Vous êtes sur la page 1sur 8

Ann Hematol (2005) 84: 557–564

DOI 10.1007/s00277-005-1046-0

ORIGINA L ARTI CLE

A. Wrzesień-Kuś . T. Robak . A. Wierzbowska . E. Lech-Marańda . A. Pluta .


E.Wawrzyniak . A. Krawczyńska . K. Kuliczkowski . G. Mazur . M. Kiebiński .
A. Dmoszyńska . M. Wach . A. Hellmann . W. Baran . J. Hołowiecki .
S. Kyrcz-Krzemień . S. Grosicki

A multicenter, open, noncomparative, phase II study


of the combination of cladribine (2-chlorodeoxyadenosine),
cytarabine, granulocyte colony-stimulating factor
and mitoxantrone as induction therapy in refractory acute
myeloid leukemia: a report of the Polish Adult Leukemia Group
Received: 8 January 2005 / Accepted: 2 April 2005 / Published online: 27 April 2005
# Springer-Verlag 2005

Abstract Purine nucleoside analogues, cladribine (2-chlo- toxicity of induction treatment consisting of 2-CdA (5 mg/
rodeoxyadenosine, 2-CdA) and fludarabine (FAMP) are m2), Ara-C (2 g/m2), mitoxantrone (MIT, 10 mg/m2) and
active agents in acute myeloid leukemias (AMLs). Syner- granulocyte colony-stimulating factor (G-CSF) (CLAG-M)
gistic interaction between FAMP or 2-CdA with cytarabine in refractory AML. In case of partial remission, a second
(cytosine arabinoside, Ara-C) has been demonstrated in CLAG-M was administered. Patients in complete remission
preclinical and clinical studies. The current multicenter (CR) received consolidation courses based on high-dose
phase II study was initiated to evaluate the efficacy and Ara-C and MIT with or without 2-CdA. Forty-three patients
from five centers were registered: 25 primary resistant and
18 relapsed. CR was achieved in 21 (49%) patients, 20
(47%) were refractory and 2 (5%) died early. Hematologic
A. Wrzesień-Kuś . T. Robak (*) . A. Wierzbowska . toxicity was the most prominent toxicity of this regimen.
E. Lech-Marańda . A. Pluta . E. Wawrzyniak . The overall survival (OS; 1 year) for the 42 patients as a
A. Krawczyńska
Department of Hematology, whole and the 20 patients in CR were 43% and 73%,
Medical University of Lódz, respectively. Disease-free survival (1 year) was 68.6%.
ul. Pabianicka 62, None of the analyzed prognostic factors influenced the CR
93-513 Lódz, Poland and OS probability significantly. We conclude that CLAG-
e-mail: robaktad@csk.am.lodz.pl
Tel.: +48-426-895191 M regimen has significant antileukemia activity in refrac-
Fax: +48-426-895192 tory AML, which seems to be better than the activity of
many other regimens. The toxicity of the treatment is
K. Kuliczkowski . G. Mazur . acceptable.
M. Kiebiński
Department of Hematology,
Medical University, Keywords Cladribine . 2-Chlorodeoxyadenosine . Purine
Wrocław, Poland nucleoside analogs . Mitoxantrone . Refractory AML .
Induction treatment
A. Dmoszyńska . M. Wach
Department of Hematology,
Medical University,
Lublin, Poland Introduction

Despite the significant progress made in the treatment of


A. Hellmann . W. Baran
Department of Hematology, acute myeloid leukemia (AML) in the last decade, 20–
Medical University, 40% of patients still do not achieve complete remission
Gdańsk, Poland (CR) with standard induction chemotherapy and 50–70%
of patients in first CR (CR1) may be expected to relapse
J. Hołowiecki . S. Kyrcz-Krzemień . S. Grosicki
Department of Hematology and Bone Marrow Transplantation, [8, 33]. Patients with primary refractory disease and with
Silesian Medical University, early relapses following a CR1 of less than 6 months in
Katowice, Poland duration have a significantly poorer response to therapy
558

and overall outcomes when compared with patients in re- Patients and methods
lapse after a CR1 of more than 6 months in duration (CR
rate 28% vs. 59%) [23]. Most salvage therapies that have Patient selection
been studied in patients with refractory or relapsed AML
are combination regimens using various doses of cytosine Patients were considered eligible if they (1) had been di-
arabinoside (Ara-C) with a variety of other agents. The agnosed as having refractory AML, (2) were in perfor-
number of patients achieving CR with these regimens is mance status ≤2 according to the WHO scale and (3) had a
low (16–44%), with usually short disease-free survival life expectancy of ≥6 weeks. Ineligibility criteria were (1)
(DFS) and overall survival (OS) [1, 2, 6, 12, 20, 23, 27, AML subtype M3 according to the French–American–
32, 38]. British (FAB) classification, (2) uncontrolled infection, (3)
The purine analogs, cladribine (2-chlorodeoxyadeno- hepatic or renal insufficiency and (4) refusal to participate
sine, 2-CdA) and fludarabine (FAMP), represent a relative- in the study.
ly novel group of cytotoxic agents with activity against Refractory AML was defined according to the follow-
AML refractory to conventional treatment [9, 14, 24, 26, ing criteria: (1) primary resistance to initial induction ther-
34]. Estey et al. developed chemotherapy consisting of apy, (2) first early relapse with a remission duration of less
FAMP, Ara-C and granulocyte colony-stimulating factor than 6 months, (3) second or subsequent relapse and (4)
(G-CSF) (FLAG) [10]. Since its original description, the recurrence, after either allogeneic or autologous stem cell
FLAG regimen has been used in a large number of pri- transplantation (SCT) [23, 37]. Written informed consent
marily single-center studies of relapsed or refractory AML, was obtained from all patients before registration. The
with CR rates of 30–81% reported [2, 13, 19, 20, 28, 29, study was approved by a local ethics committee at each
38]. The addition of mitoxantrone (MIT) or idarubicine center.
(IDA) to the FLAG regimen may result in antileukemic
synergism of this combination [4, 7, 16, 30, review in 34].
However, no prospective studies comparing the FLAG Treatment protocol
regimen with FLAG+ anthracycline have been reported to
date. The treatment protocol scheme is described in Fig. 1. Pa-
Both in vitro and ex vivo pharmacological studies dem- tients received induction chemotherapy consisting of 2-
onstrated a 50–65% increase in the rate of Ara-CTP ac- CdA 5 mg/m2, in 2-h i.v. infusions daily, for 5 consecutive
cumulation after pretreatment with 2-CdA [15, 24]. In days; Ara-C 2 g/m2, in 4 h infusions, started 2 h after 2-CdA,
the pilot study of the combination of 2-CdA with high for 5 consecutive days; MIT 10 mg/m2 i.v. for 3 days and G-
doses of Ara-C and G-CSF (CLAG), the CR rate was 50% CSF at a dose of 300 μg s.c., for 6 days, started 24 h before
in primary refractory and relapsed AML patients with CR1 chemotherapy. In cases of white blood cells (WBC) >20
of less than 12 months in duration [32]. These prelimi- G/l, G-CSF and 2-CdA were commenced simultaneously.
nary results have been recently confirmed in a phase II No dose modification was permitted. All other concurrent
multicenter study. In the group of 58 patients, where the treatment and supportive care were administered in ac-
majority of cases had been diagnosed as primary resistant cordance with local practice at the participating centers. In
AML, the CR rate after CLAG regimen was 50%, which cases of partial remission (PR), the second induction
is better than the CR rate achieved with many other reg- course was recommended. Patients not in CR (NR) after
imens in this very poor risk group of patients [39]. two induction courses were withdrawn from the study. If
However, less is known about possible synergistic anti- CR had been achieved, the patient was given two consol-
leukemic activity of MIT and Ara-C in combination with idation courses. In the group of primary refractory patients
2-CdA. So far, 2-CdA with Ara-C and IDA or daunoru- the first consolidation course consisted of MIT 10 mg/m2
bicin (DNR) has been used in the treatment of de novo (days 3–5) and Ara-C 1.5 g/m2 (days 1–3) (HAM). (Fig. 1)
AML or AML in the elderly [18, 21]. No increase in the [18]. In the group of patients with relapsed AML, the first
toxicity has been observed and the CR rate after one in- consolidation course, HAM or another CLAG-M was given
duction course has been higher in the treatment arm with according to the treatment center choice. The second con-
2-CdA, in both the de novo AML group and the elderly solidation course based on high doses (HD) of Ara C with
AML patients. or without 2-CdA in all patients.
These results and the results of our multicenter phase II Patients in CR were qualified for stem cell transplan-
study of CLAG regimen were the rationales for a mul- tation or maintenance therapy.
ticenter phase II study assessing the efficacy and toxicity
of the combination of CLAG regimen with MIT (CLAG-
M) in patients with refractory AML. To the best of our Diagnosis and response criteria
knowledge, these are the first results of a multicenter
analysis of the efficacy of 2-CdA in combination with Patients were considered relapsed when they had >5%
Ara-C, G-CSF and MIT in this poor risk group of AML blasts in the bone marrow (BM) examination and primary
patients. refractory (resistant) if they had >25% blasts or >5% in the
559
CLAG-M without either a del(9q), or being part of a complex kar-
2-CdA 5mg/m2 iv d 1-5
Ara-C 2g/m2 iv d 1-5 yotype. The intermediate risk category included patients
NR
MIT 10 mg/m2 iv d 1-3 characterized by +8, −Y, +6, del(12p) or normal karyo-
G-CSF 300 µg sc d 0-5 Off protocol type. The unfavorable risk category was defined by the
presence of one or more of −5/del(5q), −7/del(7q), inv(3q),
abn11q, 20q, 21q, del(9q), t(6;9), t(9;22), abn 17p and
PR complex karyotype defined as three or more abnormalities.
In the fourth group, patients with cytogenetic aberrations
of unknown prognostic significance were classified.

CLAG-M PR, NR Statistics


Off protocol
Sample size was calculated using the 0.05 level of sig-
nificance and assuming the power of the study at 80%.
The objective was to increase the CR of 20%, after other
historical salvage regimens, to 50% following CLAG-M.
At least 40 patients were to be included. Disease-free
survival (DFS) was defined as the time from the docu-
CR mentation of a CR to the date of relapse or death from any
Consolidation
cause or the last survival and relapse-free date. The du-
ration of overall survival (OS) was measured from the date
of entry into the study to the date of death from any cause.
or Patients surviving at the time of the analysis were censored
HAM
Ara-C 1,5 g/m2 iv d 1-3 CLAG-M on the date they were last known to be alive. The rela-
Mitox 10 mg/m2 iv d 3-5
tionships between qualitative and discrete parameters were
analyzed by use of the chi-square and Mann–Whitney
tests. Survival curves of patients were prepared using the
Kaplan–Meier method, and differences between the groups
in the OS were assessed using the log-rank test. Ninety-five
HiDAC
Ara-C 4 g/m2 iv d 1,3,5 percent confidence intervals (CI) were calculated for DFS
±2-CdA 5 mg/m2 iv d 1,3,5 and OS.

Fig. 1 Treatment schedule with CLAG-M regimen in patients with


refractory AML Results

A total of 43 patients from five centers were registered


BM examination, after the first and second standard in- between November 2000 and September 2004. The clin-
duction treatment, respectively. Response criteria were es- ical and hematological characteristics of the patients are
tablished according to those developed by the National listed in Table 1. There were 21 women and 22 men in the
Cancer Institute-sponsored workshop [3]. CR was defined study group, with the median age of 44 years (range, 20–
as less than 5% blasts in BM and normal peripheral count. 66 years). Twenty-five patients suffered from primary
PR was defined as BM blasts with percentage between 5 refractory AML and 18 patients relapsed; 17 of these
and 25%. NR was defined when these criteria were not subjects were in first relapse, with 1 in second, and among
satisfied. Early death (ED) was defined as death from any relapsed patients 6 relapsed 5–24 months after auto-SCT.
cause during the first 30 days from the commencement of In 2 patients, myelodysplastic syndrome (MDS) preceding
the induction treatment. Hematological and extra hemato- the diagnosis of AML had been documented. The median
logical toxicity were assessed according to the WHO cri- number of WBC count at diagnosis was 25.0 g/l (range
teria. The number of nights spent in hospital and days of 1.4–500.0 g/l), WBC before CLAG-M was 3.9 (range,
i.v. antibiotic therapy during CLAG-M induction course 0.8–131.7) and the median percentage of blasts in BM
were also recorded. before CLAG-M was 39.8 (range, 6–97). Twenty-seven
patients were in the WHO performance status 0–1, and 16
patients in performance status 2. According to the karyo-
Cytogenetic analysis type analysis performed at diagnosis of AML, 21 patients
were assigned to the intermediate and 11 to the unfavor-
Four cytogenetic categories were defined according to the able group; 1 patient had nonclassified abnormalities of
Southwest Oncology Group (SWOG) criteria [35]. The unknown prognostic significance. None of the patients
favorable risk category included patients with inv(16)/del was assigned to the favorable risk group. In the group of
(16q)/t(16;16) with any additional abnormalities, t(8;21) 10 patients, cytogenetics was not available.
560
Table 1 Characteristics of patients Table 2 Treatment outcome after CLAG-M regimen
No. of patients (%) Treatment No. of patients DFS, median OS, median
(n=43) outcome (%) (n=43) (range), weeks (range), weeks

Median age (range), years 44 (20–66) CR 21 (49) 26.2 (0.5–138+) 43 (3–174+)


Sex, F/M 21/22 (n=20) (n=20)
FAB ED 2 (5)
0 4 (10) NR 20 (47) 21.5 (6–79)
1 9 (21) DFS Disease-free survival, OS overall survival, CR complete
2 18 (42) remission, ED early death, NR no remission
4 10 (23)
5 1 (2)
6 1 (2)
Antecedent history CR was achieved in 40% and 61% in each group, re-
MDS/AML 2 (5) spectively (p=NS).
De novo 41 (95) Thirty-five patients had received DNR and Ara-C as the
WHO performance status first-line induction therapy (DA-7); 8 patients had received
0–1 27 (63) additional 2-CdA (DAC-7) during the first-line treatment.
2 16 (37)
The CR rates after CLAG-M regimen were 49% and 50%
WBC (g/l), range
in each group, respectively. CLAG-M regimen was ad-
ministered as a first salvage treatment in 19 out of 25
At diagnosis 25.0 (1.4–500.0)
primary resistant patients, and in 6 patients at least one
Before CLAG-M 3.9 (0.8–131.7)
other salvage regimen was used before CLAG-M. CR rate
Percent of blasts in BM before CLAG-M 39.8 (6–97)
was 42% vs 33% in both groups, respectively (p=NS).
(range)
Cytogenetics at diagnosis
Favorable 0/33 Toxicity associated with CLAG-M regimen
Intermediate 21/33
Unfavorable 11/33 Two patients (5%) died early, both from infections, one of
UN [46,XX t(4;17), t(8;16)] 1/33 them from fungal sepsis. Hematological toxicity and in-
NA 10 fections were the most prominent toxicities of the CLAG-M
Primary resistant (%) 25 (58) induction treatment (Table 4). For patients, who achieved
Relapse (%) 18 (42) CR, the median duration of neutrophils <0.5 g/l and plate-
Second relapse (%) 1/18 lets <20 g/l was 20 days (range, 13–35) and 24 days (range,
Preceding auto-SCT 6/18 12–104), respectively. Patients received a median of 6 units
F Females, M males, MDS myelodysplastic syndrome, WBC white of packed red cells (range, 10–16) and 24 units (range, 3–
blood cells, BM bone marrow, UN karyotype abnormalities of 72) of platelets. The median time of hospitalization was 33
unknown prognostic significance, NA not available days (range, 19–54). All but one patient in the whole group
had fever during the period of aplasia, in 21 (49%) of them
infection grade 3/40 according to the WHO toxicity scale
had been diagnosed (Table 5). The median duration of anti-
Response to CLAG-M induction therapy biotic therapy was 20 days (range, 11–34 days). WHO >2
extrahematological toxicity is summarized in Table 5.
After one induction with CLAG-M regimen, 20 (47%) and
5 (12%) achieved CR and PR, respectively. Three patients
with PR received a second CLAG-M regimen, 1 was Follow-up
withdrawn from the study because of poor performance
status after the first cycle, and 1 patient did not agree for Consolidation therapy
further intensive treatment. Finally CR was achieved in
21 (49%) patients, 20 (47%) were resistant and 2 pa- At the time of analysis 16 of 21 patients in CR received
tients (5%) died early (Table 2). Twelve of 21 patients consolidation therapy, 2 patients received maintenance
(57%) with intermediate karyotype achieved CR; 4 of 11 treatment or allo-SCT directly after CLAG-M regimen, 2
(36%) from the unfavorable prognostic group according to patients are awaiting allo-SCT or first consolidation course
karyotype (p=NS) and 1 patient with karyotype abnor- and 1 patient in CR has been lost from observation. Eleven
malities of unknown prognostic significance achieved CR patients received HAM and 5 CLAG-M as the first con-
(Table 3). In the analyzed group, 25 patients were primary solidation course. All but one patient stayed in CR; 1
resistant to first-line induction therapy and 18 relapsed. patient relapsed after HAM regimen. Nine patients of 15 in
561
Table 3 The efficacy of Risk groups No. of CR (no. of DFS, range OS, range No. of patients (%) treated
CLAG-M regimen according to
karyotype at diagnosis (accord- according to patients patients/%) (weeks) (weeks) with SCT after CLAG-M
ing to SWOG criteria) karyotype

Intermediate 21 12/57 53.8 61 (3–174+) 5 (42)


(2–135+) (n=12)
Unfavorable 11 4/36 7 14.5 (4–90) 1 (25)
CR Complete remission, DFS (0.5–86.5) (n=4)
disease-free survival, OS overall Unknown [46,XX 1 1 16.5 23.5 0
survival, SCT stem cell t(4;17), t (8;16)]
transplantation

CR received HD Ara-C as the second consolidation course Prognostic factors for response to remission
(3 of them with 2-CdA); 3 patients received allo-SCT di- induction therapy
rectly after consolidation 1. Three patients with AML with
relapse after auto-SCT performed in CR1 received main- The following factors were analyzed for prognostic value
tenance therapy after consolidation cycle 1 because of for CR probability: age, WHO performance status, WBC
poor tolerance of intensive treatment; 1 of them died in at diagnosis, WBC and percent of blasts in BM before
CR2. CLAG-M, karyotype abnormalities at diagnosis (interme-
diate vs unfavorable) and number of preceding CLAG-M
salvage regimens. None of these factors significantly in-
Disease-free and overall survival fluenced the probability of CR after salvage treatment with
CLAG-M (p>0.05 for all comparisons in the chi-square
One patient was lost from observation after achievement with Yate’s correction and the Mann–Whitney tests).
of CR with CLAG-M. The median OS for the group of 42
patients was 23.7 weeks (range, 3–174+weeks) and 43
weeks (range, 3–174+weeks) for patients who achieved Prognostic factors for DFS and OS
CR. The probability of 1-year OS for the group as a whole
(42 patients) and the group of 20 patients in CR was 43% The following factors were analyzed according to their in-
(95% CI, 28–58%) and 73% (95% CI, 53.5–92%), re- fluence on OS and DFS probability: age, the WHO per-
spectively (Fig. 2). Median time of DFS was 26.2 weeks formance status, WBC at diagnosis and WBC and percent
(range, 0.5–138+weeks). The probability of 1-year DFS of blasts in BM before CLAG-M and number of preceding
was 68.6% for all 20 patients in CR (95% CI, 49–89%). CLAG-M salvage regimens. Karyotype at diagnosis (inter-
mediate vs unfavorable) was not included in the analysis as
five patients in the group with karyotype of intermediate
Stem cell transplantation prognostic significance had SCT early after induction
treatment (Table 3). None of the analyzed factors influ-
Seven out of 21 patients in CR received SCT, 6 of them as enced the OS probability significantly (p>0.05 for all
the postconsolidation treatment: 5 allogeneic and 2 au- comparisons in the log-rank test).
tologous. Median time from CR achievement to SCT was
17.3 weeks (range, 7–27 weeks). All patients after allo-
SCT are alive and in CR. Both patients treated with auto- Discussion
SCT relapsed after 14 and 30 months; 1 died. Median DFS
in the group of patients treated with SCT, when compared The prognosis of adult patients with AML resistant to first-
with the group treated with chemotherapy alone, was 86.5 line, standard induction therapy or relapsed after CR1 of
weeks (range, 62–138+) and 16.5 weeks (range, 0.5–74+),
respectively. Median OS was 90 weeks (range, 69–174+) Table 5 Nonhematologic toxicity (WHO >2)
in group 1 and 23.5 weeks (range, 3–116+) in group 2.
Toxicity (WHO 3/4) No. of patients (%) (n=43)

Nausea/vomiting 5 (12)
Table 4 Hematopoietic recovery and supportive treatment in Diarrhoea 7 (16)
patients who achieved CR after CLAG-M regimen (n=21)
Mucositis/stomatitis 8 (19)
Days for neutrophils <0.5 g/l 20 (13–35) Hepatotoxicity 1 (2)
Days for platelets <20 g/l 24 (12–104) Alopecia 15 (35)
Packed red cells units (range) 6 (10–16) Cardiotoxicity 2 (4)
Platelets units (range) 24 (3–72) Haemorrhages 2 (4)
Days of hospitalization (range) 33 (19–54) Allergy 1 (2)
Days of antibiotic therapy 20 (11–34) Infection/fever of unknown origin 21 (49)
562
Fig. 2 Overall survival (OS) Overall survival
probability in the group of 42 1,0
refractory AML patients after
CLAG-M induction treatment 0,9

0,8

0,7

Proportion surviving
0,6

0,5

0,4

0,3

0,2

0,1

0,0
0 50 100 150 200
Weeks

less than 6 months’ duration is very poor. The average CR the results of the FLAG–Ida regimen in the group of 12
rates in this group of patients are estimated for 10–20% patients with resistant AML or AML relapsed after CR of
with less than 10% probability for OS of 52 weeks [11]. less than 12 months’ duration. The CR rate was only 33%.
AML relapsed after SCT or AML in second or subsequent Steinmetz et al. [36] reported the results of the treatment of
relapse has similar poor prognosis [23, 37]. There is no refractory, relapsed and secondary AML with the FLAG–
consensus on how to manage this poor-risk refractory Ida regimen. CR was achieved by 1 of 14 patients with
AML. Most salvage therapies are based on the combina- refractory AML, defined as primary resistant or relapsed
tion regimens, using various doses of Ara-C, the most within 6 months of CR1. Clavio et al. [4] treated with FLAG
active drug in AML, with a variety of other agents [1, 6, or FLAG with MIT (FLANG) protocols 8 patients with
12, 15, 17, 23, 25, 27, 37]. refractory AML and 9 with AML relapsed within 12 months
Our group has recently reported the results of a phase II from diagnosis or in second relapse. The CR rate was 33%
study of the use of 2-CdA, Ara-C and G-CSF (CLAG in the refractory group and 75% in the group with relapsed
regimen) in poor-prognosis refractory AML. The results of AML.
that study indicated that the CLAG regimen is an effective In our group of 43 patients, 8 received 2-CdA in com-
and well-tolerated induction treatment. CR was achieved bination with DNR and Ara-C (DAC-7) as the first-line
in 29 (50%) patients, 19 (33%) were refractory and 10 induction chemotherapy. They were primary refractory to
(17%) died early [39]. this induction regimen and were subsequently enrolled to
The only studies so far assessing the toxicity and ef- the CLAG-M therapy as the salvage, second-line treat-
ficacy of the combination of 2-CdA, Ara-C and IDA or ment. Four (50%) of 8 patients achieved CR. This obser-
DNR have been performed by Juliusson et al. [21] in the vation is different from our former study, where 2-CdA in
group of elderly AML and by Hołowiecki et al. [18] in the first-line treatment was the only one adverse prognos-
patients below 60 years of age with de novo AML. No tic factor for the CR probability after CLAG [39]. It cannot
increase in the toxicity has been observed and the CR rate be excluded that the addition of MIT to the CLAG reg-
after one induction course has been higher in the treatment imen significantly improved treatment results in this group
arm with 2-CdA, both in de novo AML group and in the of patients.
elderly AML patients. However, there are no existing data Karyotype abnormalities did not significantly influence
so far concerning the efficacy and toxicity of similar treat- the treatment results. This is similar to the observation
ment combination in refractory AML. With the obser- made by others [4, 7, 36]. However, it is worth mentioning
vations in mind concerning CLAG regimen, we had the that none of our patients with known karyotype at diag-
possibility to evaluate specifically the relative safety and nosis belonged to the group of favorable prognosis and
efficacy of the CLAG-M protocol in the similar group of 30% of patients belonged to the group of poor prognosis
refractory AML patients. The association of CLAG with according to the SWOG criteria. This is different from our
MIT did not increase the CR rate, when retrospectively former study with CLAG, in which only 16% of patients
compared to the CLAG regimen (49% vs 50%, respec- belonged to the poor-prognosis group according to the
tively). However, the CR rate of 49% in this very poor risk karyotype and 15% of patients had favorable cytogenetic
group of patients deserves further attention and is still better abnormalities [39]. Although the small number of patients
than the CR rate achieved with many other regimens. In precludes any conclusion in this regard, the 50% CR rate
some studies, MIT or IDA has been added to the FLAG after salvage treatment with CLAG-M in the group of
regimen [4, 20, 22, 28, 30]. De la Rubia [7] et al. reported patients with intermediate and unfavorable prognosis ac-
563

cording to karyotype abnormalities seems to be the result 4. Clavio M, Carrara P, Miglino M, Pierri I, Canepa L, Balleari E,
worth further attention. Gatti AM, Cerri R, Celesti L, Vallebella E, Sessarego M,
Patrone F, Ghio R, Damasio E, Gobbi M (1996) High efficacy
Age was not a significant prognostic factor for CR of fludarabine containing therapy (FLAG-FLANG) in poor risk
probability and survival in this study. However, only two acute myeloid leukemia. Haematologica 81:513–520
patients in our group were older than 60 years, and nine 5. Clavio M, Venturino C, Pierri I, Garrone A, Miglino M, Canepa
patients were older than 55 years. In this latter group, the L, Balleari E, Balocco M, Michelis GL, Ballerini F, Gobbi M
(2004) Combination of liposomal daunorubicin (DaunoXome),
CR rate was 22% as compared with the CR rate of 53% in fludarabine, and cytarabine (FLAD) in patients with poor-risk
younger patients (p=NS; data not shown). acute leukemia. Ann Hematol 83:696–703
DFS and OS probability of 1 year in this study’s group 6. Cortes J, Estey E, Beran M, O’Brien S, Giles F, Koller C,
of 20 patients in CR was 68.6 and 73%, respectively, Keating M, Kantarjian H (2000) Cyclophosphamide, ara-C and
topotecan (CAT) for patients with refractory or relapsed acute
which is much better than the results obtained with other leukemia. Leuk Lymphoma 36:479–484
salvage regimens or CLAG [1, 6, 12, 18, 24, 28, 38]. 7. De la Rubia J, Regadera AA, Martin G, Cervera J, Sanz G,
However, 7 of 20 patients received SCT as consolidation Martinez J, Jarque I, Garcia I, Andreu R, Moscardo F, Jimenez
or postconsolidation phase of treatment, and DFS in the C, Molla S, Benlloch L, Sanz M (2002) FLAG-IDA regimen
group of patients not treated with SCT was 48%. (fludarabine, cytarabine, idarubicin and G-CSF) in the treat-
ment of patients with high-risk myeloid malignancies. Leuk
Two (5%) of 43 patients treated with CLAG-M died Res 26:725–730
early, both from infections. This is an unexpectedly low 8. Edenfield WJ, Gore SD (1999) Stage-specific application of
mortality rate as compared with other intensive salvage allogeneic and autologous marrow transplantation in the man-
regimens, among them FLAG–Ida or FLAG–Mito and agement of acute myeloid leukemia. Semin Oncol 26:21–34
9. Estey E, Plunkett W, Gandhi V, Rios MB, Kantarjian H,
CLAG (11%–22,8%) [7, 31, 36, 39]. However, Clavio et al. Keating MJ (1993) Fludarabine and arabinosylcytosine therapy
described recently the treatment results of FLAD regimen of refractory and relapsed acute myelogenous leukemia. Leuk
(daunoXome, FAM and Ara-C) in the group of 62 patients Lymphoma 9:343–350
with high-risk acute leukemias [5]. The treatment-related 10. Estey E, Thall P, Andreeff M (1994) Use of G-CSF before,
during and after fludarabine plus Ara-C induction therapy of
mortality was 8%. Improving standards of supportive care newly diagnosed AML or MDS: comparison with fludarabine +
or decisions concerning intensive salvage treatment mak- Ara-C without G-CSF. J Clin Oncol 12:671–678
ing at earlier stage of the disease may explain these low 11. Estey EH (2000) Treatment of relapsed and refractory acute
mortality rates. The most prominent toxicity of the treat- myelogenous leukemia. Leukemia 14:476–479
ment was hematological toxicity. The median duration of 12. Ferra C, Berlanga J, Gallardo D, Ancin I, Marin D, Gonzalez JR,
Peris J, Munoz J, Sarra J, Granena A (2000) Mitoxantrone,
profound thrombocytopenia and neutropenia, numbers of etoposide, carboplatinum and ara-C combination therapy (MECA)
platelets transfused and days of hospitalization were longer in refractory and relapsed acute leukemia. Leuk Lymphoma
after CLAG-M regimen as compared with CLAG. How- 39:583–590
ever, they seem not to be different from hematological 13. Ferrara F, Melillo L, Montillo M, Leoni F, Pinto A, Mele G,
Mirto S (1999) Fludarabine, cytarabine and G-CSF (FLAG) for
toxicity observed by others after FLAG–Ida or FLAG–Mito the treatment of acute myeloid leukemia relapsing after auto-
regimens [7, 16]. logous stem cell transplantation. Ann Hematol 78:380–384
The nonhematological toxicity was low in most pa- 14. Gandhi V, Estey E, Keating MJ, Chucrallah A, Plunkett W
tients, predominantly reaching the WHO grades 1–2. Se- (1996) Chlorodeoxyadenosine and arabinosylcytosine in pa-
tients with acute myelogenous leukemia: pharmacokinetic,
vere nonhematological toxicity mainly consisted of loss of pharmacodynamic, and molecular interactions. Blood 87:256–
hair, nausea/vomiting, diarrhea and mucositis. 264
In summary, our results confirm the high antileukemic 15. Gandhi V, Estey E, Keating MJ, Plunkett W (1993) Fludarabine
efficacy of CLAG-M regimen and an acceptable toxicity potentiates metabolism of cytarabine in patients with acute
profile in patients with very poor prognosis refractory AML. myelogenous leukemia during therapy. J Clin Oncol 11:116–
124
16. Hanel M, Friedrichsen K, Hanel A, Herbst R, Morgner A,
Neser S, Nicklisch M, Teich M, Ehninger G, Fiedler F (2001)
References Mito-flag as salvage therapy for relapsed and refractory acute
myeloid leukemia. Onkologie 24:356–360
1. Archimbaud E, Thomas X, Leblond V, Michallet M, Fenaux P, 17. Hiddemann W, Martin WR, Sauerland CM, Heinecke A,
Cordonnier C, Dreyfus F, Troussard X, Jaubert J, Travade P et Buchner T (1990) Definition of refractoriness against conven-
al (1995) Timed sequential chemotherapy for previously treated tional chemotherapy in acute myeloid leukemia; a proposal
patients with acute myeloid leukemia: long-term follow-up of based on the results of retreatment by thioguanine, cytosine
the etoposide, mitoxantrone, and cytarabine-86 trial. J Clin arabinoside, and daunorubicin (TAD 9) in 150 patients with
Oncol 13:11–18 relapse after standardized first line therapy. Leukemia 4:184–
2. Carella AM, Cascavilla N, Greco MM, Melillo M, Sajeva MR, 188
Ladogana S, D’Arena G, Perla G, Carotenuto M (2001) Treat- 18. Hołowiecki J, Grosicki S, Robak T, Kyrcz-Krzemień S, Giebel
ment of “poor risk” acute myeloid leukemia with fludarabine, S, Hellamnn A, Skotnicki A, Jędrzejczak WW, Konopka L,
cytarabine and G-CSF (flag regimen): a single center study. Kuliczkowski K, Zdziarska B, Dmoszyńska A, Mariańska B,
Leuk Lymphoma 40:295–303 Pluta A, Zawilska K, Komarnicki M, Kłoczko J, Sułek K, Haus
3. Cheson BD, Cassileth PA, Head DR, Schiffer CA, Bennett JM, O, Stella-Hołowiecka B, Baran W, Jakubas B, Paluszewska M,
Bloomfield CD, Brunning R, Gale RP, Grever MR, Keating MJ Wierzbowska A, Kiełbiñski M, Jagoda K, Polish Adult Leu-
(1990) Report of the National Cancer Institute-sponsored work- kemia Group (PALG) (2004) Addition of cladribine to dau-
shop on definitions of diagnosis and response in acute myeloid norubicin and cytarabine increases complete remission rate
leukemia. J Clin Oncol 8:813–819 after a single course of induction treatment in acute myeloid
leukemia. Mulitcenter, phase III study. Leukemia 18:989–997
564
19. Huhmann IM, Watzke HH, Geissler K, Gisslinger H, Jager U, 29. Nokes TJ, Johnson S, Harvey D, Goldstone AH (1997) FLAG
Knobl P, Pabinger I, Korninger L, Mannhalter C, Mitterbauer is a useful regimen for poor prognosis adult myeloid leukemia
G, Schwarzinger I, Kahls P, Haas OA, Lechner K (1996) FLAG and myelodysplastic syndromes. Leuk Lymphoma 27:93–101
(fluadarabine, cytosine arabinoside, G-CSF) for refractory and 30. Parker JE, Pagliuca A, Mijovic A, Cullis JO, Czepulkowski B,
relapsed acute myeloid leukemia. Ann Hematol 73:265–271 Rassam SM, Samaratunga IR, Grace R, Gover PA, Mufti GJ
20. Jackson G, Taylor P, Smith GM, Marcus R, Smith A, Chu P, (1997) Fludarabine, cytarabine, G-CSF and idarubicin (FLAG-
Littlewood TJ, Duncombe A, Hutchinson M, Mehta AB, IDA) in the treatment of poor risk myelodysplastic syndromes
Johnson SA, Carey P, Mackie MJ, Ganly PS, Turner GE, Deane and acute myeloid leukaemia. Br J Haematol 99:939–944
M, Schey S, Brookes J, Tollerfield SM, Wilson MP (2001) A 31. Prebet T, Ducastelle S, Debotton S, Stamatoullas A, Deconinck
multicentre, open, non-comparative phase II study of a com- E, Fruchart C, Gratecos N, Ifrah N, Dreyfus F, Fenaux P, Wattle
bination of fludarabine phosphate, cytarabine and granulocyte E (2004) A phase II study of intensive chemotherapy with
colony-stimulating factor in relapsed and refractory acute mye- fludarabine, cytarabine, and mitoxantrone in P-glycoprotein-
loid leukemia and de novo refractory anaemia with excess of negative high-risk myelodysplastic syndromes. Hematol J
blasts in transformation. Br J Haematol 112:127–137 5:209–215
21. Juliusson G, Hoglund M, Karlsson K, Lofgren C, Mollgard L, 32. Robak T, Wrzesień-Kuś A, Lech-Marańda E, Kowal M,
Paul C, Tidefelt U, Bjorkholm M, Leukemia Group of Middle Dmoszyńska A (2000) Combination regimen of cladribine
Sweden (2003) Increased remissions from one course for in- (2-chlorodeoxyadenosine), cytarabine and G-CSF (CLAG) as
termediate-dose cytosine arabinoside and idarubicin in elderly induction therapy for patients with relapsed or refractory
acute myeloid leukaemia when combined with cladribine. A acute myeloid leukemia. Leuk Lymphoma 39:121–129
randomized population-based phase II study. Br J Haematol 33. Robak T, Wrzesień-Kuś A (2002) The search for optimal
123:810–818 treatment in relapsed and refractory acute myeloid leukemia.
22. Kern W, Schleyer E, Braess J, Wittmer E, Ohnesorge J, Leuk Lymphoma 43:281–291
Unterhalt M, Wormann B, Buchner T, Hiddemann W (2001) 34. Robak T (2003) Purine nucleoside analogues in the treatment of
Efficacy of fludarabine, intermittent sequential high-dose cyto- myeloid leukemia. Leuk Lymphoma 44:391–409
sine arabinoside and mitoxantrone (FIS-HAM) salvage therapy 35. Slovak ML, Kopecky KJ, Cassileth PA, Harrington DH, Theil
in highly resistant acute leukemias. Ann Hematol 80:334–339 KS, Mohamed A, Paietta E, Willman CL, Head DR, Rowe JM,
23. Kern W, Schoch C, Haferlach T, Braess J, Unterhalt M, Forman SJ, Appelbaum FR (2000) Karyotypic analysis predicts
Wormann B, Buchner T, Hiddemann W, German AML Co- outcome of pre-remission and postremission therapy in adult
operative Group (2000) Multivariate analysis of prognostic acute myeloid leukemia: a Southwest Oncology Group/Eastern
factors in patients with refractory and relapsed acute myeloid Cooperative Oncology Group study. Blood 96:4075–4083
leukemia undergoing sequential high-dose cytosine arabinoside 36. Steinmetz HT, Schulz A, Staib P, Scheid C, Glasmacher A,
and mitoxantrone (S-HAM) salvage therapy: relevance of cyto- Neufang A, Franklin J, Tesch H, Diehl V, Dias Wickramanayake P
genetic abnormalities. Leukemia 14:226–231 (1999) Phase-II trial of idarubicin, fludarabine, cytosine ara-
24. Kornblau SM, Gandhi V, Andreeff HM, Beran M, Kantarjian binoside, and filgrastim (Ida-FLAG) for treatment of refractory,
HM, Koller CA, O’Brien S, Plunkett W, Estey E (1996) Clin- relapsed, and secondary AML. Ann Hematol 78:418–425
ical and laboratory studies of 2-chlorodeoxyadenosine +/− 37. Tallman MS, Lee S, Sikic BI, Paietta E, Wiernik PH, Bennett
cytosine arabinoside for relapsed or refractory acute myelog- JM, Rowe JM (1999) Mitoxantrone, etoposide and cytarabine
enous leukemia in adults. Leukemia 10:1563–1569 plus cyclosporine for patients with relapsed or refractory acute
25. Kristensen J, Nygren P, Liliemark J, Fridborg H, Killander A, myeloid leukemia. Cancer 85:358–367
Simonsson B, Oberg G, Larsson R (1994) Interactions between 38. Visani G, Tosi P, Zinzani PL, Manfroi S, Ottaviani E, Testoni
cladribine (2-chlorodeoxyadenosine) and standard anti-leuke- N, Clavio M, Cenacchi A, Gamberi B, Carrara P, Gobbi M,
mic drugs in primary cultures of human tumour cells from Tura S (1994) FLAG (fludarabine+high dose cytarabine+G-
patients with acute myelocytic leukemia. Leukemia 8:1712– CSF): an effective and tolerable protocol for the treatment of
1717 “poor risk” acute myeloid leukemia. Leukemia 8:1842–1846
26. Kufe D, Spriggs D, Egan EM, Munroe D (1984) Relationships 39. Wrzesień-Kuś A, Robak T, Lech-Marańda E, Wierzbowska A,
among Ara-CTP pools, formation of (Ara-C) DNA and cy- Dmoszyńska A, Kowal M, Holowiecki J, Kyrcz-Krzemien S,
totoxicity of human leukemic cells. Blood 64:54–58 Grosicki S, Maj S, Hellmann A, Skotnicki A, Jędrzejczak W,
27. Liu Yin JA, Wheatley K, Rees JK, Burnett AK, UK MRC Kuliczkowski K (2003) A multicenter, open, non-comparative
Adult Leukemia Working Party (2001) Comparison of ‘se- phase II study of the combination of cladribine (2-chlorodeoxy-
quential’ versus ‘standard’ chemotherapy as re-induction treat- adenosine), cytarabine, and G-CSF as induction therapy in
ment, with or without cyclosporine, in refractory/relapsed acute refractory acute myeloid leukemia—a report of the Polish Adult
myeloid leukemia (AML): results of the UK medical research Leukemia Group (PALG). Eur J Haematol 71:155–162
council AML-R trial. Br J Haematol 113:713–726
28. Montillo M, Mirto S, Petti MC, Latagliata R, Magrin S, Pinto
A, Zagonel V, Mele G, Tedeschi A, Ferrara F (1998) Flu-
darabine, cytarabine and G-CSF (FLAG) for the treatment of
poor risk acute myeloid leukemia. Am J Hematol 58:105–109

Vous aimerez peut-être aussi