. Multicenter studies of the French LAME (Leuce´mie Aigue¨ Mye´loblastique Enfant) Cooperative Group. Leukemia 2005; 19: 2082–2089.

33 Lie SO, Abrahamsson J, Clausen N, Forestier E, Hasle H, Hovi L, et al., on behalf of the Nordic Society of Pediatric Hematology and Oncology (NOPHO). Long-term results in children with AML: NOPHO-AML Study Group-Report of three consecutive trials. Leukemia 2005; 19: 2090–2100.

34 Ravindranath Y, Chang M, Steuber CP, Becton D, Dahl G, Civin C, et al., for the Pediatric Oncology Group. Pediatric Oncology Group (POG) studies of acute myeloid leukemia (AML): a review of four consecutive childhood AML trials conducted between 1981 and 2000. Leukemia 2005; 19: 2101–2116.

35 Dluzniewska A, Balwierz W, Armata J, Balcerska A, Chybicka A, Kowalczyk J, et al., for the Polish Pediatric Leukemia/Lymphoma Study Group (PPLLSG). Twenty years of Polish experience with three consecutive protocols for treatment of childhood acute myelogenous leukemia. Leukemia 2005; 19: 2117–2124. 36 Ribeiro RC, Razzouk BI, Pounds S, Hijiya N, Pui C-H, Rubnitz JE.

Successive clinical trials for childhood acute myeloid leukemia at St. Jude Children’s Research Hospital, 1980 through 2000. Leukemia 2005; 19: 2125–2129.

37 Gibson BES, Wheatley K, Hann IM, Stevens RF, Webb D, Hills RK, et al., for the United Kingdom Childhood Leukaemia Working Party and the Dutch Childhood Oncology Group. Treatment strategy and long-term results in paediatric patients treated in consecutive UK AML Trials. Leukemia 2005; 19: 2130–2138. 38 Slats AM, Egeler RM, van der Does-van den Berg A, Korbijn C,

Hahlen K, Kamps WA et al. Causes of death-other than progressive leukemia – in childhood acute lymphoblastic leukemia (ALL) and myeloid leukemia (AML): the Dutch Childhood Oncology Group experience. Leukemia 2005; 19: 537–544.

39 Rowe JM, Tallman MS. Intensifying induction therapy in acute myeloid leukemia: has a new standard of care emerged? Blood 1997; 90: 2121–2126.

40 Wiernik PH, Banks PL, Case Jr DC, Arlin ZA, Periman PO, Todd MB et al. Cytarabine plus idarubicin of daunorubicin as induction and consolidation therapy for previously untreated adult patients with acute myeloid leukemia. Blood 1992; 79: 313–319.

41 Vogler WR, Velez-Garcia E, Weiner RS, Flaum MA, Bartolucci AA, Omura GA et al. A phase III trial comparing idarubicin and daunorubicin in combination with cytarabine in acute myelogen-ous leukemia: a Southeastern Cancer Study Group Study. J Clin Oncol 1992; 10: 1103–1111.

42 Stasi R, Venditti A, Del Poeta G, Aronica G, Abruzzese E, Pisani F et al. High-dose chemotherapy in adult acute myeloid leukemia: rationale and results. Leuk Res 1996; 20: 535–549.

43 Dillman RO, Davis RB, Green MR, Weiss RB, Gottlieb AJ, Caplan S et al. A comparative study of two different doses of cytarabine for acute myeloid leukemia: a phase III trial of Cancer and Leukemia Group B. Blood 1991; 78: 2520–2526.

44 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.

45 Weick JK, Kopecky KJ, Appelbaum FR, Head DR, Kingsbury LL, Balcerzak SP et al. A randomized investigation of high-dose versus standard-dose cytosine arabinoside with daunorubicin in patients with previously untreated acute myeloid leukemia: a Southwest Oncology Group study. Blood 1996; 88: 2841–2851.

46 Casazza AM, Dı´Marco A, Bonadonna G, Bonfante G, Bertazzoli C, Bellini D et al. Effects of modifications in position 4 of the chromophore or in position 40of the aminosugar, on the antitumor

activity and toxicity of daunorubicin and doxorubicin. In: Crooke ST, Reich SD (eds). Anthracyclines, Current Status and New Developments. Academic Press: New York, 1980, pp 403–430. 47 Chen ZM, Colombo T, Conforti L, Grazia Donelli M, Fiedorowicz

RJ, Marchi S et al. Effects of three new anthracyclines and doxorubicin on the rat isolated heart. J Pharm Pharmacol 1987; 39: 947–950.

48 Anderlini P, Benjamin RS, Wong FC, Kantarjian HM, Andreeff M, Kornblau SM et al. Idarubicin cardiotoxicity: a retrospective study in acute myeloid leukemia and myelodysplasia. J Clin Oncol 1995; 13: 2827–2834.

49 Preisler H, Davis RB, Kirshner J, Dupre E, Richards III F, Hoagland HC, et al., and the Cancer and Leukemia Group B. Comparison of three remission induction regimens and two postinduction strategies for the treatment of acute nonlymphocytic leukemia: a Cancer and Leukemia Group B study. Blood 1987; 69: 1441–1449.

50 Ho AD, Lipp T, Ehninger G, Illiger HJ, Meyer P, Freund M et al. Combination of mitoxantrone and etoposide in refractory acute myelogenous leukemia – an active and well-tolerated regimen. J Clin Oncol 1988; 6: 213–217.

51 Burke PJ, Owens Jr AH. Attempted recruitment of leukemic myeloblasts to proliferative activity by sequential drug treatment. Cancer 1971; 28: 830–836.

52 Woods WG, Kobrinsky N, Buckley J, Neudorf S, Sanders J, Miller L et al. Intensively timed induction therapy followed by auto-logous or allogeneic bone marrow transplantation for children with acute myeloid leukemia or myelodysplastic syndrome: a Childrens Cancer Group pilot study. J Clin Oncol 1993; 11: 1448–1457.

53 Holowiecki J, Grosicki S, Robak T, Kyrcz-Krzemien S, Giebel S, Hellmann A, et al., Polish Adult Leukemia Group (PALG). Addition of cladribine to daunorubicin and cytarabine increases complete remission rate after a single course of induction treatment in acute myeloid leukemia. Multicenter, phase III study. Leukemia 2004; 18: 989–997.

54 Bu¨chner T, Urbanitz D, Hiddemann W, Ruhl 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.

55 Mayer RJ, Davis RB, Schiffer CA, Berg DT, Powell BL, Schulman P et al. Intensive postremission chemotherapy in adults with acute myeloid leukemia. Cancer and Leukemia Group B. N Engl J Med 1994; 331: 896–903.

56 Momparler RL. A model for the chemotherapy of acute leukemia with l-b-D-arabinofranosylcytosine. Cancer Res 1974; 34: 1775–1787.

57 Hiddemann W. Cytosine arabinoside in the treatment of acute myeloid leukemia: the role and place of high-dose regimens. Ann Hematol 1991; 62: 119–128.

58 Hiddemann W, Aul C, Maschmeyer G, Schonrock-Nabulsi R, Ludwig WD, Bartholomaus A et al. High-dose versus intermediate-dose cytarabine combined with mitoxantrone for the treatment of relapsed and refractory acute myeloid leukemia: results of an age-adjusted randomized comparison. Sem Hematol 1991; 28 (Suppl 4): 35–38.

59 Ribeiro RC, Pui CH. Saving the children – improving childhood cancer treatment in developing countries. N Engl J Med 2005; 332: 2158–2160.

數據

Figure 1 5-year event-free survival (EFS) and overall survival (OS) rates for all 141 patients with AML.

參考文獻

相關文件

In this paper, we report a case of a patient who reported for bleeding gums to our department and was diagnosed with acute myeloid leukaemia incidentally on routine blood

Myeloperoxidase and leukocyte nonspecific esterase (NSE) special stain were used which showed presence of myeloblasts in the peripheral smear suggestive of acute myelocytic

Loss of vascular content, increase of fat in the bone marrow cavity, and fibrosis showed a linear relation with time after radiation and were considered the end stage of

Up-and-coming mandibular reconstruction technique with autologous human bone marrow stem cells and iliac bone graft in patients with large

The simulation results of the DWT method and the least-squares matrix inversion method in x and y direction are shown in Table 1, The computer simulation results of the

In this paper, we provide new decidability and undecidability results for classes of linear hybrid systems, and we show that some algorithms for the analysis of timed automata can

Table 7: Resident population born outside Macao by total years of residence in Macao c (2001 ). Total

MR CLEAN: A Randomized Trial of Intra-arterial Treatment for Acute Ischemic Stroke. • Multicenter Randomized Clinical trial of Endovascular treatment for Acute ischemic stroke in