You are on page 1of 22

Clinical Therapeutics/Volume 31, Theme Issue, 2009

Current and Emerging Therapies for Acute Myeloid Leukemia


Tadeusz Robak, MD, PhD; and Agnieszka Wierzbowska, MD, PhD
Department of Hematology, Medical University of Lodz, Copernicus Memorial Hospital, Lodz, Poland

ABSTRACT expected to relapse within 3 years. The optimum


Background: Acute myeloid leukemia (AML) is a strategy at the time of relapse, or for patients with the
clonal disease characterized by the proliferation and resistant disease, remains uncertain. Allogeneic stem
accumulation of myeloid progenitor cells in the bone cell transplantation has been established as the most
marrow, which ultimately leads to hematopoietic fail- effective form of antileukemic therapy in patients with
ure. The incidence of AML increases with age, and AML in first or subsequent remission. New drugs are
older patients typically have worse treatment outcomes being evaluated in clinical studies, including immuno-
than do younger patients. toxins, monoclonal antibodies, nucleoside analogues,
Objective: This review is focused on current and hypomethylating agents, farnesyltransferase inhibi-
emerging treatment strategies for nonpromyelocytic tors, alkylating agents, FMS-like tyrosine kinase 3 in-
AML in patients aged <60 years. hibitors, and multidrug-resistant modulators. How-
Methods: A literature review was conducted of the ever, determining the success of these treatment strategies
PubMed database for articles published in English. ultimately requires well-designed clinical trials, based on
Publications from 1990 through March 2009 were stratification of the patient risk, knowledge of the indi-
scrutinized, and the search was updated on August 26, vidual disease, and the drug’s performance status.
2009. The search terms used were: acute myeloid leu- Conclusions: Combinations of AraC and anthracy-
kemia in conjunction with treatment, chemotherapy, clines are still the mainstay of induction therapy, and
stem cell transplantation, and immunotherapy. Clinical use of high-dose AraC is now a standard consolida-
trials including adults with AML aged ≥19 years were tion therapy in AML patients aged <60 years. Although
selected for analysis. Conference proceedings from several new agents have shown promise in treating
the previous 5 years of The American Society of Hema- AML, it is unlikely that these agents will be curative
tology, The European Hematology Association, and when administered as monotherapy; it is more likely
The American Society for Blood and Marrow Trans- that they will be used in combination with other new
plantation were searched manually. Additional relevant agents or with conventional therapy. (Clin Ther. 2009;
publications were obtained by reviewing the references 31[Theme Issue]:2349–2370) © 2009 Excerpta
from the chosen articles. Medica Inc.
Results: Cytarabine (AraC) is the cornerstone of Key words: acute myeloid leukemia, nonpromyelo-
induction therapy and consolidation therapy for AML. cytic, cytarabine, induction therapy.
A standard form of induction therapy consists of AraC
(100–200 mg/m2), administered by a continuous infu-
sion for 7 days, combined with an anthracycline, INTRODUCTION
administered intravenously for 3 days. Consolidation Acute myeloid leukemia (AML) is a clonal disease
therapy comprises treatment with additional courses characterized by the proliferation and accumulation
of intensive chemotherapy after the patient has achieved of myeloid progenitor cells in bone marrow, which
a complete remission (CR), usually with higher doses ultimately leads to hematopoietic failure. Diagnosis of
of the same drugs as were used during the induction AML is based on cellular morphology and immuno-
period. High-dose AraC (2–3 g/m2) is now a standard logic, cytogenetic, and molecular features.1 The inci-
consolidation therapy for patients aged <60 years.
Despite substantial progress in the treatment of newly
Accepted for publication September 24, 2009.
diagnosed AML, 20% to 40% of patients do not doi:10.1016/j.clinthera.2009.11.017
achieve remission with the standard induction chemo- 0149-2918/$ - see front matter
therapy, and 50% to 70% of first CR patients are © 2009 Excerpta Medica Inc. All rights reserved.

2009 2349
Clinical Therapeutics

dence of AML increases with age, with a median diag- been cited only if the primary source could not be
nosis between 65 and 75 years of age.2 Although found or if valuable information about several clinical
AML is the most common type of leukemia in adults, trials was listed. Although the focus of this review was
it continues to have the lowest survival rate of all the on younger patients, studies involving older patients
leukemias.3 AML accounts for ~25% of all leukemias are also presented, as usual practice in AML is that
in adults in the Western world but comprises only new drugs are investigated in older patients first, as
15% to 20% of cases in patients aged ≤15 years.4 In this group usually has a particularly poor prognosis.
the United States, 44,270 new cases of leukemia were
reported in 2008, of which 13,290 were AML cases.5 INDUCTION THERAPY
In 2007, nearly 9000 patients died of this disease. A The treatment of patients with AML includes at least
number of prognostic factors have been identified in one course of intensive myelosuppressive induction
AML, including age, performance status, organ dys- chemotherapy.7,10 A widely accepted form of induction
function, white blood cell count at presentation, therapy includes standard-dose cytarabine (SDAraC)
karyotype, and molecular abnormalities.6,7 100 to 200 mg/m2, administered by a continuous infu-
AML is a heterogeneous disease in which a variety sion for 7 days, combined with daunorubicin 45 to
of cytogenetic and molecular alterations have been 60 mg/m2/d, administered intravenously for 3 days
identified.1 Specific chromosomal abnormalities have (the 3+7 induction regimen).8,11 This therapy has been
been established as particularly strong prognostic reported to induce a complete remission (CR) in 65%
markers for survival.1,7 Cytogenetic abnormalities to 75% of patients aged 18 to 60 years.1,11–13 This
can be detected in ~50% to 60% of newly diagnosed approach results in a long-term disease-free survival
AML patients. of ~30%, with a treatment-related mortality (ie, the
AML treatment includes at least one course of inten- percentage of patients who died during induction) of
sive induction chemotherapy, followed by an additional 5% to 10%.1,11–13
course of intensive consolidation therapy and then main- Efforts to improve the CR rate have included use of
tenance therapy. Moreover, autologous and allogeneic alternative anthracyclines or anthracenodiones, incor-
stem cell transplantation (autoSCT and alloSCT, respec- poration of high-dose AraC (HDAraC), or addition of
tively) procedures can be performed in selected pa- other agents such as etoposide, fludarabine, or cladri-
tients. In addition, many new agents for the treatment of bine.13–21 Some trials have reported that substitution of
AML have been developed recently and are currently idarubicin for daunorubicin in a 3+7 induction regimen
entering clinical trials.8,9 This review is focused on cur- resulted in a higher CR rate (80% vs 58% [P = 0.005],
rent and emerging treatment strategies for nonpromyelo- 71% vs 58% [P = 0.032], and 70% vs 59% [P = 0.08],
cytic AML in patients aged <60 years. respectively).13–15 Additionally, one trial found a signifi-
cant difference in overall survival (OS), which favored
METHODS the idarubicin arm (P = 0.025).13 In a meta-analysis of
A literature review was conducted of the PubMed 5 trials comparing idarubicin and daunorubicin,
database for articles published in English. Publica- members of the AML Collaborative Group reported
tions from 1990 through March 2009 were scruti- that idarubicin performed more strongly than dauno-
nized, and the search was updated on August 26, rubicin in 3+7 induction therapy in terms of longer
2009. The search terms used were: acute myeloid leu- disease-free survival (P = 0.07), 15% relapse rate re-
kemia in conjunction with treatment, chemotherapy, duction (P = 0.008), and 14% risk reduction for death
stem cell transplantation, and immunotherapy. Clini- (P = 0.03).21 However, these trials used nonequivalent
cal trials including adults with AML aged ≥19 years doses of daunorubicin and idarubicin, in favor of the
were selected for analysis. Conference proceedings latter.
from the previous 5 years of The American Society of The addition of etoposide to daunorubicin and
Hematology, The European Hematology Association, AraC did not improve the remission rate but increased
and The American Society for Blood and Marrow Trans- remission duration in a study of 264 previously un-
plantation were searched manually. Additional rele- treated AML patients aged 15 to 70 years.17 Patients
vant publications were obtained by reviewing the ref- were randomized to receive either AraC 100 mg/m2/d
erences from the chosen articles. Review articles have continuous IV infusion on days 1 through 7 and

2350 Volume 31 Theme Issue


T. Robak and A. Wierzbowska

daunorubicin 50 mg/m2/d IV on days 1 through 3 (AD induction regimen improved the CR rate and OS in
regimen), or the same drugs intensified with etoposide adults with AML aged ≤60 years, with no additional
75 mg/m2/d IV on days 1 through 7 (ADE regimen) as toxicity.19 However, this beneficial effect was not ob-
induction therapy. CR rates did not differ significantly served in patients taking daunorubicin combined with
in either arm (56% vs 59%, respectively). However, fludarabine.
ADE significantly improved remission duration in The effectiveness of an induction regimen comprising
patients aged <55 years (P = 0.01). Moreover, induc- HDAraC (2–3 g/m2) and SDAraC (100–200 mg/m2) has
tion and consolidation therapies with etoposide in- been compared in several randomized trials.23–27 In a
creased OS in this group of patients, with a median of study by the Southwest Oncology Group,24 the CR
9 months for the AD regimen and 17 months for the rate was 55% for patients aged 15 to 49 years re-
ADE regimen (P = 0.03). ceiving HDAraC and 58% for those receiving SDAraC
A randomized, multicenter trial by the Lederle Co- (Table I). In patients aged 50 to 64 years, the CR rate
operative Group involved 200 previously untreated was 45% and 53% in the HDAraC and SDAraC
AML patients (median age, 60 years).16 Mitoxantrone groups, respectively. With a median follow-up time of
in combination with AraC was more effective than the 51 months, survival was not significantly different.
standard 3+7 induction regimen in terms of CR rates The estimated survival rate at 4 years was 32% for
after a single induction course (89% vs 53%, respec- HDAraC and 22% for SDAraC in patients aged 15 to
tively), the median time to CR and CR duration, and 49 years and 13% and 11%, respectively, in patients
the median length of survival. aged 50 to 64 years.
The addition of purine nucleoside analogues (PNA) An analysis of 3 large randomized trials has con-
to anthracycline and AraC induction therapy may firmed that there is no evidence that HDAraC leads to
benefit some patients with AML.18–20,22 A Phase III an increase in CR rate.26 However, it has been re-
trial by Russo et al18 evaluated the efficacy and toxici- ported that HDAraC, used as a part of induction
ty of the FLAI regimen (fludarabine + AraC + idarubi- therapy, has led to a longer remission duration and a
cin) compared with the ICE regimen (idarubicin + better 4-year OS in patients aged <60 years.
AraC + etoposide) in newly diagnosed AML patients Double induction represents another method of
aged <60 years. After a single induction course, the intensifying induction treatment in AML using the sys-
CR rate was 74% in the FLAI arm and 51% in the tematic administration of a second induction course,
ICE arm (P = 0.01); death during the induction period irrespective of the bone marrow status after the first
was 2% and 9%, respectively. Moreover, hematologic course. A randomized study by the German AML
and nonhematologic toxicities were significantly low- Cooperative Group found that SDAraC (100 mg/m2
er in the FLAI group (P = 0.002 and P < 0.001, respec- on days 1–8) with daunorubicin (60 mg/m2 on
tively). The role of cladribine in addition to 3+7 in- days 3–5) and 6-thioguanine (100 mg/m2 every 12 hours
duction therapy in AML was also investigated in a on days 3–9) (TAD regimen) followed by HDAraC (3 g/m2
large multicenter, randomized study.18,19 A total of every 12 hours on days 1–3) with mitoxantrone
400 untreated AML patients (mean age, 45 years; age (10 mg/m2 on days 3–5) (HAM regimen) was associ-
range, 16–60 years) were randomized to receive either ated with a nonsignificant trend towards a higher CR
DAC-7 (daunorubicin + AraC + cladribine) or DA-7 incidence compared with 2 courses of TAD (71% vs
(without cladribine). The overall response (OR) and 65%, respectively; P = 0.072).25 However, in another
CR rates were similar in the DAC-7 and DA-7 arms. German study, a more intensive induction regimen us-
However, after a single course of DAC-7 induction, ing 2 courses of HAM did not result in any improve-
the CR rate was 64% and was significantly higher ment in outcome (eg, response, OS, relapse-free survival,
than in the DA-7 arm (47%) (P < 0.001). Moreover, early death rates).27
there was a nonsignificant trend towards higher A randomized study by the Acute Leukemia French
3-year leukemia-free survival for patients aged >40 Association compared the 3+7 induction regimen
years receiving DAC-7 compared with DA-7 (44% vs (daunorubicin 80 mg/m2 on days 1–3 + SDAraC) with
28%; P = 0.05). The subsequent randomized, 3-arm double induction (3+7 induction regimen followed by
study performed by the Polish Adult Leukemia Group mitoxantrone with intermediate-dose AraC [IDAraC])
proved that adding cladribine to the standard DA-7 and time-sequential therapy (3+7 induction regimen

2009 2351
Clinical Therapeutics

Table I. The results of selected randomized trials comparing high-dose cytarabine (AraC) versus standard-
dose AraC induction regimens in acute myeloid leukemia.

AraC Dose, Age, No. of CR, DFS, 4-Year OS,


Study mg/m2/d y Patients % mo %
ALSG23
High dose 6000 × 4 days 15–60 149 71 22* 34
Standard dose 100 × 7 days 15–60 152 74 12* 25
SWOG24
High dose 4000 × 6 days 15–49 85 55 12† 32†
Standard dose 200 × 7 days 15–49 293 58 10 22
High dose 4000 × 6 days 50–64 87 45 8 13
Standard dose 200 × 7 days 50–64 200 53 8 11
AMLCG25
High dose 4000 × 6 days 16–60 365 71 23‡ 36§
Standard dose 200 × 7 days 16–60 360 65 18 32

CR = complete remission; DFS = disease-free survival; OS = overall survival; ALSG = Australian Leukemia Study Group;
SWOG = Southwest Oncology Group; AMLCG = German AML Cooperative Group.
*P = 0.007.
†P = 0.021.
‡P = 0.012.
§P = 0.009.

followed by an early course of mitoxantrone + Leukemia Group B trial,33 1088 patients were treated
IDAraC) in newly diagnosed patients with AML aged with the standard 3+7 induction regimen of daunoru-
<65 years.28 No difference in CR rate, early death bicin and AraC followed by postremission therapy
incidence, or relapse-free survival was observed be- with 1 of 3 doses of AraC: 100 mg/m2/d for 3 days,
tween the 3 randomization arms. 400 mg/m2 for 5 days, or 3000 mg/m2 every 12 hours
Presently, there is no conclusive evidence to recom- on days 1, 3, and 5. For patients aged <60 years, the
mend one 3+7 induction regimen over another. How- 4-year disease-free survival was 44% in the HDAraC
ever, the results of these studies clearly support the group, compared with 29% and 24% in the intermediate-
claim that further intensification of the induction regi- dose and low-dose groups, respectively (P = 0.002).
men is not associated with an increased CR rate. Serious neurotoxicity was reported only in the HDAraC
group (12%). Subsequent studies found that 3 or 4 con-
CONSOLIDATION THERAPY secutive consolidation courses with HDAraC (cu-
Consolidation therapy comprises treatment with addi- mulative dose, 54–72 g/m2) were more effective than one
tional courses of intensive chemotherapy after the patient course (18 g/m2) among patients with core-binding
has achieved CR, usually with higher doses of the same factor leukemia (t[8;21] or inv[16]) in terms of both
drugs as were used during the induction period.10,29 The disease-free survival (P = 0.03) and OS (P = 0.04).34,35
goal of consolidation therapy is to prevent relapse and However, data from a meta-analysis indicate that
achieve a cure in patients who are in CR after induction results found with 2 cycles in combination with an
treatment. The median disease-free survival for patients anthracycline or mitoxantrone were not different
who received only the induction therapy is 4 to 8 months.30 from those using 3 or 4 cycles.36 In a randomized
However, 35% to 50% of adults aged <60 years who study by the Finnish Leukemia Group,37 there was no
receive consolidation treatment survive 2 to 3 years.30,31 significant difference in survival between patients who
HDAraC is now a standard consolidation therapy had been randomized to receive 2 courses or 6 courses
in patients aged <60 years.32–35 In the Cancer and of the consolidation therapy.

2352 Volume 31 Theme Issue


T. Robak and A. Wierzbowska

The current data on consolidation chemotherapy do not achieve remission with standard induction
for AML patients in first CR suggest that multiple chemotherapy, and 50% to 70% of first CR patients
courses of HDAraC remain the mainstay of a curative are expected to relapse over 3 years.42,43 The progno-
therapy for core-binding factor leukemia. The aug- sis for patients with AML refractory to first-line treat-
mentation of the cytogenetic risk stratification by ment or in first or subsequent relapse is generally
molecular prognostic markers may help define addi- poor. The duration of first remission in relapsed pa-
tional subgroups of AML patients who will benefit tients is the most important prognostic factor correlat-
from the intensified chemotherapy. ing with the probability of second CR and survival.44
Patients with primary refractory and early relapsed
MAINTENANCE THERAPY disease with a CR duration <6 months have a signifi-
Maintenance therapy, which is considered less myelo- cantly poorer response to therapy and OS than do pa-
suppressive than the induction and consolidation forms tients who relapsed after a first CR lasting ≥6 months
of treatment, is used in patients who have previously (CR rate 10%–30% vs 40%–60%, respectively).45–48
obtained CR.10 It is a strategy to further reduce the The optimum strategy at the time of relapse or for
number of residual leukemic cells and prevent a patients with resistant disease remains uncertain. Use
relapse.38–41 However, its role in the routine manage- of alloSCT may be curative for a minority of patients
ment of AML patients is controversial and depends who achieve a second CR and who have an available
mainly on the intensity of the induction and consoli- donor.42,49 For the majority of patients, additional che-
dation therapies.39,42 A randomized study by Sauter et motherapy is given in the hope of achieving remission.
al38 found that the use of maintenance chemotherapy Most salvage therapies use high (2–3 g/m2) or inter-
every 8 weeks for 2 years after 1 course of consolida- mediate (1–1.5 g/m2) doses of AraC in combination
tion therapy had no advantage in terms of OS compared with other agents to overcome resistance in leukemia
with observation only. However, the German AML cells.43,50,51
Cooperative Group reported that monthly mainte- The results of randomized trials in patients with
nance therapy based on AraC (100 mg/m2 SC every relapsed or refractory AML are summarized in Ta-
12 hours on days 1–5), with daunorubicin (45 mg/m2 ble II.45,52–62 There is no single regimen or approach
IV on days 3 and 4), thioguanine (100 mg/m2 orally that is considered the standard of care in relapsed and
on days 1–5), or cyclophosphamide (1 g/m2 IV on day 3) refractory AML. The factors most predictive of re-
given alternatively as a second agent, was associated sponse in relapsed patients are the duration of previ-
with 31.4% of patients being relapse free at 5 years ver- ous remission and the number of previous salvage
sus 24.7% patients in the sequential HAM arm being courses.63 Salvage therapy is associated with CR rates
relapse free (P < 0.02). Relapse-free survival after of 40% to 60% in patients with a first CR lasting
maintenance therapy was better in patients with poor ≥1 year. However, in patients with the first CR lasting
risk by unfavorable karyotype, age ≥60 years, lactate <1 year, the rate of second CR is only 10% to 15%.10,63
dehydrogenase level >700 U/L, or day 16 bone mar- In the past 10 years, several prospective Phase II clini-
row blasts >40% (P = 0.006).40 It should be empha- cal trials have been conducted to evaluate new combi-
sized that in these German studies, patients without nations of standard chemotherapies or novel agents in
maintenance therapy received only 1 course of patients with relapsed or refractory disease.7,64
HDAraC-containing consolidation therapy, which may Table III summarizes studies using combinations of
explain these results. According to the current guidelines PNAs with AraC in the treatment of adult patients
of the British Committee for Standards in Haematology, with relapsed or refractory AML.48,65–74 Several inves-
there is no evidence that maintenance therapy benefits tigations have revealed that PNAs, especially cladri-
AML patients who have undergone intensive consolida- bine and fludarabine, are active agents in relapsed or
tion therapy.31 refractory AML.64 PNAs in combination with AraC
increase the accumulation of AraC triphosphate,
TREATMENT OF RELAPSED AND which is responsible for the cytotoxic effect in leuke-
REFRACTORY DISEASE mic blasts.64 The combination of fludarabine or cla-
Despite the substantial progress in the treatment of dribine with AraC has been extensively explored in
newly diagnosed AML, 20% to 40% of patients still relapsed and refractory AML.51,65–74 The combination

2009 2353
2354

Clinical Therapeutics
Table II. The results of randomized trials in patients with relapsed or refractory acute myeloid leukemia.

No. of Age, Second CR Median Second Early Median


Study Treatment Patients y Rate, % CR Duration, mo Death, % OS, mo

Kern et al45 HDAraC + Mit vs IDAC + Mit 186 50 vs 50 52 vs 45 5.3 vs 3.3 32 vs 17 5 vs NG


Martiat et al52 HDAraC + Amsa vs HDAraC + Mit 52 NG 53 vs 60 11 vs 12 15 vs 8 8 vs 11
Larson et al53 HDAraC vs HDAraC + Amsa 36 52 14 vs 53* NG 25 vs 25 2 vs 6
Vogler et al54 HDAraC vs HDAraC + Eto 131 119 patients 40 vs 45 12 vs 25 NG 5 vs 5
aged <70 years
Ohno et al55 MAE vs MAE + G-CSF 58 47 vs 43 42 vs 54 14 vs 12 8 vs 0 NG
Karanes et al56 HDAraC vs HDAraC + Mit 162 14–76 32 vs 44 9 vs 5 10 vs 16 8 vs 6
Thomas et al57 EMA vs EMA + GM-CSF 72 46 vs 47 81 vs 89 4 vs 5 8 vs 5 8.5 vs 10
Liu Yin et al58 ADE with or without CSA vs 235 48 (4–75) 57 vs 38 NG 16 vs 24 NG
seq ADE with or without CSA (<60 y)*
48 vs 25
(≥60 y)
List et al59 HDAraC + daunorubicin vs HDAraC +
daunorubicin + CSA 226 NG 33 vs 39 NG 15 vs 18 NG
Greenberg et al60 MEC vs MEC + PSC-833 129 70% of patients 25 vs 17 9.3 vs 10 10 vs 16 5.4 vs 4.6
aged ≥50 years
Feldman et al61 MEC vs MEC + lintuzumab 191 55.4 vs 48.8 23 vs 29 NG NG 8.1 vs 5.7
Giles et al62 HDAraC vs HDAraC + laromustine 178 59 (22–84) 19 vs 35† 332 vs 275 2 vs 11‡ 177 vs 128
Volume 31 Theme Issue

CR = complete remission; OS = overall survival; HDAraC = high-dose cytarabine; Mit = mitoxantrone; IDAC = intermediate-dose AraC; NG = not given; Amsa =
amsacrine; Eto = etoposide; MAE = Mit + AraC + Eto; G-CSF = granulocyte-colony stimulating factor; EMA = Eto + Mito + AraC; GM-CSF = granulocyte,
macrophage–colony stimulating factor; ADE = AraC + daunorubicin + Eto; CSA = cyclosporine; seq ADE = sequential ADE; MEC = Mit + Eto + AraC.
* P < 0.01.
† P = 0.005.
‡ P = 0.016.
T. Robak and A. Wierzbowska

Table III. Combinations of purine nucleoside analogues with cytarabine (AraC) in the treatment of adult pa-
tients with relapsed or refractory acute myeloid leukemia.

Age, y
No. of Salvage Overall OS and Early
Study Patients Regimen Median Range CR Rate, % Time Death, %
Wierzbowska et al48 118 CLAG-M 45 20–66 58 14% at 4 years 8
Steinmetz et al65 36 FLAG-IDA 49 19–75 52 0% at 1 year 14
52 30–75 15% at 1 year
Jackson et al66 83 FLAG 48 18–69 – 9% at 2 years 18
47 21–74 81 50% at 2 years
de la Rubia et al67 32 FLAG-IDA 59 18–79 53 40% at 1 year 9
Clavio et al68 59 FLAG/FLANG 64 33–76 59 NA 10
Carella et al69 41 FLAG 52 16–72 56 20% at 2 years 7
Wrzesień-Kuśet al70 58 CLAG 45 18–67 50 42% at 1 year 17
Pastore et al71 46 FLAG-IDA 41 15–60 52 NA 6.6
Hänel et al72 29 Mit-FLAG 59 18–75 59 34% at 1 year 14
Huhmann et al73 22 FLAG 46 24–63 50 58 at 1 year, 5
24 at 2 years
Camera et al74 61 FLAD 43 26–81 52 5.8 months 12
(median)

CR = complete remission; OS = overall survival; CLAG-M = CLAG + mitoxantrone (Mit); FLAG = fludarabine + AraC +
granulocyte-colony stimulating factor (G-CSF); IDA = idarubicin; FLANG = FLAG + Mit; NA = not available; CLAG =
cladribine + AraC + G-CSF; FLAD = fludambine + AraC + daunorubicin.

of fludarabine with AraC, with or without granulo- NOVEL AND EMERGING DRUGS FOR
cyte-colony stimulating factor (G-CSF) and/or an ACUTE MYELOID LEUKEMIA
anthracycline, induced a second CR in 30% to 80% The current data on postremission therapy in AML
of patients with relapsed or refractory disease. Simi- suggest that further dose intensification of presently
lar results were obtained when cladribine was used available cytotoxic drugs is unlikely to improve the
instead of fludarabine.48,70 A multicenter study by clinical outcome.25,28,37 Optimizing the AML therapy
The Medical Research Council compared ADE (AraC will incorporate new cytotoxic treatments and new
+ daunorubicin + etoposide) with FLA (fludarabine targeted therapies into the standard approach. Target-
+ HDAraC), with patients further randomized to re- ing essential cell functions creates drugs with narrow
ceive all-trans retinoic acid and G-CSF.51 In this therapeutic indices. These new agents are unlikely to
study, 250 patients with relapsed, resistant/refractory, be curative when administered as monotherapy. Thus,
or adverse genetic AML were randomly assigned to new compounds with an antileukemic activity, which
receive either ADE or FLA. The CR rate was 61%, have previously been evaluated in small Phase I and II
with a 4-year disease-free survival of 29%. There studies, should be tested in combination with other new
were no significant differences in the CR rate, deaths agents or with already approved conventional drugs.
during CR, relapse rate, or disease-free survival be- Emerging agents potentially useful in the treatment of
tween the ADE and FLA groups. However, survival patients with AML include immunotoxins, monoclonal
at 4 years was better in the ADE arm (27%) than in antibodies, nucleoside analogues, hypomethylating
the FLA arm (16%) (P = 0.05). agents, farnesyltransferase inhibitors, alkylating agents,

2009 2355
Clinical Therapeutics

FMS-like tyrosine kinase 3 (FLT3) inhibitors, and on the surface of AML cells in 80% to 90% of pa-
multidrug-resistant modulators (Table IV).75–80 tients with AML. After binding with CD33, the
toxin is internalized, causing double-strand DNA
Immunotoxins and Monoclonal Antibodies breaks, which lead to cell death.77 In a Phase I dose-
The only antibody approved by the US Food and escalation study, the drug was evaluated in 40 patients
Drug Administration (FDA) for AML therapy is gem- with relapsed or refractory AML.79 Infusion-related
tuzumab ozogamicin* (GO).77 GO consists of a hu- toxicity, including fever and chills, occurred in 32
manized anti–CD33 antibody (hP 67.6) linked to N- of 40 patients (80%) within 2 to 4 hours from the
acetyl-calicheamicin 1,2-dimethyl hydrazine chloride, start of treatment. The maximum tolerated dose (MTD)
a potent cytotoxic antibiotic. CD33 antigen is present was established as 9 mg/m2 and dosing intervals as
14 days. Three patients achieved CR and 6 patients
*Trademark: Mylotarg ® (Wyeth Pharmaceuticals, Madison, had clearance of bone marrow blast cells with incom-
New Jersey]). plete platelet recovery. In the Phase II studies, GO as

Table IV. Novel agents potentially useful for acute myeloid leukemia.75–80

Drug Class Agents Mechanism of Action

Immunotoxins Gemtuzumab ozogamicin (Mylotarg® [Wyeth CD33 binding, toxin


Pharmaceuticals, Madison, New Jersey]) internalization, and double-
strand DNA breaks
Monoclonal Lintuzumab (HuM195 [Seattle Genetics Inc., Bothell, CD33 binding, complement-
antibodies Washington]) dependent cytotoxicity, and
antibody-directed cellular
cytotoxicity
Newer nucleoside Clofarabine (Evoltra® [Genzyme Corporation, Cambridge, Inhibition of ribonucleotide
analogues Massachusetts]); troxacitabine (Troxatyl® [SGX reductase and DNA
Pharmaceuticals, San Diego, California]); sapacitabine polymerase; induction of
(CYC682, CS-682 [Cyclacel Ltd., Dundee, United Kingdom]) apoptosis
Hypomethylating Azacitidine (Vidaza® [Celgene Corporation, Summit, Inhibition of DNA
agents New Jersey]); Decitabine (Dacogen® [Eisai Inc., Woodcliff methylation
Lake, New Jersey])
Farnesyltransferase Tipifarnib (Zarnestra™ [Johnson & Johnson Pharmaceutical Inhibition of farnesyl-
inhibitors Research & Development, L.L.C., Raritan, New Jersey]); transferase, inhibition of
lonafarnib (Sarasar® [Schering-Plough Corporation, angiogenesis, and induction
Kenilworth, New Jersey]) of cellular adhesion
Alkylating agents Laromustine (Onrigin ® [Vion Pharmaceuticals, Inc., New DNA alkylation and DNA
Haven, Connecticut]) cross-linking
Tyrosine kinase Lestaurtinib (CEP701 [Cephalon, Inc., Frazer, Pennsylvania]); Inhibition of FLT 3
inhibitors (FLT 3 tandutinib (MLN518 [Millennium Pharmaceuticals, phosphorylation,
inhibitors) Cambridge, Massachusetts]); PKC412 (Novartis induction of apoptosis
Oncology, East Hanover, New Jersey)
Multidrug-resistant Valspodar (PSC-833 [Novartis Pharmaceuticals, East Binding to P-glycoprotein
modulators Hanover, New Jersey]); zosuquidar (LY335979 [Kanisa and inactivation of its
Pharmaceuticals, Inc./Lilly, San Diego, California]) efflux function
FLT 3 = FMS-like tyrosine kinase 3.

2356 Volume 31 Theme Issue


T. Robak and A. Wierzbowska

monotherapy led to a 25% to 30% OR rate in unselected tigated at a dosage of 12 or 36 mg/m2 on days 1 to 4
patients newly diagnosed with AML.75 The drug is highly and days 15 to 18 in patients with relapsed or refrac-
myelosuppressive and causes prolonged thrombocyto- tory AML.88 Two CRs and one partial remission
penia. Infections, veno-occlusive disease, and abnormal were achieved among 49 assessable patients. In
results on liver function tests have been reported.80 9 other patients, a decrease in blast counts was ob-
The value of the combination of GO and standard served. More recently, lintuzumab in combination
chemotherapy in AML is currently being evaluated in with mitoxantrone, etoposide, and IDAraC was
randomized trials.81 Preliminary results of the Medi- evaluated in a Phase III, randomized, multicenter
cal Research Council AML 15 Trial indicate that ad- trial.61 A total of 191 patients with relapsed and
dition of GO improved outcomes in patients with in- primary resistant AML were randomly assigned to
termediate and favorable cytogenetics.82 A combination receive mitoxantrone 8 mg/m2, etoposide 80 mg/m2,
of GO (9 mg/m2) with IDAraC (1 g/m2) and mitoxan- and AraC 1 g/m2 daily for 6 days (MEC regimen) in
trone (12 mg/m2) was evaluated in 62 patients with combination with lintuzumab 12 mg/m2, or MEC
relapsed and refractory AML. Thirty-one patients alone. The CR rate was 28% in the group receiving
(50%) achieved a CR and 8 patients (13%) had a CR MEC alone and 36% in patients treated with MEC
with delayed platelet recovery. A significantly higher and lintuzumab (P = NS). The median OS was 156
OR rate was observed in relapsed (73%) than in re- days and was similar in both arms. The results of
fractory patients (39%; P = 0.007), and overall disease- these studies suggest that the addition of this mono-
free survival was 41% and 33% (P = 0.03), respec- clonal antibody to induction chemotherapy is well
tively. Candoni et al83 evaluated GO in combination tolerated but has no additional benefit compared
with IDAraC, idarubicin, and fludarabine in first-line with chemotherapy alone.
chemotherapy in patients aged <65 years. CR was
obtained in 26 of 29 treated patients (90%), and the Newer Nucleoside Analogues
probability of 1-year OS was 90%. Three newer nucleoside analogues—clofarabine,
The combination of low doses of GO with hydroxy- troxacitabine, and sapacitabine—have recently been
urea and azacitidine was assessed in previously un- studied in patients with AML.89,90 Clofarabine† is a
treated elderly patients.84 Treatment was initialized second-generation PNA synthesized to combine the
with hydroxyurea 1500 mg/m2 twice daily to decrease most favorable pharmacokinetic properties of flu-
white blood cell counts to <10,000/m2, followed by darabine and cladribine.89 This drug acts by inhibiting
azacitidine 75 mg/m2 subcutaneously for 7 days and ribonucleotide reductase and DNA polymerase, as
GO 3 mg/m2 on day 8. Consolidation therapy was well as by inducing apoptosis.91 In a Phase II study,
administered to those patients who achieved a CR. clofarabine monotherapy was used in 31 patients with
Fourteen of 18 patients (78%) achieved a CR, and the relapsed or refractory disease.92 The overall CR rate
median disease-free survival was 8 months. was 42%; however, for patients with a first remission
The results of these studies suggest that combina- lasting >12 months, the CR rate was 87%. A single-
tions of GO with chemotherapy are promising and center, Phase II trial was conducted with clofarabine
support the need for further investigations using well- as first-line treatment in 28 patients aged >70 years
designed randomized trials. with AML who were not suitable for conventional
Lintuzumab* is a humanized monoclonal antibody chemotherapy.93 Clofarabine was administered daily
consisting of a human immunoglobulin G1 frame- at a dosage of 30 mg/m2 IV for 5 days every 4 weeks.
work that contains human constant regions and Seventeen patients achieved CR, and grade 3/4 hepa-
complementarity-determining regions.61 Lintuzumb totoxicity was observed in 11 patients. In a multi-
induces cell death by complement-dependent cytotox- center, Phase II study of 26 elderly patients with newly
icity and antibody-directed cellular cytotoxicity.85 diagnosed AML who were ineligible for standard che-
Phase I studies found that lintuzumab induced a clini- motherapy, the OR rate was 44%, including a 21%
cal response in 5 of 23 patients with AML, including CR rate.94
one CR.86,87 In a Phase II trial, lintuzumab was inves-
†Trademark:Evoltra® (Genzyme Corporation, Cambridge,
*HuM195 (Seattle Genetics Inc., Bothell, Washington). Massachusetts).

2009 2357
Clinical Therapeutics

The results of the preclinical study and other en- studies reported synergistic effects with oxaliplatin,
couraging data from the single-agent experience led to gemcitabine, docetaxel, and doxorubicin in human
trials of clofarabine in combination with AraC.95,96 colon cancer cells.102 Sapacitabine is still in single-
Faderl et al95 reported the results of a Phase II study agent phases of development, and its activity in AML
in patients aged ≥50 years with previously untreated is not yet known.90
AML. For 5 days, clofarabine was given as a 1-hour IV
infusion at a dose of 40 mg/m2 followed 4 hours later Hypomethylating Agents
by AraC as a 2-hour infusion at a dose of 1 g/m2. The DNA methylation is the addition of a methyl group
OR rate was 60%, including a 52% CR rate. Subse- to specific stretches of DNA sequences often located
quently, in a randomized study, clofarabine alone was in or near promoter regions.103 DNA methylation
compared with clofarabine and low-dose AraC in occurs when a methyl group is attached to a cytosine
70 patients with AML aged ≥60 years.96 Sixteen pa- by 1 of the 3 known active DNA methyltransferases
tients received clofarabine 30 mg/m2 intravenously for (DNMTs). DNA methylation causes loss of gene func-
5 days and 54 patients received clofarabine with tion induced by epigenetic gene silencing. Such changes
20 mg/m2 AraC given SC for 14 days. The OR and CR may be reversible using methylation inhibitors. Aber-
rates were significantly higher in patients treated with rant DNA methylation has been described in several
the combination regimen than in those receiving clo- malignancies, including AML.104,105
farabine alone (67% vs 31% [P = 0.012] and 63% vs Two DNMT inhibitors, azacitidine‡ and decitabine§,
31% [P < 0.003], respectively). Toxicity was compa- are pyrimidine analogues of cytidine that can be incor-
rable in both groups. porated into RNA and/or DNA. Both agents were used
Troxacitabine* is an l-enantiomer nucleoside ana- at higher doses in salvage therapy of relapsed/
logue that differs from AraC in structure, mechanism refractory AML in the 1980s, but the treatment-related
of action, and resistance.97 The results of Phase I and toxicity, especially bone marrow suppression, was
I/II studies in AML revealed that prolonged infusion considerable.105,106 A retrospective analysis of 171 pa-
of this drug was more active than bolus adminis- tients with AML who were treated with higher doses of
tration.98–100 A Phase I study of troxacitabine given azacitidine found 18% had a CR, with an OR of
as a 30-minute IV infusion daily for 5 days found that 27%.106 Remission was achieved in a mean of 46 days
its MTD is 8 mg/m2/d.98 In this study in 30 patients (median, 112 days). More recently, lower doses of
with acute leukemia, the CR rate was 10%. In a larger azacitidine (75 mg/m2 SC on days 1–7) and decitabine
Phase I/II study of continuous troxacitabine infusion, (20 mg/m2 IV on days 1–5) were studied in myelodys-
an MTD of 12 mg/m2/d for 5 days was established.100 plastic syndrome (MDS) and AML, and were approved
Dose-limiting toxicities were mucositis and hand–foot by the FDA for the treatment of MDS.106,107 However,
syndrome. Seven patients (15%) achieved CR or CR lower doses of azacitidine and decitabine are also active
with incomplete platelet recovery. Troxacitabine was in AML and are being studied, especially in secondary
also evaluated in combination with AraC and/or ida- AML and AML in elderly patients.108,109
rubicin in untreated AML patients aged ≥50 years Sudan et al108 retrospectively analyzed 20 patients
with adverse karyotypes.101 However, neither troxacita- (median age, 66 years; age range, 40–80 years) with
bine combination was better than treatment with ida- AML who had bone marrow blast counts between
rubicin and AraC. 21% and 36% and who were treated with azacitidine
Sapacitabine† is a 2-deoxycytidine analogue that 75 mg/m2/d administered SC for 7 days every 4 weeks.
has displayed antiproliferative activity in a variety of Nine of the patients were originally reported as hav-
cell lines, including those shown to be resistant to ing MDS refractory anemia with excess blasts in
several anticancer drugs.102 The antiproliferative ef- transformation subtype. The OR was 60%, and the
fects of sapacitabine in terms of IC50 values were CR rate was 20%. The median survival of responders
better than those observed with AraC. Combination was 15 months. The most common toxic events were

*Trademark: Troxatyl® (SGX Pharmaceuticals, San Diego, ‡Trademark: Vidaza® (Celgene Corporation, Summit, New
California). Jersey).
† CYC682, CS-682 (Cyclacel Ltd., Dundee, United Kingdom). §Trademark: Dacogen® (Eisai Inc., Woodcliff Lake, New Jersey).

2358 Volume 31 Theme Issue


T. Robak and A. Wierzbowska

infections (observed in 8 patients). In another study, survival.113 Two farnesyltransferase inhibitors, tipi-
azacitidine was combined with phenylbutyrate in 29 pa- farnib* and lonafarnib†, are currently under develop-
tients with AML.110 All patients received azacitidine ment for the treatment of AML.114–117
25 to 75 mg/m2/d SC for 5, 10, or 14 days followed by Tipifarnib is an orally available, nonpeptidomi-
a continuous 7-day intravenous infusion of phenylbu- metic methylquinolone that has shown in vitro and in
tyrate. Overall, 11 of 29 evaluable patients responded, vivo activity against AML.115–117 Lancet at al117 inves-
including 14% with CR. Response duration ranged tigated tipifarnib as a single agent in 158 previously
from 8 to >19 months. untreated patients with poor-risk AML. The median
Decitabine has also been studied in patients with age was 74 years, and a majority of patients had ante-
AML.109,111,112 In a Phase II study, the drug was used cedent MDS. Tipifarnib was given orally at a dosage
in low doses of 20 mg/m2 over 1 hour for 5 consecu- of 600 mg twice daily for 21 days, followed by a rest
tive days every 4 weeks in untreated patients with period of up to 42 days to allow for recovery of pe-
AML aged >60 years.109 CR was obtained in 7 of ripheral blood counts. The CR rate was 14% and the
27 patients (26%). Garcia-Manero et al112 reported OR rate was 13%, with a median duration of response
results from a study of decitabine in combination with of 7.3 months. Unfortunately, tipifarnib showed no
the histone deacetylase inhibitor valproic acid in 54 un- benefit over best supportive care, including hydroxy-
treated patients with AML (median age, 60 years; age urea, in a large randomized Phase III study, which in-
range, 5–80 years). Decitabine was administered at a cluded 457 patients aged ≥70 years with newly diag-
dosage of 15 mg/m2 IV for 10 days concomitantly nosed AML.118 However, some patients with poor-risk
with escalating doses of valproic acid orally, also for AML, including patients with secondary AML and ad-
10 days. Twelve patients (22%) displayed an objective verse cytogenetics, may benefit from tipifarnib mainte-
response: 10 patients (19%) with CR and 2 (4%) with nance therapy.119 In addition, tipifarnib may be more
incomplete platelet recovery. The OS in responders active when combined with other antileukemic agents.
was 15.3 months (range, 20.2–46 months). This thera- Karp et al120 recently reported the results of a Phase
py was associated with DNA hypomethylation, induc- I trial of tipifarnib and etoposide. A total of 84 patients
tion of histone acetylation, and gene reactivation. aged >70 years received 222 cycles of tipifarnib at a
Blum et al111 reported the results of a similar Phase I dosage of 300 to 600 mg twice daily for 14 or 21 days
study in 25 patients (median age, 70 years), 12 previ- and oral etoposide at a dosage of 100 to 200 mg daily
ously untreated and 13 with relapsed AML. Estrogen re- on days 1 through 3 and days 8 through 10. CR was
ceptor promoter demethylation, global DNA hypomethy- achieved in 50% of two 14-day tipifarnib cohorts re-
lation, depletion of DNMT, and histone hyperacetylation ceiving either tipifarnib 600 mg with etoposide 100 mg
were observed. Response was achieved in 11 of 21 assess- or tipifarnib 400 mg with etoposide 200 mg. The re-
able patients (52%), including 4 with morphologic and sults of this study are encouraging in that they highlight
cytogenetic CRs. However, because the addition of val- the therapeutic progress in a population of poor-risk,
proic acid led to encephalopathy in some patients, no older AML patients who are very difficult to treat.
clinical benefit was observed compared with decitabine Lonafarnib is another orally bioavailable, farnesyl-
alone at a dosage of 20 mg/m2/d for 10 days. transferase inhibitor.121,122 In a Phase I study, this
agent was administered at a dosage of 200 mg twice
Farnesyltransferase Inhibitors daily for 3 courses of 4 weeks to 16 patients with
Farnesyltransferase is an enzyme involved in cell MDS and secondary AML.121 The median age was
signaling, proliferation, and differentiation. It cata- 70 years (age range, 53–77 years). Partial remission
lyzes the transfer of the farnesyl moiety to the cysteine was observed in 1 of 5 AML patients. This study indi-
terminal residue of a substrate protein in the Ras pro- cates that lonafarnib had limited activity in older pa-
tein.113 Farnesylation is an important posttransla- tients with secondary AML.
tional modification of Ras. Farnesylated protein prod-
ucts of the Ras gene family are frequently activated in
*Trademark: Zarnestra™ (Johnson & Johnson Pharmaceutical
MDS and AML. Farnesyltransferase inhibitors selec- Research & Development, L.L.C., Raritan, New Jersey).
tively target intracellular enzyme; they also affect an- † Trademark: Sarasar ® (Schering-Plough Corporation,

giogenesis, cellular adhesion, mitosis, and cellular Kenilworth, New Jersey).

2009 2359
Clinical Therapeutics

Laromustine Tyrosine Kinase Inhibitors


Laromustine* is an alkylating agent that belongs to FLT3 is a transmembrane enzyme that promotes
the group of sulfonyl hydrazine prodrugs. This drug proliferation after activation by ligand binding.76
spontaneously generates nucleophilic species that ef- FLT3 plays an important role in the survival and pro-
ficiently lead to DNA alkylation, resulting in DNA liferation of AML blasts. Several studies have shown
cross-linking and cell death.123 In in vitro studies, la- that FLT3 mutations are among the most prevalent
romustine showed significant antileukemic activity genetic abnormalities in AML and have been report-
against myeloid leukemia blast cells both as a single ed in 30% to 35% of patients with AML.45,129–131
agent and in combination with AraC or daunorubi- The most common FLT3–internal tandem duplication
cin.124 This agent was also active in patients with re- (FLT3-ITD) is observed in 25% to 30% of AML pa-
lapsed or refractory AML.125–128 In a Phase I study, tients. FLT3 mutations are associated with an adverse
28 patients with AML and 5 patients with MDS prognosis.130–132
received 52 courses of treatment.125 Dose escalation In recent years, a number of FLT3 inhibitors have
was terminated at 708 mg/m2 given as a single intra- been developed and tested in Phase I/II trials. Lestaurti-
venous infusion. In a subsequent Phase II study, nib† is an orally available, FLT3-selective indolocarba-
104 previously untreated patients (median age, 72 years; zole alkaloid compound synthetically derived from
age range, 60–84 years) were treated with laromustine compounds of microbial origin. In vitro studies noted
at a dosage of 600 mg/m2 intravenously, given as a preferential killing of AML cells with FLT3-ITD.133 In a
single dose.127 Patients were eligible to receive a sec- Phase I/II trial, lestaurtinib was evaluated in 14 patients
ond course if they had a response of less than CR or with relapsed, refractory, or poor-risk AML harboring
CR with incomplete platelet recovery after the first FLT3 mutations.134 The patients were given lestaurtinib
cycle. The OR rate was 32%, with 29 patients (28%) at an initial dosage of 60 mg orally twice daily. Five pa-
achieving a CR. Median OS was 94 days, with a tients had limited responses, including reduction in pe-
1-year survival of 14%. Nineteen patients (18%) ripheral blast count and transfusion frequency, with
died within 30 days of receiving laromustine therapy, minimal treatment-related toxicity. Knapper et al135
and 84% of the deaths occurred in patients with conducted a Phase II trial of lestaurtinib used as a single
pancytopenia. agent in previously untreated, elderly AML patients.
A randomized, double-blind, placebo-controlled Twenty-nine patients unfit for intensive chemotherapy
study of HDAraC with or without laromustine was (median age, 73 years; age range, 67–82 years) entered
subsequently conducted in patients with relapsed or the study. Initially, the drug was administered at a dos-
refractory disease.62 A total of 164 patients were en- age of 60 mg twice daily, with subsequent dose escala-
rolled (median age, 59 years; age range, 22–83 years). tion to 80 mg twice daily for 8 weeks. Transient reduc-
Patients received 1.5 g/m2/d of AraC in a continuous tion in bone marrow blasts and peripheral blood or
infusion for 3 days, with a subgroup of these patients reduction in blood transfusion was observed in 60% of
also receiving laromustine 600 mg/m2 on day 2. The patients with mutated FLT3 and in 23% of patients
CR rate was significantly higher for the laromustine with wild-type FLT3 mutations. However, the median
group (35%) than for those receiving AraC alone time to progression was 25 days.
(19%) (P = 0.005). However, early mortality was Tandutinib‡ is another quinazoline-based FLT3 in-
higher in patients treated with laromustine (11% vs hibitor with known activity in AML.136 In a Phase I
2%; P = 0.016). There were also no differences in trial, tandutinib was given orally in doses ranging
progression-free survival or OS rates in either group from 50 to 700 mg twice daily.137 Forty AML and
because of the excessive number of early deaths asso- high-risk MDS patients (median age, 70.5 years) were
ciated with combination therapy. Further studies included. The dose-limiting toxicity of the drug was re-
should concentrate on optimization of the laromus- versible and included generalized muscular weak-
tine doses in this patient population. ness and fatigue, which occurred at doses >525 mg.

†CEP701(Cephalon, Inc., Frazer, Pennsylvania).


*Trademark: Onrigin® (Vion Pharmaceuticals, Inc., New ‡MLN518 (Millenium Pharmaceuticals, Cambridge,
Haven, Connecticut). Massachusetts).

2360 Volume 31 Theme Issue


T. Robak and A. Wierzbowska

However, this Phase I trial was limited in its ability to ferences in terms of disease-free survival, OS, and OR
assess the antileukemic activity of tandutinib. rates in the interim analysis. Similarly, in a second trial
PKC412* was originally developed as a protein ki- comparing 2 cycles of induction therapy (AraC +
nase C inhibitor.138 However, it is also a potent inhibitor daunorubicin), one given with valspodar and one with-
of FLT3 phosphorylation and cell proliferation, and it out, there were no significant between-group differenc-
may induce leukemic cell apoptosis. PKC412 was evalu- es.145 In the updated analysis, there was even a nonsig-
ated as a single agent in patients with relapsed/refractory nificant trend for inferior response in the valspodar
AML and in MDS patients with FLT3 mutations.139 The arm due to a higher death rate during induction.146
drug was used at a dosage of 75 mg orally 3 times a A more potent third generation of P-gp modulator—
day. The peripheral blast count decreased by ≥50% in zosuquidar‡—has been under clinical investigation.147
14 patients, and bone marrow blast reduction by ≥50% Zosuquidar is a difluorocyclopropyl dibenzosuberane
was observed in 6 of the 20 patients. Two patients de- that binds allosterically to P-gp and inactivates its efflux
veloped fatal pulmonary events of unclear etiology. function. In a Phase I study, zosuquidar was given as a
PKC412 was also evaluated in combination with AraC 72-hour intravenous infusion together with induction
and daunorubicin in newly diagnosed AML patients therapy using AraC and daunorubicin.147 The recom-
aged ≤60 years.140 All 6 patients with FLT3 mutations mended dose of this agent for Phase II study was estab-
and 8 of 13 patients (62%) with wild-type FLT3 lished as a 72-hour continuous infusion at 700 mg/d.
achieved a CR. This combined regimen was generally Although the drug was generally well tolerated, its clini-
well tolerated, and no drug-related deaths occurred. The cal activity should be further defined in well-designed
preclinical observations and clinical studies suggest that clinical trials.
FLT3 inhibitors are promising agents in the treatment
of AML patients with FLT3 mutations, especially when HEMATOPOIETIC STEM CELL
they are used in combination with chemotherapy. TRANSPLANTATION
Use of alloSCT has been established as the most effec-
Multidrug-Resistant Modulators tive form of antileukemic therapy in patients with
P-glycoprotein (P-gp) is a cellular membrane pro- AML in first or subsequent remission.148,149 This
tein encoded by the MDR1 gene, which belongs to the treatment modality combines high-dose chemothera-
ABC superfamily and serves as an efflux pump to ex- py, with or without radiotherapy, with alloreactive
trude chemotherapeutic agents from the cell.6,131 immunotherapeutic effects against leukemia, also known
Overexpression of P-gp is an important mechanism of as graft-versus-leukemia activity. Several studies have
multidrug resistance in AML and has been incorpo- shown a reduction in relapse rate and an increased
rated into prognostic models for AML.141 disease-free survival benefit in AML patients undergo-
Several multidrug-resistant modulators, including ing transplantation in first CR, as well as in patients
cyclosporine and quinine in combination with chemo- with more advanced disease (second CR or primary
therapy, have been tested for the treatment of relapsed refractory disease), compared with patients treated
AML.6 In one study, the addition of cyclosporine to with chemotherapy alone.148–151 However, the benefi-
conventional induction treatment increased relapse-free cial effect of alloSCT on OS is in part attenuated by
survival and OS.59 However, in 2 other studies, no sig- the high nonrelapse mortality related to toxicity of
nificant difference was found in CR rate or disease-free high-dose conditioning, as well as the disparity of
survival and OS.142,143 host–recipient histocompatibility.30,49,152–154 There-
The addition of valspodar† to induction therapy was fore, alloSCT should be offered to those AML patients
tested in 2 randomized trials.144,145 In one study, AraC, with a relatively high risk of relapse and a relatively
daunorubicin, and etoposide were compared with an low risk of nonrelapse mortality.
identical regimen incorporating PSC-833.144 This study One of the most noteworthy areas of interest in the
was stopped prematurely because of increased early current clinical use of alloSCT includes the clarifica-
treatment-related mortality and lack of significant dif- tion of prognostic factors, which has led to improved

*PKX412 (Novartis Oncology, East Hanover, New Jersey). ‡LY335979 (Kanisa Pharmaceuticals, Inc./Lilly, San Diego,
† PSC-833 (Novartis Pharmaceuticals, East Hanover, New Jersey). California).

2009 2361
Clinical Therapeutics

risk determination. Currently, AML patients are mortality in AML patients undergoing transplanta-
stratified into 3 risk categories based on cytogenetics: tion in second CR.158 However, the outcome of
favorable, intermediate, and unfavorable. alloSCT recipients in terms of OS has been found to
be better than other patients treated with chemothera-
AlloSCT in Acute Myeloid Leukemia Patients in py alone or autotransplantation.157 In young patients
First Complete Remission lacking a sibling donor match, the alloSCT from a
There is evidence that sibling alloSCT improves well-matched unrelated donor is associated with 35%
disease-free survival and OS in patients with AML in to 40% disease-free survival and is comparable to re-
first CR.49,148,154,155 The findings from 5 major studies sults of alloSCT from sibling donors.159–161 Because
revealed a ~50% reduction in relapse rate in each cyto- the chance of surviving without a transplant is very
genetic risk group.49,148,152,154,155 Moreover, in a meta- low, the use of an alternative source such as a well-
analysis, a statistically significant OS benefit of 12% matched unrelated donor, cord blood, or haploidenti-
(hazard ratio, 0.84 [95% CI, 0.74–0.95]; P = 0.01) was cal family donors represents the sole possibility of
reported for all patients but those with good-risk cyto- survival.160 A similar approach may be followed in
genetics.148 In this group of patients, the relatively low primary refractory AML. There is evidence that ap-
relapse probability (≤35%) was offset by a relatively proximately one third of patients with primary re-
high nonrelapse mortality (≥20%). Therefore, myelo- fractory AML could achieve durable remission after
ablative alloSCT in first CR is not generally recom- alloSCT.151 In study by Fung et al,151 the 3-year prob-
mended for AML patients with favorable cytogenetics. abilities of disease-free survival, OS, and relapse rate
However, molecular genetics are being used to define for 68 patients with primary refractory AML were
new prognostic factors that often recategorize the new 31%, 30%, and 51%, respectively.
AML subset into another prognostic group, particu-
larly within the intermediate and favorable sub- Reduced-Intensity alloSCT in
groups.130 There is evidence that adults with favorable- Acute Myeloid Leukemia
risk AML but with c-kit mutations appear to do as During the last decade, new, less-intensive condi-
poorly with standard chemotherapy as patients with tioning regimens have been explored for alloSCT in
unfavorable cytogenetics.156 Thus, in these patients, older AML patients or in patients with significant
alloSCT might be considered in first CR. Similarly, the comorbidities who are considered incapable of toler-
mutational status of NPM1, FLT3, and CEBPA might ating traditional high-dose regimens. Several stud-
stratify AML patients with normal karyotypes into ies have confirmed that it is possible for older pa-
groups with better and worse prognosis. The patients tients with AML to undergo transplantation and that
with NPM1 or CEBPA mutations without FLT3-ITD, reduced-intensity alloSCT (RIC-alloSCT) is associated
which has also been associated with favorable progno- with lower morbidity and mortality compared with
sis, do not benefit from alloSCT in first CR.129 alloSCT with myeloablative conditioning.161–163 How-
Based on recent studies, therefore, it appears rea- ever, some trials have also found a concomitant in-
sonable to offer alloSCT from a matched sibling do- crease in relapse rates that attenuated the disease-free
nor in first CR for younger AML patients with poor- survival benefit.161,162 Other research appears to sug-
risk cytogenetics, intermediate-risk cytogenetics apart gest a better outcome for older patients treated with
from the NPM1+/FLT3-ITD and CEBPA+/FLT3-ITD RIC-alloSCT compared with conventional chemo-
subgroups, and favorable-risk cytogenetics with c-kit therapy.163 Larger studies are still required, however,
mutations. to better evaluate the long-term value of RIC-alloSCT
from sibling and unrelated donors in older patients
AlloSCT in Acute Myeloid Leukemia Patients with AML.
With Relapsed and Primary Refractory Disease
AlloSCT is the best treatment option for AML pa- AutoSCT in Acute Myeloid Leukemia
tients with relapsed and refractory disease151,157; how- A number of randomized AML trials have investi-
ever, the outcome beyond first CR is inferior to that in gated the role of autoSCT compared with other treat-
first CR. Retrospective data report 39% disease-free ment modalities.164 Over the past 10 years, a reduc-
survival and a 42% relapse rate and 32% nonrelapse tion in treatment-related mortality has been observed;

2362 Volume 31 Theme Issue


T. Robak and A. Wierzbowska

however, the results with autoSCT have remained REFERENCES


constant. In large intergroup studies,164 the treatment- 1. Estey E, Döhner H. Acute myeloid leukaemia. Lancet.
related mortality range in autoSCT was 3% to 14% 2006;368:1894–1907.
compared with 3% to 7% in conventional chemo- 2. National Cancer Institute. SEER cancer statistics review,
therapy. However, the relapse rate in the autoSCT 1975–2005. http://seer.cancer.gov./csr/1975_2005/. Ac-
cessed February 6, 2009.
group (35%–48%) was lower than that in conven-
3. Deschler B, Lübbert M. Acute myeloid leukemia: Epidemiolo-
tionally treated patients (60%–68%).164 In some stud-
gy and etiology. Cancer. 2006;107:2099–2107.
ies, a higher disease-free survival was evident in pa- 4. Aquino VM. Acute myelogenous leukemia. Curr Probl
tients assigned to autoSCT compared with those Pediatr Adolesc Health Care. 2002;32:50–58.
receiving conventional chemotherapy (48% vs 30%), 5. American Cancer Society. Cancer Facts and Figures 2008.
but no significant difference in OS was observed.165 In http://www.cancer.org/downloads/STT/Cancer_
most trials, the patients with poor-risk cytogenetics Statistics_2008.ppt#397,2,2008,Estimated US Cancer
benefited the least from autoSCT. AutoSCT produces Death. Accessed March 11, 2009.
durable second remission in 25% to 30% of patients 6. Laubach J, Rao AV. Current and emerging strategies for
with relapsed AML who have achieved second the management of acute myeloid leukemia in the elderly.
CR.166 Oncologist. 2008;13:1097–1108.
7. Ravandi F, Burnett AK, Agura ED, Kantarjian HM. Prog-
ress in the treatment of acute myeloid leukemia. Cancer.
CONCLUSIONS
2007;110:1900–1910.
Considerable advances have been made over the past 8. Tallman MS. New strategies for the treatment of acute
20 years in the prognosis and treatment of patients myeloid leukemia including antibodies and other novel
with AML. At present, risk-adapted therapy based on agents. Hematology Am Soc Hematol Educ Program. 2005:
chemotherapy response and cytogenetic abnormalities 143–150.
are recommended for use. However, there has been 9. King ME, Rowe JM. Recent developments in acute myelo-
little development in OS. Combinations of anthracy- genous leukemia therapy. Oncologist. 2007;12(Suppl 2):
clines and AraC are still the mainstay of induction 14–21.
therapy, and use of HDAraC is now a standard con- 10. Jabbour EJ, Estey E, Kantarjian HM. Adult acute myeloid
solidation therapy in AML patients aged <60 years. leukemia [published correction appears in Mayo Clin Proc.
For younger patients with poor-risk cytogenetics and 2006;81:569]. Mayo Clin Proc. 2006;81:247–260.
11. Tallman MS, Gilliland DG, Rowe JM. Drug therapy for
an available donor, alloSCT offers the best opportu-
acute myeloid leukemia [published correction appears in
nity for cure. For older patients and for younger pa-
Blood. 2005;106:2243]. Blood. 2005;106:1154–1163.
tients with relapsed or primary refractory disease,
12. Löwenberg B, Griffin JD, Tallman MS. Acute myeloid leu-
there is an obvious need to develop better strategies kemia and acute promyelocytic leukemia. Hematology Am
with new, more specific and active drugs. Recently, Soc Hematol Educ Program. 2003:82–101.
several new agents have been explored and have 13. Berman E, Heller G, Santorsa J, et al. Results of a random-
shown promise in treating AML. However, it is un- ized trial comparing idarubicin and cytosine arabinoside
likely that these agents will be curative when admin- with daunorubicin and cytosine arabinoside in adult pa-
istered as monotherapy; it is more likely that they tients with newly diagnosed acute myelogenous leukemia.
will be used in combination with other new agents or Blood. 1991;77:1666–1674.
with conventional therapy. 14. Wiernik PH, Banks PL, Case DC Jr, et al. Cytarabine plus
idarubicin or daunorubicin as induction and consolida-
ACKNOWLEDGMENTS tion therapy for previously untreated adult patients with
acute myeloid leukemia. Blood. 1992;79:313–319.
This work was supported in part by a grant (no. 503-
15. Vogler WR, Velez-Garcia E, Weiner RS, et al. A phase III
1093-1) from the Medical University of Lodz and by
trial comparing idarubicin and daunorubicin in combina-
the Foundation for the Development of Diagnostics tion with cytarabine in acute myelogenous leukemia: A
and Therapy (Warsaw, Poland). The authors have in- Southeastern Cancer Study Group study. J Clin Oncol.
dicated that they have no other conflicts of interest 1992;10:1103–1111.
regarding the content of this article. 16. Arlin Z, Case DC Jr, Moore J, et al, for the Lederle Coop-
The authors would like to thank Ms. Anna Rychter erative Group. Randomized multicenter trial of cytosine
for editorial assistance. arabinoside with mitoxantrone or daunorubicin in previ-

2009 2363
Clinical Therapeutics

ously untreated adult patients with 23. Bishop JF, Matthews JP, Young GA, myeloid leukemia in first remission.
acute nonlymphocytic leukemia et al. A randomized study of high- N Engl J Med. 1998;339:1649–1656.
(ANLL). Leukemia. 1990;4:177–183. dose cytarabine in induction in acute 31. Milligan DW, Grimwade D, Cullis
17. Bishop JF, Lowenthal RM, Joshua myeloid leukemia. Blood. 1996;87: JO, et al, for the British Committee
D, et al, for the Australian Leukemia 1710–1717. for Standards in Haematology.
Study Group. Etoposide in acute 24. Weick JK, Kopecky KJ, Appelbaum Guidelines on the management of
nonlymphocytic leukemia. Blood. FR, et al. A randomized investiga- acute myeloid leukaemia in adults.
1990;75:27–32. tion of high-dose versus standard- Br J Haematol. 2006;135:450–474.
18. Russo D, Malagola M, de V ivo A, et dose cytosine arabinoside with 32. Kohrt HE, Coutre SE. Optimizing
al. Multicentre phase III trial on daunorubicin in patients with previ- therapy for acute myeloid leukemia.
fludarabine, cytarabine (Ara-C), ously untreated acute myeloid leuke- J Natl Compr Canc Netw. 2008;6:
and idarubicin versus idarubicin, mia: A Southwest Oncology Group 1003–1016.
Ara-C and etoposide for induction study. Blood. 1996;88:2841–2851. 33. Mayer RJ, Davis RB, Schiffer CA,
treatment of younger, newly diag- 25. Büchner T, Hiddemann W, Wörmann et al, for the Cancer and Leukemia
nosed acute myeloid leukaemia pa- B, et al. Double induction strategy Group B. Intensive postremission
tients [published correction ap- for acute myeloid leukemia: The chemotherapy in adults with
pears in Br J Haematol. 2006;132: effect of high-dose cytarabine with acute myeloid leukemia. N Engl J
804]. Br J Haematol. 2005;131:172– mitoxantrone instead of standard- Med. 1994;331:896–903.
179. dose cytarabine with daunorubicin 34. Byrd JC, Dodge RK, Carroll A, et al.
19. Holowiecki J, Grosicki S, Robak T, and 6-thioguanine: A randomized Patients with t(8;21)(q22;q22) and
et al, for the Polish Adult Leukemia trial by the German AML Cooperative acute myeloid leukemia have supe-
Group (PALG). Addition of cladri- Group. Blood. 1999;93:4116–4124. rior failure-free and overall survival
bine to daunorubicin and cytarabine 26. Kern W, Estey EH. High-dose cyto- when repetitive cycles of high-dose
increases complete remission rate sine arabinoside in the treatment of cytarabine are administered. J Clin
after a single course of induction acute myeloid leukemia: Review of Oncol. 1999;17:3767–3775.
treatment in acute myeloid leuke- three randomized trials. Cancer. 2006; 35. Byrd JC, Ruppert AS, Mrózek K, et
mia. Multicenter, phase III study. 107:116–124. al. Repetitive cycles of high-dose
Leukemia. 2004;18:989–997. 27. Büchner T, Berdel WE, Schoch C, et cytarabine benefit patients with
20. Holowiecki J, Grosicki S, Kyrcz- al. Double induction containing acute myeloid leukemia and inv(16)
Krzemien S, et al. Addition of cladrib- either two courses or one course of (p13q22) or t(16;16)(p13;q22):
ine to the standard daunorubicine- high-dose cytarabine plus mitoxan- Results from CALGB 8461. J Clin
cytarabine (DA 3+7) remission trone and postremission therapy by Oncol. 2004;22:1087–1094.
induction protocol (DAC) contrary either autologous stem-cell trans- 36. Schlenk RF, Benner A, Krauter J, et
to adjunct of fludarabine (DAF) plantation or by prolonged mainte- al. Individual patient data-based
improves the overall survival in un- nance for acute myeloid leukemia. meta-analysis of patients aged 16
treated adults with acute myeloid J Clin Oncol. 2006;24:2480–2489. to 60 years with core binding factor
leukemia aged up to 60 Y: A multi- 28. Castaigne S, Chevret S, Archimbaud acute myeloid leukemia: A survey of
center, randomized, phase III PALG E, et al. Randomized comparison of the German Acute Myeloid Leukemia
AML 1/2004 DAF/DAC/DA study double induction and timed-sequential Intergroup. J Clin Oncol. 2004;22:
in 673 patients. Blood (ASH Annual induction to a “3 + 7” induction in 3741–3750.
Meeting Abstracts). 2008;112:Abstract adults with AML: Long-term analysis 37. Elonen E, Almqvist A, Hänninen A,
133. of the Acute Leukemia French Asso- et al. Comparison between four
21. AML Collaborative Group. A sys- ciation (ALFA) 9000 study. Blood. and eight cycles of intensive chemo-
tematic collaborative overview of 2004;104:2467–2474. therapy in adult acute myeloid leu-
randomized trials comparing idaru- 29. Rowe JM. Consolidation therapy: kemia: A randomized trial of the
bicin with daunorubicin (or other What should be the standard of Finnish Leukemia Group. Leukemia.
anthracyclines) as induction thera- care? Best Pract Res Clin Haematol. 1998;12:1041–1048.
py for acute myeloid leukaemia. Br 2008;21:53–60. 38. Sauter C, Berchtold W, Fopp M, et
J Haematol. 1998;103:100–109. 30. Cassileth PA, Harrington DP, Ap- al. Acute myelogenous leukaemia:
22. Robak T. Purine nucleoside ana- pelbaum FR, et al. Chemotherapy Maintenance chemotherapy after
logues in the treatment of myeloid compared with autologous or allo- early consolidation treatment does
leukemias. Leuk Lymphoma. 2003;44: geneic bone marrow transplanta- not prolong survival. Lancet. 1984;
391–409. tion in the management of acute 1:379–382.

2364 Volume 31 Theme Issue


T. Robak and A. Wierzbowska

39. Büchner T, Hiddemann W, Wör- potecan (CAT) for patients with re- with acute myeloid leukaemia in re-
mann B, et al, for the German fractory or relapsed acute leukemia. lapse: A Leukemia Intergroup study.
AMLCG. Acute myeloid leukemia in Leuk Lymphoma. 2000;36:479–484. Br J Haematol. 1992;82:337–346.
adults: Is postconsolidation main- 47. Ferrá C, Berlanga JJ, Gallardo D, et 54. Vogler WR, McCarley DL, Stagg M,
tenance therapy necessary? Int J He- al. Mitoxantrone, etoposide, carbo- et al, for the Southeastern Cancer
matol. 2000;72:285–289. platinum and ara-C combination Study Group trial. A phase III trial of
40. Büchner T, Hiddemann W, Berdel therapy (MECA) in refractory and high-dose cytosine arabinoside with
WE, et al, for the German AML Co- relapsed acute leukemia. Leuk Lym- or without etoposide in relapsed and
operative Group. 6-Thioguanine, phoma. 2000;39:583–590. refractory acute myelogenous leuke-
cytarabine, and daunorubicin (TAD) 48. Wierzbowska A, Robak T, Pluta A, mia. Leukemia. 1994;8:1847–1853.
and high-dose cytarabine and mi- et al, for the Polish Adult Leukemia 55. Ohno R, Naoe T, Kanamaru A, et
toxantrone (HAM) for induction, Group. Cladribine combined with al, for the Kohseisho Leukemia
TAD for consolidation, and either high doses of arabinoside cytosine, Study Group. A double-blind con-
prolonged maintenance by reduced mitoxantrone, and G-CSF (CLAG-M) trolled study of granulocyte colony-
monthly TAD or TAD-HAM-TAD and is a highly effective salvage regimen stimulating factor started two days
one course of intensive consolida- in patients with refractory and re- before induction chemotherapy in
tion by sequential HAM in adult lapsed acute myeloid leukemia of refractory acute myeloid leukemia.
patients at all ages with de novo the poor risk: A final report of the Blood. 1994;83:2086–2092.
acute myeloid leukemia (AML): A Polish Adult Leukemia Group. Eur J 56. Karanes C, Kopecky KJ, Head DR,
randomized trial of the German AML Haematol. 2008;80:115–126. et al. A phase III comparison of high
Cooperative Group. J Clin Oncol. 49. Burnett AK, Wheatley K, Goldstone dose ARA-C (HIDAC) versus HIDAC
2003;21:4496–4504. AH, et al, for the Medical Research plus mitoxantrone in the treatment
41. Büchner T, Krug U, Berdel WE, et Council Adult and Paediatric Work- of first relapsed or refractory acute
al. Maintenance for acute myeloid ing Parties. The value of allogeneic myeloid leukemia Southwest On-
leukemia revisited. Curr Treat Options bone marrow transplant in patients cology Group Study. Leuk Res.
Oncol. 2007;8:296–304. with acute myeloid leukaemia at 1999;23:787–794.
42. Robak T, Wrzesień-Kuś A. The search differing risk of relapse: Results of 57. Thomas X, Fenaux P, Dombret H, et
for optimal treatment in relapsed the UK MRC AML 10 trial. Br J Hae- al. Granulocyte-macrophage colony-
and refractory acute myeloid leuke- matol. 2002;118:385–400. stimulating factor (GM-CSF) to
mia. Leuk Lymphoma. 2002;43:281– 50. Kell J. Treatment of relapsed acute increase efficacy of intensive se-
291. myeloid leukaemia. Rev Recent Clin quential chemotherapy with etopo-
43. Mato AR, Morgans A, Luger SM. Trials. 2006;1:103–111. side, mitoxantrone and cytarabine
Novel strategies for relapsed and re- 51. Milligan DW, Wheatley K, Little- (EMA) in previously treated acute
fractory acute myeloid leukemia. Curr wood T, et al, for the NCRI Haema- myeloid leukemia: A multicenter
Opin Hematol. 2008;15:108–114. tological Oncology Clinical Studies randomized placebo-controlled trial
44. Stahnke K, Boos J, Bender-Götze C, Group. Fludarabine and cytosine (EMA91 Trial). Leukemia. 1999;13:
et al. Duration of first remission are less effective than standard ADE 1214–1220.
predicts remission rates and long- chemotherapy in high-risk acute 58. Liu Yin JA, Wheatley K, Rees JK, Bur-
term survival in children with re- myeloid leukemia, and addition of nett AK, for the UK MRC Adult Leu-
lapsed acute myelogenous leukemia. G-CSF and ATRA are not beneficial: kemia Working Party. Comparison of
Leukemia. 1998;12:1534–1538. Results of the MRC AML-HR ran- ‘sequential’ versus ‘standard’ chemo-
45. Kern W, Schoch C, Haferlach T, et domized trial. Blood. 2006;107:4614– therapy as re-induction treatment,
al. Multivariate analysis of prognos- 4622. with or without cyclosporine, in
tic factors in patients with refrac- 52. Martiat P, Ghilain JM, Ferrant A, et refractory/relapsed acute myeloid
tory and relapsed acute myeloid al. High-dose cytosine arabinoside leukaemia (AML): Results of the UK
leukemia undergoing sequential and amsacrine or mitoxantrone in Medical Research Council AML-R tri-
high-dose cytosine arabinoside and relapsed and refractory acute myel- al. Br J Haematol. 2001;113:713–726.
mitoxantrone (S-HAM) salvage oid leukemia: A prospective ran- 59. List AF, Kopecky KJ, Willman CL, et
therapy: Relevance of cytogenetic domized study. Eur J Haematol. al. Benefit of cyclosporine modula-
abnormalities. Leukemia. 2000;14: 1990;45:164–167. tion of drug resistance in patients
226–231. 53. Larson RA, Day RS, Azarnia N, et al. with poor-risk acute myeloid leuke-
46. Cortes J, Estey E, Beran M, et al. The selective use of AMSA following mia: A Southwest Oncology Group
Cyclophosphamide, ara-C and to- high-dose cytarabine in patients study. Blood. 2001;98:3212–3220.

2009 2365
Clinical Therapeutics

60. Greenberg PL, Lee SJ, Advani R, et rabine, cytarabine, idarubicin and 76. Blum W, Marcucci G. New ap-
al. Mitoxantrone, etoposide, and G-CSF) in the treatment of patients proaches in acute myeloid leuke-
cytarabine with or without valspo- with high-risk myeloid malignancies. mia. Best Pract Res Clin Haematol.
dar in patients with relapsed or re- Leuk Res. 2002;26:725–730. 2008;21:29–41.
fractory acute myeloid leukemia 68. Clavio M, Carrara P, Miglino M, et al. 77. Stasi R, Evangelista ML, Buccisano
and high-risk myelodysplastic syn- High efficacy of fludarabine- F, et al. Gemtuzumab ozogamicin
drome: A phase III trial (E2995). containing therapy (FLAG-FLANG) in the treatment of acute myeloid
J Clin Oncol. 2004;22:1078–1086. in poor risk acute myeloid leukemia. leukemia. Cancer Treat Rev. 2008;34:
61. Feldman EJ, Brandwein J, Stone R, Haematologica. 1996;81:513–520. 49–60.
et al. Phase III randomized multi- 69. Carella AM, Cascavilla N, Greco 78. Stone RM. Targeted agents in AML:
center study of a humanized anti- MM, et al. Treatment of “poor risk” Much more to do. Best Pract Res Clin
CD33 monoclonal antibody, lintu- acute myeloid leukemia with flu- Haematol. 2007;20:39–48.
zumab, in combination with chemo- darabine, cytarabine and G-CSF 79. Sievers EL, Appelbaum FR, Spiel-
therapy, versus chemotherapy alone (flag regimen): A single center study. berger RT, et al. Selective ablation
in patients with refractory or first- Leuk Lymphoma. 2001;40:295–303. of acute myeloid leukemia using anti-
relapsed acute myeloid leukemia. 70. Wrzesień
-KuśA, Robak T, Lech-Marán- body-targeted chemotherapy: A
J Clin Oncol. 2005;23:4110–4116. da E, et al, for the Polish Adult Leuke- phase I study of an anti-CD33 cali-
62. Giles FJ, Stock W, Vey N, et al. A mia Group. A multicenter, open, non- cheamicin immunoconjugate. Blood.
double blind placebo-controlled comparative, phase II study of the 1999;93:3678–3684.
randomized phase III study of high combination of cladribine (2-chlo- 80. Stapnes C, Gjertsen BT, Reikvam H,
dose continuous infusion cytosine rodeoxyadenosine), cytarabine, and Bruserud Ø. Targeted therapy in acute
arabinoside (araC) with or without G-CSF as induction therapy in re- myeloid leukaemia: Current status
Cloretazine® in patients with first fractory acute myeloid and future directions. Expert Opin
relapse of acute myeloid leukemia leukemia—a report of the Polish Investig Drugs. 2009;18:433–455.
(AML). Blood (ASH Annual Meeting Adult Leukemia Group (PALG). Eur 81. Gleissner B, Schlenk R, Bornhäuser
Abstracts). 2006;108:Abstract 1970. J Haematol. 2003;71:155–162. M, Berdel WE. Gemtuzumab ozo-
63. Estey EH. Treatment of relapsed and 71. Pastore D, Specchia G, Carluccio P, gamicin (Mylotarg) for the treat-
refractory acute myelogenous leuke- et al. FLAG-IDA in the treatment of ment of acute myeloid leukemia—
mia. Leukemia. 2000;14:476–479. refractory/relapsed acute myeloid ongoing trials. Onkologie. 2007;30:
64. Robak T, Wierzbowska A, Robak E. leukemia: Single-center experience. 657–662.
Recent clinical trials of cladribine in Ann Hematol. 2003;82:231–235. 82. Chevallier P, Delaunay J, Turlure P, et
hematological malignancies and 72. Hänel M, Friedrichsen K, Hänel A, al. Long-term disease-free survival af-
autoimmune disorders. Rev Recent et al. Mito-flag as salvage therapy ter gemtuzumab, intermediate-dose
Clin Trials. 2006;1:15–34. for relapsed and refractory acute cytarabine, and mitoxantrone in pa-
65. Steinmetz HT, Schulz A, Staib P, et myeloid leukemia. Onkologie. 2001; tients with CD33(+) primary resistant
al. Phase-II trial of idarubicin, flu- 24:356–360. or relapsed acute myeloid leukemia.
darabine, cytosine arabinoside, and 73. Huhmann IM, Watzke HH, Geissler J Clin Oncol. 2008;26:5192–5197.
filgrastim (Ida-FLAG) for treatment K, et al. FLAG (fludarabine, cytosine 83. Candoni A, Martinelli G, Toffoletti
of refractory, relapsed, and second- arabinoside, G-CSF) for refractory E, et al. Gemtuzumab-ozogamicin
ary AML. Ann Hematol. 1999;78: and relapsed acute myeloid leukemia. in combination with fludarabine,
418–425. Ann Hematol. 1996;73:265–271. cytarabine, idarubicin (FLAI-GO) as
66. Jackson G, Taylor P, Smith GM, 74. Camera A, Rinaldi CR, Palmieri S, induction therapy in CD33-positive
et al. A multicentre, open, non- et al. Sequential continuous infu- AML patients younger than 65 years.
comparative phase II study of a com- sion of fludarabine and cytarabine Leuk Res. 2008;32:1800–1808.
bination of fludarabine phosphate, associated with liposomal dauno- 84. Nand S, Godwin J, Smith S, et al.
cytarabine and granulocyte colony- rubicin (DaunoXome) (FLAD) in Hydroxyurea, azacitidine and gem-
stimulating factor in relapsed and primary refractory or relapsed adult tuzumab ozogamicin therapy in pa-
refractory acute myeloid leukaemia acute myeloid leukemia patients. tients with previously untreated
and de novo refractory anaemia with Ann Hematol. 2009;88:151–158. non-M3 acute myeloid leukemia
excess of blasts in transformation. Br J 75. Giles F, Estey E, O’Brien S. Gemtu- and high-risk myelodysplastic syn-
Haematol. 2001;112:127–137. zumab ozogamicin in the treatment dromes in the elderly: Results from
67. de la Rubia J, Regadera A, Martín G, of acute myeloid leukemia. Cancer. a pilot trial. Leuk Lymphoma. 2008;
et al. FLAG-IDA regimen (fluda- 2003;98:2095–2104. 49:2141–2147.

2366 Volume 31 Theme Issue


T. Robak and A. Wierzbowska

85. Vitale C, Romagnani C, Falco M, 94. Burnett AK, Baccarani M, John- 2-deoxycytidine-derivative, in hu-
et al. Engagement of p75/AIRM1 son P, et al. Effectiveness of clo- man cancer cells. Br J Cancer. 2007;
or CD33 inhibits the proliferation farabine in elderly AML patients 97:628–636.
of normal or leukemic myeloid with adverse cytogenetics unfit for 103. Ma WW, Adjei AA. Novel agents
cells. Proc Natl Acad Sci U S A. 1999; intensive chemotherapy. Blood (ASH on the horizon for cancer therapy.
96:15091–15096. Annual Meeting Abstracts). 2006; CA Cancer J Clin. 2009;59:111–137.
86. Caron PC, Jurcic JG, Scott AM, et 108:Abstract 1985. 104. Garcia-Manero G. Demethylating
al. A phase 1B trial of humanized 95. Faderl S, Verstovsek S, Cortes J, et agents in myeloid malignancies.
monoclonal antibody M195 (anti- al. Clofarabine and cytarabine Curr Opin Oncol. 2008;20:705–710.
CD33) in myeloid leukemia: Spe- combination as induction therapy 105. Kuendgen A, Germing U. Emerg-
cific targeting without immunoge- for acute myeloid leukemia (AML) ing treatment strategies for acute
nicity. Blood. 1994;83:1760–1768. in patients 50 years of age or old- myeloid leukemia (AML) in the
87. Caron PC, Dumont L, Scheinberg er. Blood. 2006;108:45–51. elderly. Cancer Treat Rev. 2009;35:
DA. Supersaturating infusional hu- 96. Faderl S, Ravandi F, Huang X, et 97–120.
manized anti-CD33 monoclonal al. A randomized study of clofara- 106. Saiki JH, McCredie KB, Vietti TJ, et
antibody HuM195 in myelogenous bine versus clofarabine plus low- al. 5-Azacytidine in acute leuke-
leukemia. Clin Cancer Res. 1998;4: dose cytarabine as front-line thera- mia. Cancer. 1978;42:2111–2114.
1421–1428. py for patients aged 60 years and 107. Fenaux P, Mufti GJ, Hellstrom-
88. Feldman E, Kalaycio M, Weiner G, older with acute myeloid leukemia Lindberg E, et al, for the Interna-
et al. Treatment of relapsed or re- and high-risk myelodysplastic tional Vidaza High-Risk MDS Sur-
fractory acute myeloid leukemia syndrome. Blood. 2008;112:1638– vival Study Group. Efficacy of
with humanized anti-CD33 mono- 1645. azacitidine compared with that of
clonal antibody HuM195. Leukemia. 97. Giles FJ. Troxacitabine-based thera- conventional care regimens in the
2003;17:314–318. py of refractory leukemia. Expert Rev treatment of higher-risk myelodys-
89. Robak T, Korycka A, Lech-Maranda Anticancer Ther. 2002;2:261–266. plastic syndromes: A randomised,
E, Robak P. Current status of older 98. Giles FJ, Cortes JE, Baker SD, et al. open-label, phase III study. Lancet
and new purine nucleoside ana- Troxacitabine, a novel dioxolane Oncol. 2009;10:223–232.
logues in the treatment of lympho- nucleoside analog, has activity in 108. Sudan N, Rossetti JM, Shadduck
proliferative diseases. Molecules. patients with advanced leukemia. RK, et al. Treatment of acute
2009;14:1183–1226. J Clin Oncol. 2001;19:762–771. myelogenous leukemia with out-
90. Faderl S, Gandhi V, Kantarjian HM. 99. Giles FJ, Garcia-Manero G, Cortes patient azacitidine. Cancer. 2006;
Potential role of novel nucleoside JE, et al. Phase II study of troxacit- 107:1839–1843.
analogs in the treatment of acute abine, a novel dioxolane nucleo- 109. Cashen A, Schiller GJ, Larsen JS, et
myeloid leukemia. Curr Opin Hema- side analog, in patients with re- al. Phase II study of low-dose
tol. 2008;15:101–107. fractory leukemia. J Clin Oncol. decitabine for the front-line treat-
91. Lech-Maranda E, Korycka A, Robak 2002;20:656–664. ment of older patients with acute
T. Pharmacological and clinical 100. Roboz GJ, Giles FJ, Ritchie EK, et myeloid leukemia (AML). Blood
studies on purine nucleoside ana- al. Phase I/II study of continuous- (ASH Annual Meeting Abstracts).
logs—new anticancer agents. Mini infusion troxacitabine in refracto- 2006;108:Abstract 1984.
Rev Med Chem. 2006;6:575–581. ry acute myeloid leukemia. J Clin 110. Gore SD, Baylin S, Sugar E, et al.
92. Kantarjian H, Gandhi V, Cortes J, et Oncol. 2007;25:10–15. Combined DNA methyltransferase
al. Phase 2 clinical and pharmaco- 101. Giles FJ, Kantarjian HM, Cortes and histone deacetylase inhibition
logic study of clofarabine in pa- JE, et al. Adaptive randomized in the treatment of myeloid neo-
tients with refractory or relapsed study of idarubicin and cytarabine plasms. Cancer Res. 2006;66:6361–
acute leukemia. Blood. 2003;102: versus troxacitabine and cytara- 6369.
2379–2386. bine versus troxacitabine and idaru- 111. Blum W, Klisovic RB, Hackanson B,
93. Burnett AK, Russell N, Kell JW, et al. bicin in untreated patients 50 years et al. Phase I study of decitabine
A Phase 2 evaluation of single agent or older with adverse karyotype alone or in combination with valpro-
clofarabine as first line treatment for acute myeloid leukemia. J Clin On- ic acid in acute myeloid leukemia.
older patients with AML who are not col. 2003;21:1722–1727. J Clin Oncol. 2007;25:3884–3891.
considered fit for intensive chemo- 102. Serova M, Galmarini CM, Ghoul 112. Garcia-Manero G, Kantarjian HM,
therapy. Blood (ASH Annual Meeting A, et al. Antiproliferative effects of Sanchez-Gonzalez B, et al. Phase
Abstracts). 2004;104:Abstract 869. sapacitabine (CYC682), a novel 1/2 study of the combination of

2009 2367
Clinical Therapeutics

5-aza-2-deoxycytidine with val- risk features. Clin Cancer Res. 127. Giles F, Rizzieri D, Karp J, et al.
proic acid in patients with leuke- 2008;14:3077–3082. Cloretazine (VNP40101M), a novel
mia. Blood. 2006;108:3271–3279. 120. Karp JE, Flatten K, Feldman EJ, et sulfonylhydrazine alkylating agent,
113. Braun T, Fenaux P. Farnesyltrans- al. Active oral regimen for elderly in patients age 60 years or older
ferase inhibitors and their poten- adults with newly diagnosed acute with previously untreated acute
tial role in therapy for myelodys- myelogenous leukemia: A preclini- myeloid leukemia. J Clin Oncol.
plastic syndromes and acute cal and phase 1 trial of the farnesyl- 2007;25:25–31.
myeloid leukaemia. Br J Haematol. transferase inhibitor tipifarnib 128. Giles F, Verstovsek S, Faderl S, et
2008;141:576–586. (R115777, Zarnestra) combined al. A phase II study of cloretazine
114. Gore SD, Hermes-DeSantis ER. with etoposide. Blood. 2009;113: (VNP40101M), a novel sulfonyl-
Future directions in myelodysplas- 4841–4852. hydrazine alkylating agent, in pa-
tic syndrome: Newer agents and the 121. Ravoet C, Mineur P, Robin V, et tients with very high risk relapsed
role of combination approaches. al. Farnesyl transferase inhibitor acute myeloid leukemia. Leuk Res.
Cancer Control. 2008;15(Suppl):40– (lonafarnib) in patients with my- 2006;30:1591–1595.
49. elodysplastic syndrome or second- 129. Schlenk RF, Döhner K, Krauter J,
115. Korycka A, Smolewski P, Robak T. ary acute myeloid leukaemia: A et al, for the German-Austrian
The influence of farnesyl protein phase II study. Ann Hematol. Acute Myeloid Leukemia Study
transferase inhibitor R115777 2008;87:881–885. Group. Mutations and treatment
(Zarnestra) alone and in combi- 122. Feldman EJ, Cortes J, DeAngelo outcome in cytogenetically normal
nation with purine nucleoside ana- DJ, et al. On the use of lonafarnib acute myeloid leukemia. N Engl J
logs on acute myeloid leukemia in myelodysplastic syndrome and Med. 2008;358:1909–1918.
progenitors in vitro. Eur J Haema- chronic myelomonocytic leukemia. 130. Mrózek K, Marcucci G, Paschka P,
tol. 2004;73:418–426. Leukemia. 2008;22:1707–1711. et al. Clinical relevance of muta-
116. Karp JE, Lancet JE, Kaufmann SH, 123. Finch RA, Shyam K, Penketh tions and gene-expression changes
et al. Clinical and biologic activity PG, Sartorelli AC. 1,2- in adult acute myeloid leukemia
of the farnesyltransferase inhibitor Bis(methylsulfonyl)-1-(2- with normal cytogenetics: Are we
R115777 in adults with refractory chloroethyl)-2-(methylamino) ready for a prognostically priori-
and relapsed acute leukemias: A carbonylhydrazine(101M): A nov- tized molecular classification?
phase 1 clinical-laboratory correla- el sulfonylhydrazine prodrug with Blood. 2007;109:431–448.
tive trial. Blood. 2001;97:3361– broad-spectrum antineoplastic 131. Fröhling S, Schlenk RF, Breitruck J,
3369. activity. Cancer Res. 2001;61:3033– et al, for the AML Study Group
117. Lancet JE, Gojo I, Gotlib J, et al. A 3038. Ulm. Acute myeloid leukemia.
phase 2 study of the farnesyltrans- 124. Morris KL, Adams JA, Liu Yin JA. Prognostic significance of activat-
ferase inhibitor tipifarnib in poor- Effect of cloretazine (VNP40101M) ing FLT3 mutations in younger
risk and elderly patients with previ- on acute myeloid leukaemia blast adults (16 to 60 years) with acute
ously untreated acute myelogenous cells in vitro as a single agent and myeloid leukemia and normal cy-
leukemia. Blood. 2007;109:1387– combined with cytarabine and togenetics: A study of the AML
1394. daunorubicin. Leuk Res. 2009;33: Study Group Ulm. Blood. 2002;
118. Harousseau JL, Martinelli G, Je- 1096–1099. 100:4372–4380.
drzejczak WW, et al, for the 125. Giles F, Thomas D, Garcia-Mane- 132. Kolitz JE. Current therapeutic
FIGHT-AML-301 Investigators. A ro G, et al. A Phase I and pharma- strategies for acute myeloid leu-
randomized phase 3 study of tipi- cokinetic study of VNP40101M, a kaemia. Br J Haematol. 2006;134:
farnib compared with best sup- novel sulfonylhydrazine alkylating 555–572.
portive care, including hydroxyu- agent, in patients with refractory 133. Brown P, Meshinchi S, Levis M, et
rea, in the treatment of newly leukemia. Clin Cancer Res. 2004;10: al. Pediatric AML primary samples
diagnosed acute myeloid leuke- 2908–2917. with FLT3/ITD mutations are pref-
mia in patients 70 years or older. 126. Giles F, Verstovsek S, Thomas D, erentially killed by FLT3 inhibition.
Blood. 2009;114:1166–1173. et al. Phase I study of cloretazine Blood. 2004;104:1841–1849.
119. Karp JE, Smith BD, Gojo I, et al. (VNP40101M), a novel sulfonyl- 134. Smith BD, Levis M, Beran M, et al.
Phase II trial of tipifarnib as main- hydrazine alkylating agent, com- Single-agent CEP-701, a novel
tenance therapy in first complete bined with cytarabine in patients FLT3 inhibitor, shows biologic
remission in adults with acute with refractory leukemia. Clin Cancer and clinical activity in patients
myelogenous leukemia and poor- Res. 2005;11:7817–7824. with relapsed or refractory acute

2368 Volume 31 Theme Issue


T. Robak and A. Wierzbowska

myeloid leukemia. Blood. 2004; mia identifies novel biologic clus- inhibition in patients with acute
103:3669–3676. ters for risk classification and out- myeloid leukemia treated with zo-
135. Knapper S, Burnett AK, Little- come prediction. Blood. 2006;108: suquidar trihydrochloride, dauno-
wood T, et al. A phase 2 trial of 685–696. rubicin and cytarabine. Haemato-
the FLT3 inhibitor lestaurtinib 142. Solary E, Witz B, Caillot D, et al. logica. 2004;89:782–790.
(CEP701) as first-line treatment Combination of quinine as a po- 148. Cornelissen JJ, van Putten WL,
for older patients with acute my- tential reversing agent with mitox- Verdonck LF, et al. Results of a
eloid leukemia not considered fit antrone and cytarabine for the HOVON/SAKK donor versus no-
for intensive chemotherapy. Blood. treatment of acute leukemias: A donor analysis of myeloablative
2006;108:3262–3270. randomized multicenter study. HLA-identical sibling stem cell
136. Cheng Y, Paz K. Tandutinib, an Blood. 1996;88:1198–1205. transplantation in first remission
oral, small-molecule inhibitor of 143. Daenen S, van der Holt B, Verhoef acute myeloid leukemia in young
FLT3 for the treatment of AML GE, et al. Addition of cyclosporin and middle-aged adults: Benefits
and other cancer indications. A to the combination of mitoxan- for whom? Blood. 2007;109:3658–
IDrugs. 2008;11:46–56. trone and etoposide to overcome 3666.
137. DeAngelo DJ, Stone RM, Heaney resistance to chemotherapy in re- 149. Vicente D, Lamparelli T, Gualandi
ML, et al. Phase 1 clinical results fractory or relapsing acute myelo- F, et al. Improved outcome in
with tandutinib (MLN518), a nov- id leukaemia: A randomised phase young adults with de novo acute
el FLT3 antagonist, in patients II trial from HOVON, the Dutch- myeloid leukemia in first remis-
with acute myelogenous leukemia Belgian Haemato-Oncology Work- sion, undergoing an allogeneic
or high-risk myelodysplastic syn- ing Group for adults. Leuk Res. bone marrow transplant. Bone Mar-
drome: Safety, pharmacokinetics, 2004;28:1057–1067. row Transplant. 2007;40:349–354.
and pharmacodynamics. Blood. 144. Baer MR, George SL, Dodge RK, 150. Gorin NC, Labopin M, Fouillard
2006;108:3674–3681. et al. Phase 3 study of the multidrug L, et al, for the Eurpoean Coop-
138. Knapper S, Mills KI, Gilkes AF, et resistance modulator PSC-833 erative Group for Blood and Mar-
al. The effects of lestaurtinib in previously untreated patients row Transplantation (EBMT).
(CEP701) and PKC412 on prima- 60 years of age and older with Retrospective evaluation of autolo-
ry AML blasts: The induction of acute myeloid leukemia: Cancer gous bone marrow transplanta-
cytotoxicity varies with depen- and Leukemia Group B Study 9720. tion vs allogeneic bone marrow
dence on FLT3 signaling in both Blood. 2002;100:1224–1232. transplantation from an HLA iden-
FLT3-mutated and wild-type cas- 145. van der Holt B, Löwenberg B, Bur- tical related donor in acute myelo-
es. Blood. 2006;108:3494–3503. nett AK, et al. The value of the cytic leukemia. Bone Marrow Trans-
139. Stone RM, DeAngelo DJ, Klimek V, MDR1 reversal agent PSC-833 in plant. 1996;18:111–117.
et al. Patients with acute myeloid addition to daunorubicin and 151. Fung HC, Stein A, Slovak M, et al.
leukemia and an activating muta- cytarabine in the treatment of el- A long-term follow-up report on
tion in FLT3 respond to a small- derly patients with previously un- allogeneic stem cell transplanta-
molecule FLT3 tyrosine kinase in- treated acute myeloid leukemia tion for patients with primary re-
hibitor, PKC412. Blood. 2005;105: (AML), in relation to MDR1 status fractory acute myelogenous leuke-
54–60. at diagnosis. Blood. 2005;106: mia: Impact of cytogenetic
140. Stone RM, Fischer T, Paquette R, 2646–2654. characteristics on transplantation
et al. Phase IB study of PKC412, 146. Burnett AK, Milligan D, Goldstone outcome. Biol Blood Marrow Trans-
an oral FLT3 kinase inhibitor, in A, et al, for the United Kingdom plant. 2003;9:766–771.
sequential and simultaneous com- National Cancer Research Insti- 152. Keating S, de Witte T, Suciu S, et
binations with daunorubicin and tute Haematological Oncology al, for the European Organization
cytarabine (DA) induction and Study Group. The impact of dose for Research and Treatment of
high-dose cytarabine consolida- escalation and resistance modula- Cancer, Gruppo Italiano Mallattie
tion in newly diagnosed patients tion in older patients with acute Ematologiche Maligne dell’Adulto.
with AML. Blood (ASH Annual Meet- myeloid leukaemia and high risk The influence of HLA-matched
ing Abstracts). 2005;106:Abstract myelodysplastic syndrome: The sibling donor availability on treat-
404. results of the LRF AML 14 trial. Br ment outcome for patients with
141. Wilson CS, Davidson GS, Martin J Haematol. 2009;145:318–332. AML: An analysis of the AML 8A
SB, et al. Gene expression profil- 147. Gerrard G, Payne E, Baker RJ, et al. study of the EORTC Leukaemia
ing of adult acute myeloid leuke- Clinical effects and P-glycoprotein Cooperative Group and GIMEMA.

2009 2369
Clinical Therapeutics

Br J Haematol. 1998;102:1344– experience. Bone Marrow Transplant. stem cell transplantation (HSCT)
1353. 2000;26:397–404. in elderly patients with acute
153. Slovak ML, Kopecky KJ, Cassileth 160. Rocha V, Labopin M, Sanz G, et myeloid leukemia (AML) and
PA, et al. Karyotypic analysis pre- al, for the Acute Leukemia Work- high-risk myelodysplastic syndrome
dicts outcome of preremission ing Party of European Blood and (MDS). Blood. 2007;109:1395–
and postremission therapy in adult Marrow Transplant Group, Euro- 1400.
acute myeloid leukemia: A South- cord-Netcord Registry. Trans- 164. Levi I, Grotto I, Yerushalmi R, et al.
west Oncology Group/Eastern plants of umbilical-cord blood or Meta-analysis of autologous bone
Cooperative Group Study. Blood. bone marrow from unrelated do- marrow transplantation versus
2000;96:4075–4083. nors in adults with acute leukemia. chemotherapy in adult patients
154. Suciu S, Mandelli F, De Witte T, et N Engl J Med. 2004;351:2276–2285. with acute myeloid leukemia in
al, for the EORTC and GIMENA 161. Hegenbart U, Niederwieser D, first remission. Leuk Res. 2004;28:
Leukemia Groups. Allogeneic Sandmaier BM, et al. Treatment 605–612.
compared with autologous stem for acute myelogenous leukemia 165. Zittoun RA, Mandelli F, Willemze
cell transplantation in the treat- by low-dose, total-body, irradia- R, et al, for the European Organi-
ment of patients younger than tion-based conditioning and he- zation for Research and Treat-
46 years old with acute myeloid matopoietic cell transplantation ment of Cancer (EORTC) and the
leukemia (AML) in first complete from related and unrelated do- Gruppo Italiano Malattie Ema-
remission (CR1): An intention to nors. J Clin Oncol. 2006;24:444– tologiche Maligne dell’Adulto
treat analysis of the EORTC/ 453. (GIMEMA) Leukemia Cooperative
GIMEMAAML-10 trial. Blood. 2003; 162. Shimoni A, Hardan I, Shem-Tov Groups. Autologous or allogeneic
102:1232–1240. N, et al. Allogeneic hematopoietic bone marrow transplantation
155. Harousseau JL, Cahn JY, Pignon B, stem-cell transplantation in AML compared with intensive chemo-
et al, for the Groupe Ouest Est and MDS using myeloablative ver- therapy in acute myelogenous leu-
Leucémies Aiguës Myéloblastiques sus reduced-intensity condition- kemia. N Engl J Med. 1995;332:
(GOELAM). Comparison of au- ing: The role of dose intensity. 217–223.
tologous bone marrow transplan- Leukemia. 2006;20:322–328. 166. Breems DA, Löwenberg B. Acute
tation and intensive chemothera- 163. Estey E, de Lima M, Tibes R, et al. myeloid leukemia and the posi-
py as postremission therapy in Prospective feasibility analysis of tion of autologous stem cell trans-
adult acute myeloid leukemia. reduced-intensity conditioning plantation. Semin Hematol. 2007;
Blood. 1997;90:2978–2986. (RIC) regimens for hematopoietic 44:259–266.
156. Paschka P, Marcucci G, Ruppert
AS, et al, for the Cancer and
Leukemia Group B. Adverse prog-
nostic significance of KIT muta-
tions in adult acute myeloid leuke-
mia with inv(16) and t(8;21): A
Cancer and Leukemia Group B
Study. J Clin Oncol. 2006;24:3904–
3911.
157. Breems DA, Van Putten WL,
Huijgens PC, et al. Prognostic in-
dex for adult patients with acute
myeloid leukemia in first relapse.
J Clin Oncol. 2005;23:1969–1978.
158. Gorin NC. Autologous stem cell
transplantation for adult acute
leukemia. Curr Opin Oncol. 2002;
14:152–159.
159. Sierra J, Storer B, Hansen JA, et al. Address correspondence to: Tadeusz Robak, MD, PhD, Department of
Unrelated donor marrow trans- Hematology, Medical University of Lodz, Copernicus Memorial Hospital,
plantation for acute myeloid leu- 93-510 Lodz, Ul. Ciołkowskiego 2, Poland. E-mail: robaktad@csk.umed.
kemia: An update of the Seattle lodz.pl

2370 Volume 31 Theme Issue

You might also like