Vous êtes sur la page 1sur 10

From www.bloodjournal.org by guest on July 24, 2016. For personal use only.

Review Series

ADVANCES IN ACUTE MYELOID LEUKEMIA

An update of current treatments for adult acute myeloid leukemia


Hervé Dombret1,2 and Claude Gardin2,3
1
Department of Hematology, Hôpital Saint-Louis, Assistance Publique - Hôpitaux de Paris (AP-HP), Paris, France; 2Leukemia Translational Laboratory,
EA3518, Institut Universitaire d’Hématologie, Université Paris Diderot, Paris, France; and 3Department of Hematology, Hôpital Avicenne, AP-HP,
Bobigny, France

Recent advances in acute myeloid leukemia transplantation (HSCT), at least in younger analogs or gemtuzumab ozogamicin, is still
(AML) biology and its genetic landscape patients who can tolerate such intensive under investigation, whereas promising
should ultimately lead to more subset- treatments. Nevertheless, progress has new targeted agents are under clinical de-
specific AML therapies, ideally tailored to been made, notably in terms of standard velopment. In contrast, minimal advances
each patient’s disease. Although a growing drug dose intensification and safer alloge- have been made for patients unable to
number of distinct AML subsets have been neic HSCT procedures, allowing a larger tolerate intensive treatment, mostly rep-
increasingly characterized, patient man- proportion of patients to achieve durable resenting older patients. The availability
agement has remained disappointingly uni- remission. In addition, improved identifica- of hypomethylating agents likely repre-
form. If one excludes acute promyelocytic tion of patients at relatively low risk of re- sents an encouraging first step for this
leukemia, current AML management still lapse should limit their undue exposure to latter population, and it is hoped will allow
relies largely on intensive chemotherapy the risks of HSCT in first remission. The role for more efficient combinations with novel
and allogeneic hematopoietic stem cell of new effective agents, such as purine agents. (Blood. 2016;127(1):53-61)

Introduction
Long-term cure of patients with acute promyelocytic leukemia (APL) induction. However, it is very difficult to compare these studies, because
using retinoic acid and arsenic trioxide (ATO) therapy represents the they differ significantly in several key parameters, notably the number of
only dramatic therapeutic advance over the last 2 decades in acute induction courses, doses in the control arm, and subsequent therapies
myeloid leukemia (AML).1 Although increasingly refined knowledge offered to responders or to patients with persistent marrow blasts after the
of AML biology has led to the development of new targeted agents such first induction course. Differences in study results may thus be caused by
as the mutated FLT3 or IDH inhibitors, current advances in non-APL differences in the design of either experimental or control arms.
patients lack innovation, relying instead on modifications to doses and
schedules of standard cytotoxic drugs or progress in hematopoietic stem Anthracyclines
cell transplantation (HSCT) techniques. In younger patients, complete
remission (CR) rates of $80% may be reached, with 5-year overall Recently reported randomized studies exploring daunorubicin or
survival (OS) ;40%. In older patients, the use of hypomethylating agents idarubicin doses during induction courses are summarized in Table 1.2-8
has improved median and short-term OS but has not translated into Four studies have evaluated higher daunorubicin doses.2,4-6 The
improved cure rates, which remain disappointingly very low. Guidelines Eastern Cooperative Oncology Group (ECOG) trial included patients
for AML management are widely available. This review aims to provide aged #60 years,2 whereas the European trial from a Dutch, Belgian,
a balanced perspective of available data supporting AML treatment used German, and Swiss consortium included patients aged $60 years.4
in routine practice today. We have focused on data from randomized Both studies compared a daily dose of 45 mg/m2 vs a doubled dose of
clinical trials using approved drugs. Early results with new inves- 90 mg/m2 for 3 days as part of 713 induction therapy. The higher
tigational drugs will be discussed in depth in another article of this daunorubicin dose was associated with higher CR rates, without
Review Series. delaying hematologic recovery or affecting the feasibility of planned
postremission therapies. In the younger population ECOG trial, OS was
significantly prolonged in the 90 mg/m2 arm.2 The OS benefit was
observed in all cytogenetic groups and in patients with internal tandem
Front-line induction therapy duplication (ITD) of the FLT3 gene, as well as in those with NPM1 and
DNMT3A gene mutations.3 In the older population trial, the OS benefit
Induction therapy with cytarabine and an anthracycline remains a was restricted to patients between the age of 60 and 65 years and to the
standard of care in AML. The standard combination is the 713, with a few patients with core-binding factor (CBF) AML.4
7-day continuous infusion of cytarabine at the dosage of 100 or 200 mg/m2 These results demonstrate that the 45 mg/m2 daunorubicin daily
per day on days 1 to 7 and daunorubicin at 60 mg/m2 per day on days 1 dose is suboptimal up until the age of 65 years. They do not, however,
to 3. Recent randomized studies have investigated higher doses of establish that the 90 mg/m2 daily dose should be preferred over the
anthracyclines or cytarabine or the addition of a third agent during 60 mg/m2 daily dose endorsed by the international 2010 guidelines.9

Submitted August 1, 2015; accepted October 3, 2015. Prepublished online as © 2016 by The American Society of Hematology
Blood First Edition paper, December 10, 2015; DOI 10.1182/blood-2015-08-
604520.

BLOOD, 7 JANUARY 2016 x VOLUME 127, NUMBER 1 53


From www.bloodjournal.org by guest on July 24, 2016. For personal use only.

54 DOMBRET and GARDIN BLOOD, 7 JANUARY 2016 x VOLUME 127, NUMBER 1

Table 1. Randomized studies of anthracycline dose and type for AML induction therapy
Study Age (y) Patients (N) Experimental arm Control arm Key conclusions

ECOG2,3 17-60 657 D, 90 mg/m2 d1-3 D, 45 mg/m2 d1-3 Higher response rate
A, 100 mg/m2 CIV d1-7 A, 100 mg/m2 CIV d1-7 Similar toxicity
cycle 1* cycle 1* Longer OS
HOVON-SAKK-AMLSG4 .60 813 D, 90 mg/m2 d1-3 D, 45 mg/m2 d1-3 Higher response rate
A, 200 mg/m2 CIV d1-7 A, 200 mg/m2 CIV d1-7 Similar toxicity
cycle 1 cycle 1 Similar OS†
Korean Group5 15-60 383 D, 90 mg/m2 d1-3 D, 45 mg/m2 d1-3 Higher response rate
A, 200 mg/m2 CIV d1-7 A, 200 mg/m2 CIV d1-7 Similar toxicity
cycle 1‡ cycle 1‡ Longer EFS and OS
NCRI AML176 16-72 1206 D, 90 mg/m2 d1/3/5 D, 60 mg/m2 d1/3/5 Similar response rate
A, 100 mg/m2/12 h d1-10 A, 100 mg/m2/12 h d1-10 Higher early death rate with 90 mg/m2
cycle 1§ cycle 1§ Similar EFS and OS
ALFA-98017 50-70 468 I, 12 mg/m2 d1-3 or d1-4 D, 80 mg/m2 d1-3 Higher response rate in one course
A, 200 mg/m2 CIV d1-7 A, 200 mg/m2 CIV d1-7 Similar early death rates
cycle 1 cycle 1 Similar EFS and OS
JALSG AML2018 15-64 1057 D, 50 mg/m2 d1-5 I, 12 mg/m2 d1-3 Similar response rate
A, 100 mg/m2 CIV d1-7 A, 100 mg/m2 CIV d1-7 Higher early death rate with control arm
cycle 1|| cycle 1|| Similar RFS and OS

A, cytarabine; CIV, continuous IV infusion; D, daunorubicin; EFS, event-free survival; I, idarubicin; OS, overall survival.
*A second 713 DA cycle with 45 mg/m2 per day daunorubicin was given to patients with residual marrow blasts on d12/14 of cycle 1.
†A significant EFS and OS benefit was observed in the subgroup of patients aged 60 to 65 years.
‡A second 512 DA cycle with 45 mg/m2 per day daunorubicin was given to patients with persistent leukemia after cycle 1.
§In this study, all patients but poor-risk received a second DA course with 50 mg/m2 per day daunorubicin.
||A second cycle, identical to the first one, was given to patients with persistent leukemia after cycle 1.

The recent UK NCRI AML17 trial addressed this question in an up- for 5 days during the first course and 2000 mg/m2 every 12 hours on
front comparison of the 60 and 90 mg/m2 doses in the first course.6 All days 1, 2, 4, and 6 during the second course.12 Similar rates of CR,
favorable- and intermediate-risk patients went on to receive a second event-free survival (EFS), and OS were observed in the 2 arms, with
course with a 50 mg/m2 dose. In an interim analysis, a significant in- more toxicity associated with the HiDAC arm.
crease in day 60 mortality was observed in the 90 mg/m2 arm, leading to In the second study, conducted by the EORTC and GIMEMA Leu-
premature trial termination. No OS benefit was observed in this kemia Groups, patients were randomized to receive either standard doses
analysis, although evidence of a survival benefit in subgroups may at 100 mg/m2 per day cytarabine for 10 days or HiDAC at 3000 mg/m2
require longer follow-up. every 12 hours on days 1, 3, 5, and 7 during the first course.13 A higher
The Acute Leukemia French Association (ALFA) 9801 study com- CR rate was observed in the HiDAC arm, with a trend for a longer OS
paring daunorubicin vs idarubicin, failed to demonstrate superiority of that reached statistical significance in the subset of patients aged
80 mg/m2 daunorubicin over a standard 12 mg/m2 idarubicin dose.7 #45 years. Finally, in the randomized German Intergroup study that
A similar outcome was reported in the Japan Acute Leukemia Study included a double induction as a “common” arm or an IDAC or
Group (JALSG) AML201 study, which compared 2 courses of 50 mg/m2 HiDAC sequence during induction in at least 3 study arms, no
daunorubicin for 5 days with 12 mg/m2 idarubicin for 3 days.8 difference in OS was observed.14 It thus remains unclear whether
Antileukemic activity of 60 and 90 mg/m2 daunorubicin or 12 mg/m2 increasing the cytarabine dose during induction may benefit patients
idarubicin daily doses thus appears to be similar, as are the toxicity planned to receive IDAC or HiDAC during postremission therapy.
profiles, at least in cases when a second anthracycline-containing
cycle is not given systematically to responders. “Dose-dense” regimens

Another approach to increasing induction intensity relies on systematic


Cytarabine administration of a second sequence of chemotherapy starting earlier
than normal after the completion of the first sequence (generally be-
Randomized studies exploring cytarabine dose and schedule during
tween day 7 and day 14). This timed-sequential concept was initially
induction are summarized in Table 2. Historical studies from the
developed by the Johns Hopkins group in Baltimore,15 then prospec-
Southwest Oncology Group (SWOG) and the Australian Leukemia
tively evaluated by the ALFA group without incorporating HiDAC.16
Study Group (ALSG) failed to demonstrate clinically relevant gains in
After investigating double induction containing 1 or 2 HiDAC se-
efficacy with higher cytarabine doses with alternative administration
quences (TAD-HAM or HAM-HAM),17 the German AML Coop-
regimens, whereas both reported increased toxicity.10,11 Two recent
erative Group recently conducted a phase 2 trial investigating a
studies further addressed this question, one in the context of idarubicin
sequential S-HAM.18 However, none of these studies provide
or amsacrine and the other using lower etoposide doses and alternative
evidence that a dose-dense regimen is superior to the standard 713
administration schedules.
regimen, especially when high doses of daunorubicin are used.
In the study conducted by the Dutch-Belgian Cooperative Trial
Group for Hemato-Oncology (HOVON) and the Swiss Group for Addition of a third drug
Clinical Cancer Research (SAKK), patients were randomized between
an IDAC arm, comprising 200 mg/m2 per day cytarabine during the first Gemtuzumab ozogamicin (GO). Randomized studies exploring
course and 1000 mg/m2 every 12 hours for 6 days during the second the addition of GO to intensive chemotherapy (ICT) are summarized
course, or a high-dose (HiDAC) arm with 1000 mg/m2 every 12 hours in Table 3.19-26 Six studies evaluating the addition of 3 or 6 mg/m2
From www.bloodjournal.org by guest on July 24, 2016. For personal use only.

BLOOD, 7 JANUARY 2016 x VOLUME 127, NUMBER 1 TREATMENT OF ADULT AML 55

Table 2. Randomized studies of cytarabine dose for AML induction therapy


Study Age (y) Patients (N) Experimental arm Control arm Conclusions
SWOG10 15-64 723 A, 2000 mg/m2/12 h d1-6* A, 200 mg/m2 CIV d1-7 Similar response rate
D, 45 mg/m2 d7-9 D, 45 mg/m2 d5-7 Higher early death rate
cycle 1† cycle 1† Longer RFS in patients aged ,50 y
Similar OS
ALSG11 15-60 301 A, 3000 mg/m2/12 h d1/3/5/7 A, 100 mg/m2 CIV d1-7 Higher response rate in one course
D, 50 mg/m2 d5-7 D, 50 mg/m2 d5-7 Non significantly higher early death rate
E, 75 mg/m2 d1-7 E, 75 mg/m2 d1-7 Longer RFS
cycle 1‡ cycle 1‡ Similar OS
HOVON-SAKK12 18-60 860 A, 1000 mg/m2/12 h d1-5 I, 12 mg/m2 A, 200 mg/m2 CIV d1-7 I, 12 mg/m2 Similar response rate
d5-7 cycle 1 d5-7 cycle 1
A, 2000 mg/m2/12 h d1/2/4/6 Am, A, 1000 mg/m2/12 h d1-6 Am, Similar EFS and OS
120 mg/m2 d3/5/7 cycle 2§ 120 mg/m2 d3/5/7 cycle 2§
EORTC-GIMEMA 15-60 1942 A, 3000 mg/m2/12 h d1/3/5/7 A, 100 mg/m2 CIV d1-10 Higher response rate
AML1213 D, 50 mg/m2 d1/3/5 D, 50 mg/m2 d1/3/5 Similar early death rate
E, 50 mg/m2 d1-5 E, 50 mg/m2 d1-5 Longer RFS and OS in patients aged #45 y
cycle 1|| cycle 1||

A, cytarabine; Am, amsacrine; CIV, continuous IV infusion; D, daunorubicin; E, etoposide; I, idarubicin; RFS, relapse-free survival; OS, overall survival.
*During the first 2 years of the study, cytarabine was given at 3000 mg/m2 per bolus to patients aged ,50 years, then reduced to 2000 mg/m2 per bolus because of
excessive neurologic toxicity.
†In both arms a second cycle, identical to the first one, was given to patients with persistent leukemia after cycle 1; all CR patients from the control arm were then
randomized to either standard-dose cytarabine or HiDAC during consolidation, whereas all CR patients from the experimental arm received HiDAC during consolidation.
‡A second, then a third, cycle, identical to the first one, was given to patients with persistent leukemia after cycle 1 or 1-2, respectively.
§After cycle 2, CR patients received either allogeneic or autologous HSCT or a third chemotherapy cycle for consolidation.
||In both arms, a second cycle, identical to the first one, was given to patients with persistent leukemia after cycle 1, then all CR patients received one IDAC-containing
consolidation course followed by allogeneic or autologous HSCT.

GO during induction have been performed.19-24 The first of them, newly diagnosed patients.29 Front-line results from 2 other trials are
SWOG S0106, was negative and closed prematurely because of a imminent, including the combination of clofarabine with 713,35 and
higher early mortality rate despite a reduced 45 mg/m2 per day clofarabine compared with 713 in an ECOG trial. The British AML15
daunorubicin dose in the GO arm, leading to GO withdrawal by the trial in a younger population demonstrated that the fludarabine,
Food and Drug Administration in the United States.19 Conversely, 4 cytarabine, granulocyte colony-stimulating factor (G-CSF), and
studies reported significant improvements when GO was combined idarubicin regimen (FLAG-Ida) yielded more frequent remissions with
with induction or induction-and-consolidation chemotherapy.20-23 This one course and also reduced the risk of relapse.30
finding was confirmed in a recent meta-analysis.27 Nonetheless, the Sorafenib. Sorafenib is a multikinase inhibitor with in vitro
addition of GO was associated with more frequent severe liver toxicity activity in FLT3-ITD AML. In an AML trial from the German Study
and persistent thrombocytopenia. The benefit/risk ratio appeared to be at Alliance Leukemia, combination of sorafenib with standard induction
least as good with 3 mg/m2 per dosing,24 a dose that can be administered and IDAC consolidation was evaluated in older patients. Treatment in
to older patients,21 and ultimately be repeated as successfully developed the sorafenib arm did not result in significant improvement in EFS or
by the ALFA group.22 In contrast, increased toxicity and an absence of OS.36 This was also true for subgroup analyses, including the subgroup
clinical benefit were observed in the EORTC/GIMEMA study, in which positive for FLT3-ITD. Results of induction therapy were worse in the
single-agent GO administration preceded induction chemotherapy.25 sorafenib arm, with higher early mortality and a lower CR rate. The
Finally, the 2 studies that evaluated GO maintenance therapy were same group has reported preliminary results of a similar trial conducted
negative.19,26 Clinical response to GO is likely to be influenced by in younger patients in which sorafenib was added to 713 induction and
CD33 expression level, clonal hierarchy of the leukemic population, HiDAC consolidations.37 No difference in the CR rate was observed,
and drug efflux intensity, all factors related to AML genetics. It has been whereas EFS and relapse-free survival (RFS) were significantly im-
widely demonstrated that GO benefits patients of favorable and proved in the sorafenib arm. In the subgroup of FLT3-ITD–positive
intermediate risk, including those with FLT3-ITD, although not those patients, no difference in EFS was observed; however, there was a trend
with an adverse karyotype.27 Together, these data suggest that the for prolonged RFS and OS favoring sorafenib.
license status of GO might need to be reviewed, at least for some patient
subsets. Early outcomes with the SGN-CD33A immunoconjugate
in AML are discussed elsewhere in this Review Series.
Purine analogs. Randomized studies exploring the addition of Postremission therapy
purine analogs to ICT are summarized in Table 4.28-32 In the Polish
HiDAC consolidation
Acute Leukemia Group (PALG) study, the addition of cladribine was
associated with prolonged OS,28 although it should be noted that For younger patients not undergoing HSCT, administration of several
outcomes in the control arm appeared to be suboptimal. Interestingly, HiDAC consolidation courses using cytarabine twice daily at a 3 g/m2
addition of cladribine appears to have particularly benefited high-risk dose on days 1, 3, and 5 has been a widely used option since 1994.38
patients, notably those aged $50 years or those with unfavorable Even if the optimal cytarabine dose, schedule of administration, and
cytogenetics. Clofarabine also demonstrated significant antileukemic number of cycles need yet to be defined,39,40 the use of bolus admin-
activity, both as a single agent and in combination with cytarabine in istration of HiDAC or IDAC during consolidation should be recom-
various patient populations.33,34 However, it failed to show significant mended. Interestingly, HiDAC and IDAC regimens are well suited to
benefit when combined with daunorubicin in the British AML16 trial in evaluate the effects of added targeted or nontargeted drugs during
From www.bloodjournal.org by guest on July 24, 2016. For personal use only.

56 DOMBRET and GARDIN BLOOD, 7 JANUARY 2016 x VOLUME 127, NUMBER 1

Table 3. Randomized trials of gemtuzumab ozogamicin (GO) associated with intensive chemotherapy
Treatment phase
Study (age range [y]) Patients (N) GO dose and schedule Conclusions
SWOG S010619 Induction (18-60) 637 GO, 6 mg/m2 Similar response rate
d4 cycle 1* Higher early death rate
Similar RFS
Similar OS
NCRI AML1520 Induction (18-60) 1113 GO, 3 mg/m2 Similar response rate
d1 cycle 1 Similar RFS and OS
d1 cycle 3† Longer OS in favorable AML
NCRI AML1621 Induction (.60) 1115 GO, 3 mg/m2 Similar response rate
d1 cycle 1 Longer RFS, OS from CR, and OS
ALFA-070122 Induction 278 GO, 3 mg/m2 Similar response rate
Consolidation d1/4/7 cycle 1 Longer EFS, RFS, and OS
(50-70) d1 conso 1
d1 conso 2
GOELAMS 200623 Induction (18-60) 254 GO, 6 mg/m2 Similar response rate
d4 cycle 1 Similar EFS and OS
d4 conso 1 Longer EFS in non–allo-HSCT patients
NCRI AML1724 Induction (0-81) 788 GO, 3 vs 6 mg/m2 Similar responses rate
d1 cycle 1 Lower early death rate with 3 mg/m2
Similar RFS and OS
EORTC-GIMEMA Prior to induction (61-75) 472 GO, 6 mg/m2 Similar response rate
AML-1725 d1/15 before cycle 1 Higher early death rate
Similar RFS
Similar EFS and OS
HOVON-SAKK-AMLSG26 Maintenance (.60) 232 GO, 6 mg/m2 Similar relapse incidence
3 monthly cycles Similar RFS and OS
SWOG S010619 Maintenance (18-60) 174 GO, 5 mg/m2 Similar RFS
3 monthly cycles

EFS, event-free survival; OS, overall survival; RFS, relapse-free survival.


*Daunorubicin dose was reduced from 60 mg/m2 per day in the control arm to 45 mg/m2 per day in the GO arm.
†Additional randomization for GO during cycle 3, irrespective of cycle 1 GO treatment.

consolidation. For instance, a prolonged RFS was observed in the ran- given patient. Transplantation offers the best means of preventing AML
domized ALFA-0702/CLARA study in intermediate- and unfavorable- recurrence, but remains associated with higher treatment-related mor-
risk patients with clofarabine/IDAC compared with HiDAC consolidation bidity and mortality (TRM), especially in older patients. In patients with
cycles.31 favorable-risk AML, the relapse risk may be low enough and the
salvage rate high enough to postpone HSCT to second remission. This
Allogeneic HSCT
strategy has been validated in several donor vs no-donor studies.41,42 In
these studies, favorable patients (ie, those with CBF-AML) from the no-
One of the most important treatment decisions in AML is to estimate the donor group did as well as those from the donor group, whereas all other
benefit/risk associated with allogeneic HSCT in first remission for a patients appeared to benefit from undergoing allograft. One should

Table 4. Randomized trials of purine analogs associated to intensive chemotherapy


Treatment phase Patients
Study Analog (age range [y]) (N) Treatment modalities Conclusions

PALG28 Cladribine or fludarabine Induction (16-60) 652 DAC vs DAF vs DA DAC: Higher response rate, similar RFS, longer OS*
cycle 1 DAF: Similar response rate, similar RFS and OS†
NCRI AML1629 Clofarabine Induction (.60) 806 DA vs DClo Similar response rate‡
cycle 1-2 Similar RFS and OS
NCRI AML1530 Fludarabine Induction (0-73) 3106 DA vs ADE vs FLAG-Ida ADE: similar response rate, RFS and OS
cycle 1-2 FLAG-Ida: Similar response rate§, higher rate of death
in CR, longer RFS, similar OS
ALFA-070231 Clofarabine Postremission (18-60) 221 CLARA vs HiDAC Longer RFS in non allo-HSCT patients
consolidation cycle 1-3 Similar OS
CLASSIC I32 Clofarabine First salvage (.55) 320 CLARA vs IDAC Higher response rate
up to 3 cycles Higher early death rate
Longer EFS
Similar OS

ADE, DA 1 etoposide; CLARA, clofarabine 1 IDAC; DA, daunorubicin 1 cytarabine; DAC, DA 1 cladribine; DAF, DA 1 fludarabine; DClo, daunorubicin 1 clofarabine;
FLAG-Ida, fludarabine 1 IDAC 1 G-CSF 1 idarubicin; HiDAC, high-dose cytarabine; HSCT, hematopoietic stem cell transplantation; IDAC, intermediate-dose cytarabine.
*Longer OS in patients .50 years of age, those with initial leukocyte count .50 3 109/L, and those with unfavorable cytogenetics.
†Longer OS in patients with unfavorable cytogenetics.
‡Lower CR in one course after DClo.
§Higher CR in one course after FLAG-Ida.
From www.bloodjournal.org by guest on July 24, 2016. For personal use only.

BLOOD, 7 JANUARY 2016 x VOLUME 127, NUMBER 1 TREATMENT OF ADULT AML 57

keep in mind that patients in these studies mostly underwent sibling Frequent KIT mutations or overexpression of the KIT receptor were
donor myeloablative conditioning (MAC) transplantation and as such, described some time ago in CBF-AML. Recent studies have evaluated
the benefit associated with HSCT was only demonstrated for patients the potential benefit of dasatinib, a potent KIT inhibitor, in these
,40 years of age. Based on another donor vs no-donor analysis, patients. In a French study, patients with persistent MRD or molecular
patients with cytogenetically normal (CN) AML and a favorable geno- relapse after intensive consolidations were eligible to receive 12 months
type (defined as mutated CEBPA or NPM1 without FLT3-ITD) were of maintenance with dasatinib if they were not a candidate for HSCT.60
recently categorized in the favorable subgroup.43 Because the outcome No significant impact of dasatinib on time-to-relapse was seen. Two
after allogeneic HSCT from fully matched unrelated donors appears phase 1-2 trials combined dasatinib with up-front ICT led to a currently
to be similar compared with allogeneic HSCT from matched related ongoing phase 3 trial.61,62
donors, all younger patients with intermediate- and unfavorable-risk
AML are generally considered candidates for allogeneic HSCT from AML with FLT3 gene mutation
sibling or fully-matched unrelated donors in cases of first CR.7
The FLT3-ITD mutation is one of the most frequent bad-prognosis
This HSCT benefit/risk assessment, based on the European
mutations observed in AML, at least in younger or relapsed patients,
LeukemiaNet (ELN) genetic classification only,7 needs however to be
and has led to the evaluation of multikinase or more specific kinase
reconsidered in the near future. As discussed in another article of this
inhibitors in this AML subset. Lestaurtinib associated with chemother-
Review Series, the incidence of TRM has been substantially reduced
apy failed to improve outcomes in relapsed/refractory (R/R) FLT3-ITD
over the last few decades,44 particularly in older patients, by using
AML patients or during front-line consolidation in the MRC AML15
reduced-intensity conditioning (RIC). Alternative stem cell sources are
trial.63,64 Positive results from a large international phase 3 trial in newly
more widely used and are safer, as illustrated by post-transplant ad-
diagnosed FLT3-ITD–positive patients in which midostaurin
ministration of cyclophosphamide in haplo-identical HSCT.45,46 Of
was compared to placebo in association with 7+3 should be available
particular interest are the recent results of studies evaluating RIC
soon. As mentioned before, to date there is no clear evidence of
transplantation in middle-aged patients.47-50 Using a time-dependent
increased clinically significant activity when sorafenib is added to ICT
Mantel-Byar comparison in patients .45 years of age, the MRC
in FLT3-ITD–positive patients. A phase 2 study of sorafenib combined
AML15 trial showed that, compared with chemotherapy, RIC trans-
with ICT in older patients, restricted to those diagnosed with FLT3-
plantation was associated with longer survival.47 Two recent studies
mutated AML, is expected to shed light on this issue (Clinical-
have even suggested that RIC might be preferred to MAC transplan-
Trials.gov #NCT01253070). Conversely, sorafenib delivered after
tation in this age range, given the lower TRM as well as better survival
HSCT seems to be particularly effective in FLT3-ITD–positive
in some risk subgroups.49,50
patients.65 Selection of FLT3-resistant mutants has been a general
Reliance on genetic profiles as the main treatment-stratifying tool is
phenomenon observed in most early trials with first- or second-
being increasingly challenged because of multiple recently described
generation inhibitors.66 An overview of novel promising FLT3
genetic mutations and the potential impact of mutated allele frequencies.
inhibitors is presented elsewhere in this Review Series.
Not all patients with ELN favorable-risk AML have a favorable out-
come; for instance, should we take into account the presence of ad- AML with NPM1 gene mutation
ditional KIT or FLT3 poor-risk mutations in a patient with CBF-AML,
or of additional ASXL1, IDH1 or DNMT3A mutations in a patient with Approximately 30% of CN-AML patients have an NPM1 mutation
NPM1-mutated CN-AML and no FLT3-ITD? Likewise, should HSCT and, in contrast to the frequently associated FLT3-ITD, the NPM1
be considered the best postremission option in ELN intermediate-risk mutation seems to be an early event in most of these cases. Several trials
patients with NPM1-mutated FLT3-ITD–positive CN-AML but a low have reported the effects of adding all-trans retinoic acid (ATRA) to
FLT3-ITD allelic ratio?51-54 chemotherapy in non-APL AML, with conflicting results. In the large
Assessment of minimal residual disease (MRD) using molecular MRC trial, no significant effect of the addition of ATRA was
markers or leukemia-associated aberrant immunophenotypes is therefore observed.67 Alternatively, 2 studies from the German AML Study
proposed as a more straightforward, easy-to-use prognostic decision Group reported a benefit of the addition of ATRA restricted to NPM1-
tool, including deciding which patients should be advised to undergo mutated AML patients.68,69 An explanation for these contradictory
allogeneic HSCT.55,56 As in acute lymphoblastic leukemia, MRD has results is yet to be defined. More recently, preclinical studies of arsenic
been suggested as ultimately superseding other prognostic factors for trioxide (ATO) in NPM1-mutated AML from 2 groups strongly
HSCT decision-making, including genetic factors,57 as is already the case suggested that ATO may have a beneficial effect in this subset.70,71
for CBF-AML.58 Nevertheless, whether allogeneic HSCT may be the Because extensive clinical experience with ATO has been gained in the
option of choice in poor early MRD responders remains an open issue.59 therapy of APL, clinical studies are planned to confirm whether there is
a beneficial role of ATO in the treatment of NPM1-mutated patients.

AML with mutated IDH

Targeted treatments for specific Isocitrate dehydrogenase 1 and 2 gene mutations are present in 10% to
AML subgroups 15% of AML patients. As detailed in another article of this Review
Series, early phase 1/2 results of specific inhibition of IDH2- and IDH1-
Core binding factor AML mutated enzymes are impressive. A phase 3 study will soon be initiated
in relapsing IDH2-mutated patients.
A subgroup analysis of the MRC AML15 younger population
AML trial, along with a subsequent meta-analysis,20-24,27 strongly AML with adverse cytogenetic features
suggests that the combination of GO with ICT is associated with a
significant OS benefit in CBF-AML, renewing interest in this drug, In this hard-to-treat AML population, improvement of early response
which is currently unavailable outside the context of clinical studies, for rate remains an important clinical end point, especially when HSCT can
this AML subset. be envisioned in a timely manner. However, results of trials with novel
From www.bloodjournal.org by guest on July 24, 2016. For personal use only.

58 DOMBRET and GARDIN BLOOD, 7 JANUARY 2016 x VOLUME 127, NUMBER 1

drugs are disappointing. In the PALG cladribine study,28 the subgroup


of patients with unfavorable cytogenetics appeared, nevertheless, Treatment of relapsed/refractory AML
to have a greater benefit. Similarly, a combination of cladribine,
cytarabine, priming with G-CSF, and mitoxantrone (CLAG-M) was Treatment of relapsed AML remains poorly defined. Simple clinical
reported to be of benefit compared with 713 in a large, retrospective, and disease parameters such as age, duration of first remission,
single-center study that primarily included high-risk AML patients.72 cytogenetics, and prior HSCT remain the most useful parameters
Among other nontargeted agents, amonafide combined with cytarabine to evaluate treatment effects at relapse.89 In most AML subsets
failed to yield a benefit over 713 in a secondary AML study.73 Early (other than APL) the main clinical objective of salvage therapy is
results of new drugs, including the CPX-351 liposomal combination, to “bridge” patients to HSCT, either with targeted therapies such as
are discussed elsewhere in this Review Series. FLT3 inhibitors or with ICT, at least in patients fit enough to
tolerate it. A very large cohort of relapsed AML patients subjected
to salvage therapy after being treated frontline in successive MRC
AML trials has recently been described.90 Analysis of the long-
Treatment of older AML patients term outcomes clearly established that allogeneic transplan-
tation when a second response was obtained, strongly benefited
Outcome in older patients with AML remains dismal, with lower CR intermediate- and high-risk AML subsets based on initial disease
rates and very few long-term survivors compared with younger characteristics.
patients. Treatment of older AML patients is thus an active field of Optimal salvage drug combinations, drug doses, and administration
antileukemic drug investigation. Even when short-term benefits have schedules remain open issues. In particular, the benefits of the addition
been demonstrated, very few older patients survive beyond 2 years of of an anthracycline, or any other drug, to cytarabine, and what
follow-up, raising the question of the appropriateness of OS as an cytarabine dose should be recommended are unclear at best. Recently,
efficacy end point for drug development in this patient population. Age 2 large company-sponsored studies evaluated the combination of
itself does not, however, define a homogeneous patient population. In clofarabine or vosaroxin to IDAC, with OS as the end point in R/R
older patients, prognosis is governed by patient-related and AML- AML patients. In the CLASSIC I trial, addition of clofarabine increased
related factors that are only partly age-related. General health status the response rate in patients .55 years of age, albeit at the expense of
and the presence of organ dysfunctions or comorbidities affect ICT increased toxicity and early mortality, but failed to improve OS despite
tolerance. As observed in younger patients, cytogenetics can also affect a higher number of patients bridged to HSCT.32 In the preliminary
efficacy. One predominant bad-risk characteristic of older AML is the results of the largest trial ever performed in R/R patients, the VALOR
increased frequency of prior myelodysplastic syndromes, although trial, combination of vosaroxin to IDAC similarly increased the re-
clonal hematopoiesis characterized by the presence of myelodysplastic sponse rate across all ages in both relapsed and refractory patients.
syndromes–related gene mutations has also been associated with aging Nonetheless, despite the absence of a significant increase in early
in healthy individuals.74,75 mortality and a large proportion of younger patients bridged to HSCT,
The most important clinical decision remains to estimate the benefit/ this did not translate into general improvement in OS. A significant,
risk associated with ICT in the individual older patient. As for the although modest, survival improvement was only observed in older
decision to recommend HSCT in first CR, there is no unique decision- patients $60 years of age.91
guiding score. Older patients with favorable-risk AML according to
ELN classification are likely to benefit from a standard treatment.76,77 In
those unlikely to benefit from ICT, low-dose cytarabine (LDAC) has
been considered as a possible standard, based on the nonintensive Conclusion
AML14 MRC trial results, despite the fact that patients with adverse
cytogenetics did not draw any benefit from LDAC therapy.78 In 2 Despite some advances in the treatment of adult AML patients, many
large international trials, the hypomethylating agents decitabine and issues remain to be addressed, as reflected by current recommenda-
azacitidine yielded better mid-term results, with longer median and tions.7,92 Given the poor long-term outcome for most adult AML
higher 1-year survival than those observed in LDAC arms, even if they patients, although many novel promising drugs or therapeutic ap-
did not result in a higher proportion of long-term survivors.79,80 Of proaches are currently under development for this population, their
interest, azacitidine appeared to offer particular benefit to patients with inclusion into prospective clinical trials should be strongly encouraged.
adverse cytogenetics and/or those with myelodysplasia-related In addition, standardization of trial procedures and control arms would
changes.80 greatly assist comparison of trial results and strengthen future treatment
Current trials in older AML patients often address the effects of the recommendations.
addition of a new agent to LDAC, azacitidine, or decitabine. To date,
negative results with tipifarnib, ATO, GO, and vosaroxin in combi-
nation with LDAC have been reported by the British group.81-84 Single-
agent sapacitabine or clofarabine also failed to improve outcomes over Authorship
LDAC.85,86 Addition of quizartinib, ganetespib, tosedostat, or selinexor
to LDAC is currently under investigation. Interesting phase 2 results Contribution: H.D. and C.G. wrote this review article jointly.
have been observed when combining the polo-like kinase inhibitor Conflict-of-interest disclosure: H.D. is on the Advisory Board of
volasertib with LDAC,87 or vosaroxin with decitabine in a single-center Celgene, Pfizer, Sunesis, and Agios Pharmaceuticals. C.G. is on the
evaluation.88 Too frequently, the evaluation of the efficacy of the com- Advisory Board of Celgene.
binations is hindered by their poor tolerance in older patients deemed Correspondence: Hervé Dombret, Hopital Saint-Louis, Assis-
unfit for standard ICT, raising the issue of their evaluation in fitter tance Publique - Hopitaux de Paris (AP-HP), Paris, France; e-mail:
patients, potentially against ICT. herve.dombret@aphp.fr.
From www.bloodjournal.org by guest on July 24, 2016. For personal use only.

BLOOD, 7 JANUARY 2016 x VOLUME 127, NUMBER 1 TREATMENT OF ADULT AML 59

References
1. Lo-Coco F, Avvisati G, Vignetti M, et al. Retinoic 16. Castaigne S, Chevret S, Archimbaud E, et al. 29. Russell N, Burnett A, Kjeldsen L, et al.
acid and arsenic trioxide for acute promyelocytic Randomized comparison of double induction and A comparison of daunorubicin/ara-c versus
leukemia. N Engl J Med. 2013;369(2):111-121. timed-sequential induction to a « 317 » induction daunorubicin/clofarabine as induction treatment in
in adults with AML: long-term analysis of the older patients with AML and high-risk MDS: Long
2. Fernandez HF, Sun Z, Yao X, et al. Anthracycline
Acute Leukemia French Association (ALFA) 9000 term results of the UK NCRI AML16 trial in 806
dose intensification in acute myeloid leukemia.
study. Blood. 2004;104(8):2467-2474. patients [abstract]. Haematologica. 2015;100(s1).
N Engl J Med. 2009;361(13):1249-1259.
17. Büchner T, Berdel WE, Schooch C, et al. Double Abstract 514.
3. Luskin MR, Lee J-W, Fernandez HF, et al. High induction containing either two courses or one 30. Burnett AK, Russell NH, Hills RK, et al.
dose daunorubicin improves survival in AML up course of high-dose cytarabine plus mitoxantrone Optimization of chemotherapy for younger
to age 60, across all cytogenetic risk groups and post-remission therapy by either autologous patients with acute myeloid leukemia: results of
including patients with unfavorable cytogenetic stem-cell transplantation or by prolonged the medical research council AML15 trial. J Clin
risk and FLT3-ITD mutant AML: updated analysis maintenance for acute myeloid leukemia. J Clin Oncol. 2013;31(27):3360-3368.
from Eastern Cooperative Oncology Trial E1900 Oncol. 2006;24(16):2480-2489.
[abstract]. Blood. 2014;124(21). Abstract 373. 31. Thomas X, de Botton S, Chevret S, et al.
18. Braess J, Spiekermann K, Staib P, et al. Dose- Clofarabine-based consolidation improves
4. Löwenberg B, Ossenkoppele GJ, van Putten W, dense induction with sequential high-dose relapse-free survival of younger adults with
et al. High-dose daunorubicin in older patients cytarabine and mitoxantrone (S-HAM) and non-favorable acute myeloid leukemia (AML) in
with acute myeloid leukemia. N Engl J Med. 2009; pelfilgrastim results in a high efficacy and a short first remission: results of the randomized
361(13):1235-1248. duration of critical neutropenia in de novo acute ALFA-0702/CLARA study [abstract]. Blood.
5. Lee JH, Joo YD, Kim H, et al. A randomized myeloid leukemia: a pilot study of the AMLCG. 2015;126(23). Abstract 218.
trial comparing standard versus high-dose Blood. 2009;113(17):3903-3910.
32. Faderl S, Wetzler M, Rizzieri D, et al. Clofarabine
daunorubicin induction in patients with acute 19. Petersdorf SH, Kopecky KJ, Slovak M, et al. plus cytarabine compared with cytarabine alone
myeloid leukemia. Blood. 2011;118(14): A Phase 3 study of gemtuzumab ozogamicin in older patients with relapsed or refractory acute
3832-3841. during induction and postconsolidation therapy myelogenous leukemia: results from the
in younger patients with acute myeloid leukemia. CLASSIC I Trial. J Clin Oncol. 2012;30(20):
6. Burnett AK, Russell NH, Hills RK, et al.
Blood. 2013;121(24):4854-4860. 2492-2499.
A randomized comparison of daunorubicin
90 mg/m2 vs 60 mg/m2 in AML induction: 20. Burnett AK, Hills RK, Milligan D, et al. 33. Faderl S, Verstovsek S, Cortes J, et al.
results from the UK NCRI AML17 trial in 1206 Identification of patients with acute myeloid Clofarabine and cytarabine combination as
patients. Blood. 2015;125(25):3878-3885. leukemia who benefit from the addition of induction therapy for acute myeloid leukemia
gemtuzumab ozogamicin: results of the MRC (AML) in patients 50 years of age or older. Blood.
7. Pautas C, Merabet F, Thomas X, et al. AML15 trial. J Clin Oncol. 2011;29(4):369-377. 2006;108(1):45-51.
Randomized study of intensified anthracycline
doses for induction and recombinant interleukin-2 21. Burnett AK, Russell NH, Hills RK, et al. Addition 34. Kantarjian HM, Erba HP, Claxton D, et al. Phase II
for maintenance in patients with acute myeloid of gemtuzumab ozogamicin to induction study of clofarabine monotherapy in previously
leukemia aged 50 to 70 years: Results of the chemotherapy improves survival in older patients untreated older adults with acute myeloid
ALFA-9801 Study. J Clin Oncol. 2010;28(5): with acute myeloid leukemia. J Clin Oncol. 2012; leukemia and unfavorable prognostic factors.
808-814. 30(32):3924-3931. J Clin Oncol. 2010;28(4):549-555.
22. Castaigne S, Pautas C, Terré C, et al. Effect of 35. Willemze R, Suciu S, Muus P, et al. Clofarabine in
8. Ohtake S, Miyawaki S, Fujita H, et al.
gemtuzumab ozogamicin on survival of adult combination with a standard remission induction
Randomized trial of induction therapy comparing
patients with de-novo acute myeloid leukaemia regimen (cytosine arabinoside and idarubicin) in
standard-dose idarubicin with high-dose
(ALFA-0701): a randomised, open-label, phase 3 patients with previously untreated intermediate
daunorubicin in adult patients with previously
study. Lancet. 2012;379(9825):1508-1516. and bad-risk acute myelogenous leukemia (AML)
untreated acute myeloid leukemia: the JALSG
AML201 Study. Blood. 2011;117(8):2358-2365. 23. Delaunay J, Recher C, Pigneux A, et al. Addition or high-risk myelodysplastic syndrome (HR-MDS):
of gemtuzumab ozogamicin to chemotherapy phase I results of an ongoing phase I/II study of
9. Döhner H, Estey EH, Amadori S, et al. Diagnosis improves event-free survival but not overall the leukemia groups of EORTC and GIMEMA
and management of acute myeloid leukemia in survival of AML patients with intermediate (EORTC GIMEMA 06061/AML-14A trial). Ann
adults: recommendations from an international cytogenetics not eligible for allogeneic Hematol. 2014;93(6):965-975.
expert panel, on behalf of the European transplantation: results of the GOELAMS AML
LeukemiaNet. Blood. 2010;115(3):453-474. 36. Serve H, Krug U, Wagner R, et al. Sorafenib in
2006 IR study [abstract]. Blood. 2011;118(21). combination with intensive chemotherapy in
10. Weick JL, Kopecky KJ, Appelbaum FR, et al. Abstract 79. elderly patients with acute myeloid leukemia:
A randomized investigation of high-dose versus 24. Burnett AK, Russell N, Hills RK, et al. A results from a randomized, placebo-controlled
standard-dose cytosine arabinoside with comparison of single-dose gemtuzumab trial. J Clin Oncol. 2013;31(25):3110-3118.
daunorubicin in patients with previously untreated ozogamicin 3 mg/m2 and 6 mg/m2 combined with 37. Röllig C, Müller-Tidow C, Hüttmann A, et al.
acute myeloid leukemia: a Southwest Oncology induction chemotherapy in younger patients Sorafenib versus placebo in addition to standard
Group study. Blood. 1996;88(8):2841-2851. with AML: data from the UK NCRI AML17 trial therapy in younger patients with newly diagnosed
11. Bishop JF, Matthews JP, Young GA, et al. [abstract]. Blood. 2014;124(21). Abstract 2308. acute myeloid leukemia: results from 267 patients
Randomized study of high-dose cytarabine in 25. Amadori S, Suciu S, Stasi R, et al. Sequential treated in the randomized placebo-controlled
induction in acute myeloid leukemia. Blood. 1996; combination of gemtuzumab ozogamicin and SAL-SORAML trial [abstract]. Blood. 2014;
87(5):1710-1717. standard chemotherapy in older patients with 124(21). Abstract 6.
newly diagnosed acute myeloid leukemia: results
12. Löwenberg B, Pabst T, Vellenga E, et al. 38. Mayer RJ, Davis RB, Schiffer CA, et al. Intensive
of a randomized phase III trial by the EORTC and
Cytarabine dose for acute myeloid leukemia. postremission chemotherapy in adults with acute
GIMEMA consortium (AML-17). J Clin Oncol.
N Engl J Med. 2011;364(11):1027-1036. myeloid leukemia. N Engl J Med. 1994;331(14):
2013;31(35):4424-4430.
896-903.
13. Willemze R, Suciu S, Meloni G, et al. High-dose 26. Löwenberg B, Beck J, Graux C, et al.
cytarabine in induction treatment improves the 39. Löwenberg B. Sense and nonsense of high-dose
Gemtuzumab ozogamicin as post-remission
outcome of adult patients younger than age 46 cytarabine for acute myeloid leukemia. Blood.
treatment in AML at 60 years of age or more:
years with acute myeloid leukemia: results of the 2013;121(1):26-28.
results of a multicenter phase 3 study. Blood.
EORTC-GIMEMA AML-12 trial. J Clin Oncol. 2010;115(13):2586-2591. 40. Schlenk RF. Post-remission therapy for acute
2014;32(3):219-228. myeloid leukemia. Haematologica. 2014;99(11):
27. Hills RK, Castaigne S, Appelbaum FR, et al.
14. Büchner T, Schlenk RF, Schaich M, et al. Acute 1663-1670.
Addition of gemtuzumab ozogamicin to induction
myeloid leukemia (AML): different treatment chemotherapy in adult patients with acute myeloid 41. Cornelissen JJ, van Putten WL, Verdonck LF,
strategies versus a common standard arm: leukaemia: a meta-analysis of individual patient et al. Results of a HOVON/SAKK donor versus
combined prospective analysis by the German data from randomised controlled trials. Lancet no-donor analysis of myeloablative HLA-identical
AML Intergroup. J Clin Oncol. 2012;30(29): Oncol. 2014;15(9):986-996. sibling stem cell transplantation in first remission
3604-3610. acute myeloid leukemia in young and middle-aged
28. Holowiecki J, Grosicki S, Giebel S, et al.
adults: benefits for whom? Blood. 2007;109(9):
15. Karp JE, Donehower RC, Enterline JP, Dole GB, Cladribine, but not fludarabine, added to
3658-3666.
Fox MG, Burke PJ. In vivo cell growth and daunorubicin and cytarabine during induction
pharmacologic determinants of clinical response prolongs survival of patients with acute myeloid 42. Koreth J, Schlenk R, Kopecky KJ, et al. Allogeneic
in acute myelogeneous leukemia. Blood. 1989; leukemia: a multicenter, randomized phase III stem cell transplantation for acute myeloid
73(1):24-30. study. J Clin Oncol. 2012;30(20):2441-2448. leukemia in first complete remission: systematic
From www.bloodjournal.org by guest on July 24, 2016. For personal use only.

60 DOMBRET and GARDIN BLOOD, 7 JANUARY 2016 x VOLUME 127, NUMBER 1

review and meta-analysis of prospective clinical 59. Walter RB, Buckley SA, Pagel JM, et al. syndrome after azanucleoside failure. Leuk Res.
trials. JAMA. 2009;301(22):2349-2361. Significance of minimal residual disease before 2014;38(4):443-446.
43. Schlenk RF, Döhner K, Krauter J, et al. Mutations myeloablative allogeneic hematopoietic cell
73. Stone RM, Mazzola E, Neuberg D, et al. Phase III
and treatment outcome in cytogenetically normal transplantation for AML in first and second
open-label randomized study of cytarabine in
acute myeloid leukemia. N Engl J Med. 2008; complete remission. Blood. 2013;122(10):
combination with amonafide L-malate or
358(18):1909-1918. 1813-1821.
daunorubicin as induction therapy for patients with
44. Gooley TA, Chien JW, Pegam SA, et al. Reduced 60. Boissel N, Renneville A, Leguay T, et al. secondary acute myeloid leukemia. J Clin Oncol.
mortality after allogeneic hematopoietic-cell Dasatinib in high-risk core binding factor 2015;33(11):1252-1257.
transplantation. N Engl J Med. 2010;363(22): acute myeloid leukemia in first complete
74. Genovese G, Kähler AK, Handsaker RE, et al.
2091-2101. remission: a French Acute Myeloid Leukemia
Clonal hematopoiesis and blood-cancer risk
Intergroup trial. Haematologica. 2015;100(6):
45. Luznik L, O’Donnell PV, Fuchs EJ. Post- inferred from blood DNA sequence. N Engl J Med.
780-785.
transplantation cyclophosphamide for tolerance 2014;371(26):2477-2487.
induction in HLA-haploidentical bone marrow 61. Paschka P, Schlenk RF, Weber D, et al. Dasatinib
75. Jaiswal S, Fontanillas P, Flannick J, et al. Age-
transplantation. Semin Oncol. 2012;39(6): (DAS) in combination with chemotherapy and as
related clonal hematopoiesis associated with
683-693. maintenance in core-binding factor (CBF) acute
adverse outcomes. N Engl J Med. 2014;371(26):
myeloid leukemia (AML): a phase Ib/IIa study of
46. Ciurea SO, Zhang MJ, Bacigalupo AA, et al. 2488-2498.
the German-Austrian AML Study Group (AMLSG)
Haploidentical transplant with post-transplant
[abstract]. Haematologica. 2015;100(s1). Abstract 76. Becker H, Marcucci G, Maharry K, et al.
cyclophosphamide versus matched unrelated
515. Favorable prognostic impact of NPM1 mutations
donor transplant for acute myeloid leukemia.
62. Marcucci G, Geyer S, Zhao W, et al. Adding in older patients with cytogenetically normal de
Blood. 2015;126(8):1033-1040.
KIT inhibitor dasatinib (DAS) to chemotherapy novo acute myeloid leukemia and associated
47. Russell NH, Kjeldsen L, Craddock C, et al. gene- and microRNA-expression signatures: a
overcomes the negative impact of KIT mutation/
A comparative assessment of the curative Cancer and Leukemia Group B study. J Clin
over-expression in core binding factor (CBF)
potential of reduced intensity allografts in acute Oncol. 2010;28(4):596-604.
acute myeloid leukemia (AML): results from
myeloid leukaemia. Leukemia. 2015;29(7):
CALGB 10801 (Alliance) [abstract]. Blood. 2014; 77. Gardin C, Chevret S, Pautas C, et al. Superior
1478-1484.
124(21). Abstract 8. long-term outcome with idarubicin compared with
48. Lioure B, Béné MC, Pigneux A, et al. Early high-dose daunorubicin in patients with acute
matched sibling hematopoietic cell transplantation 63. Levis M, Ravandi F, Wang ES, et al. Results from
a randomized trial of salvage chemotherapy myeloid leukemia age 50 years and older. J Clin
for adult AML in first remission using an age- Oncol. 2013;31(3):321-327.
adapted strategy: long-term results of a followed by lestaurtinib for patients with FLT3
prospective GOELAMS study. Blood. 2012; mutant AML in first relapse. Blood. 2011;117(12): 78. Burnett AK, Milligan D, Prentice AG, et al.
119(12):2943-2948. 3294-3301. A comparison of low-dose cytarabine and
64. Knapper S, Hills RK, Cavenagh JD, et al. A hydoxyurea with or without all-trans retinoic acid
49. Cornelissen JJ, Versluis J, Passweg JR, et al. for acute myeloid leukemia and high-risk
Comparative therapeutic value of post-remission randomised comparison of the sequential addition
of the FLT3 inhibitor lestaurtinib (CEP701) to myelodysplastic syndrome in patients not
approaches in patients with acute myeloid considered fit for intensive treatment. Cancer.
leukemia aged 40-60 years. Leukemia. 2015; standard first line chemotherapy for FLT3-
mutated acute myeloid leukemia: The UK 2007;109(6):1114-1124.
29(5):1041-1050.
experience [abstract]. Blood. 2014;124(21). 79. Kantarjian HM, Thomas XG, Dmoszynska A,
50. Passweg JR, Labopin M, Cornelissen J, et al. Abstract 3736. et al. Multicenter, randomized, open-label,
Conditioning intensity in middle-aged patients with
65. Chen YB, Shuli L, Andrew LA, et al. Phase I phase III trial of decitabine versus patient
AML in first CR: no advantage for myeloablative
trial of maintenance sorafenib after allogeneic choice, with physician advice, of either
regimens irrespective of the risk group-an
hematopoietic stem cell transplantation for supportive care or low-dose cytarabine for
observational analysis by the Acute Leukemia
patients with FLT3-ITD AML [abstract]. Blood. the treatment of older patients with newly
Working Party of the EBMT. Bone Marrow
2014;124(21). Abstract 671. diagnosed acute myeloid leukemia. J Clin
Transplant. 2015;50(8):1063-1068.
Oncol. 2012;30(21):2670-2677.
51. Brunet S, Martino R, Sierra J. Hematopoietic 66. Daver N, Cortes J, Ravandi F, et al. Secondary
transplantation for acute myeloid leukemia with mutations as mediators of resistance to targeted 80. Dombret H, Seymour JF, Butrym A, et al.
internal tandem duplication of FLT3 gene (FLT3/ITD). therapy in leukemia. Blood. 2015;125(21): International phase 3 study of azacitidine vs
Curr Opin Oncol. 2013;25(2):195-204. 3236-3245. conventional care regimens in older patients with
newly diagnosed AML with .30% blasts. Blood.
52. Pratcorona M, Brunet S, Nomdedéu J, et al. 67. Burnett AK, Hills RK, Green C, et al. The impact 2015;126(3):291-299.
Favorable outcome of patients with acute myeloid on outcome of the addition of all-trans retinoic
leukemia harboring a low-allelic burden FLT3-ITD acid to intensive chemotherapy in younger 81. Burnett AK, Russell NH, Culligan D, et al. The
mutation and concomitant NPM1 mutation: patients with nonacute promyelocytic acute addition of the farnesyl transferase inhibitor,
relevance to post-remission therapy. Blood. 2013; myeloid leukemia: overall results and results in tipifarnib, to low dose cytarabine does not improve
121(14):2734-2738. genotypic subgroups defined by mutations in outcome for older patients with AML. Br J
NPM1, FLT3, and CEBPA. Blood. 2010;115(5): Haematol. 2012;158(4):519-522.
53. Linch DC, Hills RK, Burnett AK, Khwaja A, Gale
948-956. 82. Burnett AK, Hills RK, Hunter A, et al. The addition
RE. Impact of FLT3(ITD) mutant allele level on
relapse risk in intermediate-risk acute myeloid 68. Schlenk RF, Döhner K, Kneba M, et al. Gene of arsenic trioxide to low-dose Ara-C in older
leukemia. Blood. 2014;124(2):273-276. mutations and response to treatment with all-trans patients with AML does not improve outcome.
retinoic acid in elderly patients with acute myeloid Leukemia. 2011;25(7):1122-1127.
54. Schlenk RF, Kayser S, Bullinger L, et al.
Differential impact of allelic ratio and insertion leukemia. Results from the AMLSG trial AML 83. Burnett AK, Hills RK, Hunter AE, et al. The
site in FLT3-ITD-positive AML with respect to HD98B. Haematologica. 2009;94(1):54-60. addition of gemtuzumab ozogamicin to low-
allogeneic transplantation. Blood. 2014;124(23): 69. Schlenk RF, Döhner K, Krauter J, et al. All-trans dose Ara-C improves remission rate but does
3441-3449. retinoic acid improves outcome in younger not significantly prolong survival in older
55. Grimwade D, Freeman SD. Defining minimal patients with nucleophosmin-1 mutated acute patients with acute myeloid leukaemia: results
residual disease in acute myeloid leukemia: which myeloid leukemia. Results of the AMLSG 07-04 from the LRF AML14 and NCRI AML16 pick-a-
platforms are ready for “prime time”? Blood. 2014; randomized treatment trial [abstract]. Blood. winner comparison. Leukemia. 2013;27(1):
124(23):3345-3355. 2011;118(21). Abstract 80. 75-81.

56. Kayser S, Walter RB, Stock W, Schlenk RF. 70. El Hajj H, Dassouki Z, Berthier C, et al. Retinoic 84. Dennis M, Russell N, Hills RK, et al. Vosaroxin
Minimal residual disease in acute myeloid acid and arsenic trioxide trigger degradation of and vosaroxin plus low-dose Ara-C (LDAC) vs
leukemia–current status and future perspectives. mutated NPM1, resulting in apoptosis of AML low-dose Ara-C alone in older patients with acute
Curr Hematol Malig Rep. 2015;10(2):132-144. cells. Blood. 2015;125(22):3447-3454. myeloid leukemia. Blood. 2015;125(19):
2923-2932.
57. Chen X, Xie H, Wood BL, et al. Relation of clinical 71. Martelli MP, Gionfriddo I, Mezzasoma F, et al.
response and minimal residual disease and their Arsenic trioxide and all-trans retinoic acid target 85. Burnett AK, Russell N, Hills RK, et al.
prognostic impact on outcome in acute myeloid NPM1 mutant oncoprotein levels and induce A randomised comparison of the novel nucleoside
leukemia. J Clin Oncol. 2015;33(11):1258-1264. apoptosis in NPM1-mutated AML cells. Blood. analogue sapacitabine with low-dose cytarabine
2015;125(22):3455-3465. in older patients with acute myeloid leukaemia.
58. Jourdan E, Boissel N, Chevret S, et al.
Leukemia. 2015;29(6):1312-1319.
Prospective evaluation of gene mutations and 72. Jaglal MV, Duong VH, Bello CM, et al. Cladribine,
minimal residual disease in patients with core cytarabine, filgrastim, and mitoxantrone 86. Burnett AK, Russell NH, Hunter AE, et al.
binding factor acute myeloid leukemia. Blood. (CLAG-M) compared to standard induction in Clofarabine doubles the response rate in older
2013;121(12):2213-2223. acute myeloid leukemia from myelodysplastic patients with acute myeloid leukemia but does
From www.bloodjournal.org by guest on July 24, 2016. For personal use only.

BLOOD, 7 JANUARY 2016 x VOLUME 127, NUMBER 1 TREATMENT OF ADULT AML 61

not improve survival. Blood. 2013;122(8): and high risk myelodysplastic syndrome 91. Ravandi F, Ritchie E, Sayar H, et al. Improved
1384-1394. (MDS) [abstract]. Blood. 2014;124(21). survival in patients with first relapsed or refractory
87. Döhner H, Lübbert M, Fiedler W, et al. Abstract 385. acute myeloid leukemia (AML) treated with
Randomized, phase 2 trial of low-dose cytarabine 89. Breems DA, Van Putten WL, Huijgens PC, et al. vosaroxin plus cytarabine versus placebo plus
with or without volasertib in AML patients not Prognostic index for adult patients with acute cytarabine: results of a phase 3 double-blind
suitable for induction therapy. Blood. 2014;124(9): myeloid leukemia in first relapse. J Clin Oncol. randomized controlled multinational study
1426-1433. 2005;23(9):1969-1978. (VALOR) [abstract]. Blood. 2014;124(21).
Abstract LBA 6.
88. Naval D, Kantarjian HM, Garcia-Manero G, 90. Burnett AK, Goldstone A, Hills RK, et al. Curability
et al. Phase I/II study of vosaroxin and of patients with acute myeloid leukemia who did 92. O’Donnell MR, Abboud CN, Altman J, et al. Acute
decitabine in newly diagnosed older patients not undergo transplantation in first remission. myeloid leukemia. J Natl Compr Canc Netw.
(pts) with acute myeloid leukemia (AML) J Clin Oncol. 2013;31(10):1293-1301. 2012;10(8):984-1021.
From www.bloodjournal.org by guest on July 24, 2016. For personal use only.

2016 127: 53-61


doi:10.1182/blood-2015-08-604520 originally published
online December 10, 2015

An update of current treatments for adult acute myeloid leukemia


Hervé Dombret and Claude Gardin

Updated information and services can be found at:


http://www.bloodjournal.org/content/127/1/53.full.html
Articles on similar topics can be found in the following Blood collections
Clinical Trials and Observations (4361 articles)
Free Research Articles (3946 articles)
Myeloid Neoplasia (1519 articles)
Review Articles (645 articles)
Review Series (109 articles)

Information about reproducing this article in parts or in its entirety may be found online at:
http://www.bloodjournal.org/site/misc/rights.xhtml#repub_requests

Information about ordering reprints may be found online at:


http://www.bloodjournal.org/site/misc/rights.xhtml#reprints

Information about subscriptions and ASH membership may be found online at:
http://www.bloodjournal.org/site/subscriptions/index.xhtml

Blood (print ISSN 0006-4971, online ISSN 1528-0020), is published weekly by the American Society
of Hematology, 2021 L St, NW, Suite 900, Washington DC 20036.
Copyright 2011 by The American Society of Hematology; all rights reserved.

Vous aimerez peut-être aussi