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

Long-term cure of patients with acute promyelocytic leukemia (APL) using retinoic acid and arsenic trioxide (ATO) therapy represents the only dramatic therapeutic advance over the last 2 decades in acute myeloid leukemia (AML).1  Although increasingly refined knowledge of AML biology has led to the development of new targeted agents such as the mutated FLT3 or IDH inhibitors, current advances in non-APL patients lack innovation, relying instead on modifications to doses and schedules of standard cytotoxic drugs or progress in hematopoietic stem cell transplantation (HSCT) techniques. In younger patients, complete remission (CR) rates of ≥80% may be reached, with 5-year overall survival (OS) ∼40%. In older patients, the use of hypomethylating agents has improved median and short-term OS but has not translated into improved cure rates, which remain disappointingly very low. Guidelines for AML management are widely available. This review aims to provide a balanced perspective of available data supporting AML treatment used in routine practice today. We have focused on data from randomized clinical trials using approved drugs. Early results with new investigational drugs will be discussed in depth in another article of this Review Series.

Induction therapy with cytarabine and an anthracycline remains a standard of care in AML. The standard combination is the 7+3, with a 7-day continuous infusion of cytarabine at the dosage of 100 or 200 mg/m2 per day on days 1 to 7 and daunorubicin at 60 mg/m2 per day on days 1 to 3. Recent randomized studies have investigated higher doses of anthracyclines or cytarabine or the addition of a third agent during induction. However, it is very difficult to compare these studies, because they differ significantly in several key parameters, notably the number of induction courses, doses in the control arm, and subsequent therapies offered to responders or to patients with persistent marrow blasts after the first induction course. Differences in study results may thus be caused by differences in the design of either experimental or control arms.

Anthracyclines

Recently reported randomized studies exploring daunorubicin or idarubicin doses during induction courses are summarized in Table 1.2-8  Four studies have evaluated higher daunorubicin doses.2,4-6  The Eastern Cooperative Oncology Group (ECOG) trial included patients aged ≤60 years,2  whereas the European trial from a Dutch, Belgian, German, and Swiss consortium included patients aged ≥60 years.4  Both studies compared a daily dose of 45 mg/m2 vs a doubled dose of 90 mg/m2 for 3 days as part of 7+3 induction therapy. The higher daunorubicin dose was associated with higher CR rates, without 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 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 was restricted to patients between the age of 60 and 65 years and to the few patients with core-binding factor (CBF) AML.4 

Table 1

Randomized studies of anthracycline dose and type for AML induction therapy

StudyAge (y)Patients (N)Experimental armControl armKey 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 
StudyAge (y)Patients (N)Experimental armControl armKey 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 7+3 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 5+2 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.

These results demonstrate that the 45 mg/m2 daunorubicin daily dose is suboptimal up until the age of 65 years. They do not, however, establish that the 90 mg/m2 daily dose should be preferred over the 60 mg/m2 daily dose endorsed by the international 2010 guidelines.9  The recent UK NCRI AML17 trial addressed this question in an up-front comparison of the 60 and 90 mg/m2 doses in the first course.6  All favorable- and intermediate-risk patients went on to receive a second course with a 50 mg/m2 dose. In an interim analysis, a significant increase in day 60 mortality was observed in the 90 mg/m2 arm, leading to premature trial termination. No OS benefit was observed in this analysis, although evidence of a survival benefit in subgroups may require longer follow-up.

The Acute Leukemia French Association (ALFA) 9801 study comparing daunorubicin vs idarubicin, failed to demonstrate superiority of 80 mg/m2 daunorubicin over a standard 12 mg/m2 idarubicin dose.7  A similar outcome was reported in the Japan Acute Leukemia Study Group (JALSG) AML201 study, which compared 2 courses of 50 mg/m2 daunorubicin for 5 days with 12 mg/m2 idarubicin for 3 days.8 

Antileukemic activity of 60 and 90 mg/m2 daunorubicin or 12 mg/m2 idarubicin daily doses thus appears to be similar, as are the toxicity profiles, at least in cases when a second anthracycline-containing cycle is not given systematically to responders.

Cytarabine

Randomized studies exploring cytarabine dose and schedule during induction are summarized in Table 2. Historical studies from the Southwest Oncology Group (SWOG) and the Australian Leukemia Study Group (ALSG) failed to demonstrate clinically relevant gains in efficacy with higher cytarabine doses with alternative administration regimens, whereas both reported increased toxicity.10,11  Two recent studies further addressed this question, one in the context of idarubicin or amsacrine and the other using lower etoposide doses and alternative administration schedules.

Table 2

Randomized studies of cytarabine dose for AML induction therapy

StudyAge (y)Patients (N)Experimental armControl armConclusions
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 d5-7 cycle 1 A, 200 mg/m2 CIV d1-7 I, 12 mg/m2 d5-7 cycle 1 Similar response rate 
A, 2000 mg/m2/12 h d1/2/4/6 Am, 120 mg/m2 d3/5/7 cycle 2§ A, 1000 mg/m2/12 h d1-6 Am, 120 mg/m2 d3/5/7 cycle 2§ Similar EFS and OS 
EORTC-GIMEMA AML1213  15-60 1942 A, 3000 mg/m2/12 h d1/3/5/7 A, 100 mg/m2 CIV d1-10 Higher response rate 
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  
StudyAge (y)Patients (N)Experimental armControl armConclusions
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 d5-7 cycle 1 A, 200 mg/m2 CIV d1-7 I, 12 mg/m2 d5-7 cycle 1 Similar response rate 
A, 2000 mg/m2/12 h d1/2/4/6 Am, 120 mg/m2 d3/5/7 cycle 2§ A, 1000 mg/m2/12 h d1-6 Am, 120 mg/m2 d3/5/7 cycle 2§ Similar EFS and OS 
EORTC-GIMEMA AML1213  15-60 1942 A, 3000 mg/m2/12 h d1/3/5/7 A, 100 mg/m2 CIV d1-10 Higher response rate 
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.

In the study conducted by the Dutch-Belgian Cooperative Trial Group for Hemato-Oncology (HOVON) and the Swiss Group for Clinical Cancer Research (SAKK), patients were randomized between an IDAC arm, comprising 200 mg/m2 per day cytarabine during the first course and 1000 mg/m2 every 12 hours for 6 days during the second course, or a high-dose (HiDAC) arm with 1000 mg/m2 every 12 hours for 5 days during the first course and 2000 mg/m2 every 12 hours on days 1, 2, 4, and 6 during the second course.12  Similar rates of CR, event-free survival (EFS), and OS were observed in the 2 arms, with more toxicity associated with the HiDAC arm.

In the second study, conducted by the EORTC and GIMEMA Leukemia Groups, patients were randomized to receive either standard doses at 100 mg/m2 per day cytarabine for 10 days or HiDAC at 3000 mg/m2 every 12 hours on days 1, 3, 5, and 7 during the first course.13  A higher CR rate was observed in the HiDAC arm, with a trend for a longer OS that reached statistical significance in the subset of patients aged ≤45 years. Finally, in the randomized German Intergroup study that included a double induction as a “common” arm or an IDAC or HiDAC sequence during induction in at least 3 study arms, no difference in OS was observed.14  It thus remains unclear whether increasing the cytarabine dose during induction may benefit patients planned to receive IDAC or HiDAC during postremission therapy.

“Dose-dense” regimens

Another approach to increasing induction intensity relies on systematic administration of a second sequence of chemotherapy starting earlier than normal after the completion of the first sequence (generally between day 7 and day 14). This timed-sequential concept was initially developed by the Johns Hopkins group in Baltimore,15  then prospectively evaluated by the ALFA group without incorporating HiDAC.16  After investigating double induction containing 1 or 2 HiDAC sequences (TAD-HAM or HAM-HAM),17  the German AML Cooperative Group recently conducted a phase 2 trial investigating a sequential S-HAM.18  However, none of these studies provide evidence that a dose-dense regimen is superior to the standard 7+3 regimen, especially when high doses of daunorubicin are used.

Addition of a third drug

Gemtuzumab ozogamicin (GO).

Randomized studies exploring the addition of GO to intensive chemotherapy (ICT) are summarized in Table 3.19-26  Six studies evaluating the addition of 3 or 6 mg/m2 GO during induction have been performed.19-24  The first of them, SWOG S0106, was negative and closed prematurely because of a higher early mortality rate despite a reduced 45 mg/m2 per day daunorubicin dose in the GO arm, leading to GO withdrawal by the Food and Drug Administration in the United States.19  Conversely, 4 studies reported significant improvements when GO was combined with induction or induction-and-consolidation chemotherapy.20-23  This finding was confirmed in a recent meta-analysis.27  Nonetheless, the addition of GO was associated with more frequent severe liver toxicity and persistent thrombocytopenia. The benefit/risk ratio appeared to be at least as good with 3 mg/m2 per dosing,24  a dose that can be administered to older patients,21  and ultimately be repeated as successfully developed by the ALFA group.22  In contrast, increased toxicity and an absence of clinical benefit were observed in the EORTC/GIMEMA study, in which single-agent GO administration preceded induction chemotherapy.25  Finally, the 2 studies that evaluated GO maintenance therapy were negative.19,26  Clinical response to GO is likely to be influenced by CD33 expression level, clonal hierarchy of the leukemic population, and drug efflux intensity, all factors related to AML genetics. It has been widely demonstrated that GO benefits patients of favorable and intermediate risk, including those with FLT3-ITD, although not those with an adverse karyotype.27  Together, these data suggest that the 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.

Table 3

Randomized trials of gemtuzumab ozogamicin (GO) associated with intensive chemotherapy

StudyTreatment phase (age range [y])Patients (N)GO dose and scheduleConclusions
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 
StudyTreatment phase (age range [y])Patients (N)GO dose and scheduleConclusions
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.

Purine analogs.

Randomized studies exploring the addition of purine analogs to ICT are summarized in Table 4.28-32  In the Polish Acute Leukemia Group (PALG) study, the addition of cladribine was associated with prolonged OS,28  although it should be noted that outcomes in the control arm appeared to be suboptimal. Interestingly, addition of cladribine appears to have particularly benefited high-risk patients, notably those aged ≥50 years or those with unfavorable cytogenetics. Clofarabine also demonstrated significant antileukemic activity, both as a single agent and in combination with cytarabine in various patient populations.33,34  However, it failed to show significant benefit when combined with daunorubicin in the British AML16 trial in newly diagnosed patients.29  Front-line results from 2 other trials are imminent, including the combination of clofarabine with 7+3,35  and clofarabine compared with 7+3 in an ECOG trial. The British AML15 trial in a younger population demonstrated that the fludarabine, cytarabine, granulocyte colony-stimulating factor (G-CSF), and idarubicin regimen (FLAG-Ida) yielded more frequent remissions with one course and also reduced the risk of relapse.30 

Table 4

Randomized trials of purine analogs associated to intensive chemotherapy

StudyAnalogTreatment phase (age range [y])Patients (N)Treatment modalitiesConclusions
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  
StudyAnalogTreatment phase (age range [y])Patients (N)Treatment modalitiesConclusions
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 + etoposide; CLARA, clofarabine + IDAC; DA, daunorubicin + cytarabine; DAC, DA + cladribine; DAF, DA + fludarabine; DClo, daunorubicin + clofarabine; FLAG-Ida, fludarabine + IDAC + G-CSF + 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 × 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.

Sorafenib.

Sorafenib is a multikinase inhibitor with in vitro activity in FLT3-ITD AML. In an AML trial from the German Study Alliance Leukemia, combination of sorafenib with standard induction and IDAC consolidation was evaluated in older patients. Treatment in the sorafenib arm did not result in significant improvement in EFS or OS.36  This was also true for subgroup analyses, including the subgroup positive for FLT3-ITD. Results of induction therapy were worse in the sorafenib arm, with higher early mortality and a lower CR rate. The same group has reported preliminary results of a similar trial conducted in younger patients in which sorafenib was added to 7+3 induction and HiDAC consolidations.37  No difference in the CR rate was observed, whereas EFS and relapse-free survival (RFS) were significantly improved in the sorafenib arm. In the subgroup of FLT3-ITD–positive patients, no difference in EFS was observed; however, there was a trend for prolonged RFS and OS favoring sorafenib.

HiDAC consolidation

For younger patients not undergoing HSCT, administration of several HiDAC consolidation courses using cytarabine twice daily at a 3 g/m2 dose on days 1, 3, and 5 has been a widely used option since 1994.38  Even if the optimal cytarabine dose, schedule of administration, and number of cycles need yet to be defined,39,40  the use of bolus administration of HiDAC or IDAC during consolidation should be recommended. Interestingly, HiDAC and IDAC regimens are well suited to evaluate the effects of added targeted or nontargeted drugs during consolidation. For instance, a prolonged RFS was observed in the randomized ALFA-0702/CLARA study in intermediate- and unfavorable-risk patients with clofarabine/IDAC compared with HiDAC consolidation cycles.31 

Allogeneic HSCT

One of the most important treatment decisions in AML is to estimate the benefit/risk associated with allogeneic HSCT in first remission for a given patient. Transplantation offers the best means of preventing AML recurrence, but remains associated with higher treatment-related morbidity and mortality (TRM), especially in older patients. In patients with favorable-risk AML, the relapse risk may be low enough and the salvage rate high enough to postpone HSCT to second remission. This 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-donor group did as well as those from the donor group, whereas all other patients appeared to benefit from undergoing allograft. One should keep in mind that patients in these studies mostly underwent sibling donor myeloablative conditioning (MAC) transplantation and as such, the benefit associated with HSCT was only demonstrated for patients <40 years of age. Based on another donor vs no-donor analysis, patients with cytogenetically normal (CN) AML and a favorable genotype (defined as mutated CEBPA or NPM1 without FLT3-ITD) were recently categorized in the favorable subgroup.43  Because the outcome after allogeneic HSCT from fully matched unrelated donors appears to be similar compared with allogeneic HSCT from matched related donors, all younger patients with intermediate- and unfavorable-risk AML are generally considered candidates for allogeneic HSCT from sibling or fully-matched unrelated donors in cases of first CR.7 

This HSCT benefit/risk assessment, based on the European LeukemiaNet (ELN) genetic classification only,7  needs however to be reconsidered in the near future. As discussed in another article of this Review Series, the incidence of TRM has been substantially reduced over the last few decades,44  particularly in older patients, by using reduced-intensity conditioning (RIC). Alternative stem cell sources are more widely used and are safer, as illustrated by post-transplant administration of cyclophosphamide in haplo-identical HSCT.45,46  Of particular interest are the recent results of studies evaluating RIC transplantation in middle-aged patients.47-50  Using a time-dependent Mantel-Byar comparison in patients >45 years of age, the MRC AML15 trial showed that, compared with chemotherapy, RIC transplantation was associated with longer survival.47  Two recent studies have even suggested that RIC might be preferred to MAC transplantation in this age range, given the lower TRM as well as better survival in some risk subgroups.49,50 

Reliance on genetic profiles as the main treatment-stratifying tool is being increasingly challenged because of multiple recently described genetic mutations and the potential impact of mutated allele frequencies. Not all patients with ELN favorable-risk AML have a favorable outcome; for instance, should we take into account the presence of additional KIT or FLT3 poor-risk mutations in a patient with CBF-AML, or of additional ASXL1, IDH1 or DNMT3A mutations in a patient with NPM1-mutated CN-AML and no FLT3-ITD? Likewise, should HSCT be considered the best postremission option in ELN intermediate-risk patients with NPM1-mutated FLT3-ITD–positive CN-AML but a low FLT3-ITD allelic ratio?51-54 

Assessment of minimal residual disease (MRD) using molecular markers or leukemia-associated aberrant immunophenotypes is therefore proposed as a more straightforward, easy-to-use prognostic decision tool, including deciding which patients should be advised to undergo allogeneic HSCT.55,56  As in acute lymphoblastic leukemia, MRD has been suggested as ultimately superseding other prognostic factors for HSCT decision-making, including genetic factors,57  as is already the case for CBF-AML.58  Nevertheless, whether allogeneic HSCT may be the option of choice in poor early MRD responders remains an open issue.59 

Core binding factor AML

A subgroup analysis of the MRC AML15 younger population AML trial, along with a subsequent meta-analysis,20-24,27  strongly suggests that the combination of GO with ICT is associated with a significant OS benefit in CBF-AML, renewing interest in this drug, which is currently unavailable outside the context of clinical studies, for this AML subset.

Frequent KIT mutations or overexpression of the KIT receptor were described some time ago in CBF-AML. Recent studies have evaluated the potential benefit of dasatinib, a potent KIT inhibitor, in these patients. In a French study, patients with persistent MRD or molecular relapse after intensive consolidations were eligible to receive 12 months of maintenance with dasatinib if they were not a candidate for HSCT.60  No significant impact of dasatinib on time-to-relapse was seen. Two phase 1-2 trials combined dasatinib with up-front ICT led to a currently ongoing phase 3 trial.61,62 

AML with FLT3 gene mutation

The FLT3-ITD mutation is one of the most frequent bad-prognosis mutations observed in AML, at least in younger or relapsed patients, and has led to the evaluation of multikinase or more specific kinase inhibitors in this AML subset. Lestaurtinib associated with chemotherapy failed to improve outcomes in relapsed/refractory (R/R) FLT3-ITD AML patients or during front-line consolidation in the MRC AML15 trial.63,64  Positive results from a large international phase 3 trial in newly diagnosed FLT3-ITD–positive patients in which midostaurin was compared to placebo in association with 7+3 should be available soon. As mentioned before, to date there is no clear evidence of increased clinically significant activity when sorafenib is added to ICT in FLT3-ITD–positive patients. A phase 2 study of sorafenib combined with ICT in older patients, restricted to those diagnosed with FLT3-mutated AML, is expected to shed light on this issue (ClinicalTrials.gov #NCT01253070). Conversely, sorafenib delivered after HSCT seems to be particularly effective in FLT3-ITD–positive patients.65  Selection of FLT3-resistant mutants has been a general phenomenon observed in most early trials with first- or second-generation inhibitors.66  An overview of novel promising FLT3 inhibitors is presented elsewhere in this Review Series.

AML with NPM1 gene mutation

Approximately 30% of CN-AML patients have an NPM1 mutation and, in contrast to the frequently associated FLT3-ITD, the NPM1 mutation seems to be an early event in most of these cases. Several trials have reported the effects of adding all-trans retinoic acid (ATRA) to chemotherapy in non-APL AML, with conflicting results. In the large MRC trial, no significant effect of the addition of ATRA was observed.67  Alternatively, 2 studies from the German AML Study Group reported a benefit of the addition of ATRA restricted to NPM1-mutated AML patients.68,69  An explanation for these contradictory results is yet to be defined. More recently, preclinical studies of arsenic trioxide (ATO) in NPM1-mutated AML from 2 groups strongly suggested that ATO may have a beneficial effect in this subset.70,71  Because extensive clinical experience with ATO has been gained in the 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

Isocitrate dehydrogenase 1 and 2 gene mutations are present in 10% to 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-mutated enzymes are impressive. A phase 3 study will soon be initiated in relapsing IDH2-mutated patients.

AML with adverse cytogenetic features

In this hard-to-treat AML population, improvement of early response rate remains an important clinical end point, especially when HSCT can be envisioned in a timely manner. However, results of trials with novel drugs are disappointing. In the PALG cladribine study,28  the subgroup of patients with unfavorable cytogenetics appeared, nevertheless, to have a greater benefit. Similarly, a combination of cladribine, cytarabine, priming with G-CSF, and mitoxantrone (CLAG-M) was reported to be of benefit compared with 7+3 in a large, retrospective, single-center study that primarily included high-risk AML patients.72  Among other nontargeted agents, amonafide combined with cytarabine failed to yield a benefit over 7+3 in a secondary AML study.73  Early results of new drugs, including the CPX-351 liposomal combination, are discussed elsewhere in this Review Series.

Outcome in older patients with AML remains dismal, with lower CR rates and very few long-term survivors compared with younger patients. Treatment of older AML patients is thus an active field of antileukemic drug investigation. Even when short-term benefits have been demonstrated, very few older patients survive beyond 2 years of follow-up, raising the question of the appropriateness of OS as an efficacy end point for drug development in this patient population. Age itself does not, however, define a homogeneous patient population. In older patients, prognosis is governed by patient-related and AML-related factors that are only partly age-related. General health status and the presence of organ dysfunctions or comorbidities affect ICT tolerance. As observed in younger patients, cytogenetics can also affect efficacy. One predominant bad-risk characteristic of older AML is the increased frequency of prior myelodysplastic syndromes, although clonal hematopoiesis characterized by the presence of myelodysplastic syndromes–related gene mutations has also been associated with aging in healthy individuals.74,75 

The most important clinical decision remains to estimate the benefit/risk associated with ICT in the individual older patient. As for the decision to recommend HSCT in first CR, there is no unique decision-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 AML14 MRC trial results, despite the fact that patients with adverse cytogenetics did not draw any benefit from LDAC therapy.78  In 2 large international trials, the hypomethylating agents decitabine and azacitidine yielded better mid-term results, with longer median and higher 1-year survival than those observed in LDAC arms, even if they did not result in a higher proportion of long-term survivors.79,80  Of interest, azacitidine appeared to offer particular benefit to patients with adverse cytogenetics and/or those with myelodysplasia-related changes.80 

Current trials in older AML patients often address the effects of the addition of a new agent to LDAC, azacitidine, or decitabine. To date, negative results with tipifarnib, ATO, GO, and vosaroxin in combination with LDAC have been reported by the British group.81-84  Single-agent sapacitabine or clofarabine also failed to improve outcomes over LDAC.85,86  Addition of quizartinib, ganetespib, tosedostat, or selinexor to LDAC is currently under investigation. Interesting phase 2 results have been observed when combining the polo-like kinase inhibitor volasertib with LDAC,87  or vosaroxin with decitabine in a single-center evaluation.88  Too frequently, the evaluation of the efficacy of the combinations is hindered by their poor tolerance in older patients deemed unfit for standard ICT, raising the issue of their evaluation in fitter patients, potentially against ICT.

Treatment of relapsed AML remains poorly defined. Simple clinical and disease parameters such as age, duration of first remission, cytogenetics, and prior HSCT remain the most useful parameters to evaluate treatment effects at relapse.89  In most AML subsets (other than APL) the main clinical objective of salvage therapy is to “bridge” patients to HSCT, either with targeted therapies such as 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-term outcomes clearly established that allogeneic transplantation when a second response was obtained, strongly benefited intermediate- and high-risk AML subsets based on initial disease characteristics.

Optimal salvage drug combinations, drug doses, and administration schedules remain open issues. In particular, the benefits of the addition of an anthracycline, or any other drug, to cytarabine, and what cytarabine dose should be recommended are unclear at best. Recently, 2 large company-sponsored studies evaluated the combination of clofarabine or vosaroxin to IDAC, with OS as the end point in R/R AML patients. In the CLASSIC I trial, addition of clofarabine increased the response rate in patients >55 years of age, albeit at the expense of increased toxicity and early mortality, but failed to improve OS despite a higher number of patients bridged to HSCT.32  In the preliminary results of the largest trial ever performed in R/R patients, the VALOR trial, combination of vosaroxin to IDAC similarly increased the response rate across all ages in both relapsed and refractory patients. Nonetheless, despite the absence of a significant increase in early mortality and a large proportion of younger patients bridged to HSCT, this did not translate into general improvement in OS. A significant, although modest, survival improvement was only observed in older patients ≥60 years of age.91 

Despite some advances in the treatment of adult AML patients, many issues remain to be addressed, as reflected by current recommendations.7,92  Given the poor long-term outcome for most adult AML patients, although many novel promising drugs or therapeutic approaches are currently under development for this population, their inclusion into prospective clinical trials should be strongly encouraged. In addition, standardization of trial procedures and control arms would greatly assist comparison of trial results and strengthen future treatment recommendations.

Contribution: H.D. and C.G. wrote this review article jointly.

Conflict-of-interest disclosure: H.D. is on the Advisory Board of Celgene, Pfizer, Sunesis, and Agios Pharmaceuticals. C.G. is on the Advisory Board of Celgene.

Correspondence: Hervé Dombret, Hopital Saint-Louis, Assistance Publique - Hopitaux de Paris (AP-HP), Paris, France; e-mail: herve.dombret@aphp.fr.

1
Lo-Coco
 
F
Avvisati
 
G
Vignetti
 
M
, et al. 
Retinoic acid and arsenic trioxide for acute promyelocytic leukemia.
N Engl J Med
2013
, vol. 
369
 
2
(pg. 
111
-
121
)
2
Fernandez
 
HF
Sun
 
Z
Yao
 
X
, et al. 
Anthracycline dose intensification in acute myeloid leukemia.
N Engl J Med
2009
, vol. 
361
 
13
(pg. 
1249
-
1259
)
3
Luskin
 
MR
Lee
 
J-W
Fernandez
 
HF
, et al. 
High dose daunorubicin improves survival in AML up to age 60, across all cytogenetic risk groups including patients with unfavorable cytogenetic risk and FLT3-ITD mutant AML: updated analysis from Eastern Cooperative Oncology Trial E1900 [abstract].
Blood
2014
, vol. 
124
 
21
 
Abstract 373
4
Löwenberg
 
B
Ossenkoppele
 
GJ
van Putten
 
W
, et al. 
High-dose daunorubicin in older patients with acute myeloid leukemia.
N Engl J Med
2009
, vol. 
361
 
13
(pg. 
1235
-
1248
)
5
Lee
 
JH
Joo
 
YD
Kim
 
H
, et al. 
A randomized trial comparing standard versus high-dose daunorubicin induction in patients with acute myeloid leukemia.
Blood
2011
, vol. 
118
 
14
(pg. 
3832
-
3841
)
6
Burnett
 
AK
Russell
 
NH
Hills
 
RK
, et al. 
A randomized comparison of daunorubicin 90 mg/m2 vs 60 mg/m2 in AML induction: results from the UK NCRI AML17 trial in 1206 patients.
Blood
2015
, vol. 
125
 
25
(pg. 
3878
-
3885
)
7
Pautas
 
C
Merabet
 
F
Thomas
 
X
, et al. 
Randomized study of intensified anthracycline doses for induction and recombinant interleukin-2 for maintenance in patients with acute myeloid leukemia aged 50 to 70 years: Results of the ALFA-9801 Study.
J Clin Oncol
2010
, vol. 
28
 
5
(pg. 
808
-
814
)
8
Ohtake
 
S
Miyawaki
 
S
Fujita
 
H
, et al. 
Randomized trial of induction therapy comparing standard-dose idarubicin with high-dose daunorubicin in adult patients with previously untreated acute myeloid leukemia: the JALSG AML201 Study.
Blood
2011
, vol. 
117
 
8
(pg. 
2358
-
2365
)
9
Döhner
 
H
Estey
 
EH
Amadori
 
S
, et al. 
Diagnosis and management of acute myeloid leukemia in adults: recommendations from an international expert panel, on behalf of the European LeukemiaNet.
Blood
2010
, vol. 
115
 
3
(pg. 
453
-
474
)
10
Weick
 
JL
Kopecky
 
KJ
Appelbaum
 
FR
, et al. 
A randomized investigation of high-dose versus standard-dose cytosine arabinoside with daunorubicin in patients with previously untreated acute myeloid leukemia: a Southwest Oncology Group study.
Blood
1996
, vol. 
88
 
8
(pg. 
2841
-
2851
)
11
Bishop
 
JF
Matthews
 
JP
Young
 
GA
, et al. 
Randomized study of high-dose cytarabine in induction in acute myeloid leukemia.
Blood
1996
, vol. 
87
 
5
(pg. 
1710
-
1717
)
12
Löwenberg
 
B
Pabst
 
T
Vellenga
 
E
, et al. 
Cytarabine dose for acute myeloid leukemia.
N Engl J Med
2011
, vol. 
364
 
11
(pg. 
1027
-
1036
)
13
Willemze
 
R
Suciu
 
S
Meloni
 
G
, et al. 
High-dose cytarabine in induction treatment improves the outcome of adult patients younger than age 46 years with acute myeloid leukemia: results of the EORTC-GIMEMA AML-12 trial.
J Clin Oncol
2014
, vol. 
32
 
3
(pg. 
219
-
228
)
14
Büchner
 
T
Schlenk
 
RF
Schaich
 
M
, et al. 
Acute myeloid leukemia (AML): different treatment strategies versus a common standard arm: combined prospective analysis by the German AML Intergroup.
J Clin Oncol
2012
, vol. 
30
 
29
(pg. 
3604
-
3610
)
15
Karp
 
JE
Donehower
 
RC
Enterline
 
JP
Dole
 
GB
Fox
 
MG
Burke
 
PJ
In vivo cell growth and pharmacologic determinants of clinical response in acute myelogeneous leukemia.
Blood
1989
, vol. 
73
 
1
(pg. 
24
-
30
)
16
Castaigne
 
S
Chevret
 
S
Archimbaud
 
E
, et al. 
Randomized comparison of double induction and timed-sequential induction to a « 3+7 » induction in adults with AML: long-term analysis of the Acute Leukemia French Association (ALFA) 9000 study.
Blood
2004
, vol. 
104
 
8
(pg. 
2467
-
2474
)
17
Büchner
 
T
Berdel
 
WE
Schooch
 
C
, et al. 
Double induction containing either two courses or one course of high-dose cytarabine plus mitoxantrone and post-remission therapy by either autologous stem-cell transplantation or by prolonged maintenance for acute myeloid leukemia.
J Clin Oncol
2006
, vol. 
24
 
16
(pg. 
2480
-
2489
)
18
Braess
 
J
Spiekermann
 
K
Staib
 
P
, et al. 
Dose-dense induction with sequential high-dose cytarabine and mitoxantrone (S-HAM) and pelfilgrastim results in a high efficacy and a short duration of critical neutropenia in de novo acute myeloid leukemia: a pilot study of the AMLCG.
Blood
2009
, vol. 
113
 
17
(pg. 
3903
-
3910
)
19
Petersdorf
 
SH
Kopecky
 
KJ
Slovak
 
M
, et al. 
A Phase 3 study of gemtuzumab ozogamicin during induction and postconsolidation therapy in younger patients with acute myeloid leukemia.
Blood
2013
, vol. 
121
 
24
(pg. 
4854
-
4860
)
20
Burnett
 
AK
Hills
 
RK
Milligan
 
D
, et al. 
Identification of patients with acute myeloid leukemia who benefit from the addition of gemtuzumab ozogamicin: results of the MRC AML15 trial.
J Clin Oncol
2011
, vol. 
29
 
4
(pg. 
369
-
377
)
21
Burnett
 
AK
Russell
 
NH
Hills
 
RK
, et al. 
Addition of gemtuzumab ozogamicin to induction chemotherapy improves survival in older patients with acute myeloid leukemia.
J Clin Oncol
2012
, vol. 
30
 
32
(pg. 
3924
-
3931
)
22
Castaigne
 
S
Pautas
 
C
Terré
 
C
, et al. 
Effect of gemtuzumab ozogamicin on survival of adult patients with de-novo acute myeloid leukaemia (ALFA-0701): a randomised, open-label, phase 3 study.
Lancet
2012
, vol. 
379
 
9825
(pg. 
1508
-
1516
)
23
Delaunay
 
J
Recher
 
C
Pigneux
 
A
, et al. 
Addition of gemtuzumab ozogamicin to chemotherapy improves event-free survival but not overall survival of AML patients with intermediate cytogenetics not eligible for allogeneic transplantation: results of the GOELAMS AML 2006 IR study [abstract].
Blood
2011
, vol. 
118
 
21
 
Abstract 79
24
Burnett
 
AK
Russell
 
N
Hills
 
RK
, et al. 
A comparison of single-dose gemtuzumab ozogamicin 3 mg/m2 and 6 mg/m2 combined with induction chemotherapy in younger patients with AML: data from the UK NCRI AML17 trial [abstract].
Blood
2014
, vol. 
124
 
21
 
Abstract 2308
25
Amadori
 
S
Suciu
 
S
Stasi
 
R
, et al. 
Sequential combination of gemtuzumab ozogamicin and standard chemotherapy in older patients with newly diagnosed acute myeloid leukemia: results of a randomized phase III trial by the EORTC and GIMEMA consortium (AML-17).
J Clin Oncol
2013
, vol. 
31
 
35
(pg. 
4424
-
4430
)
26
Löwenberg
 
B
Beck
 
J
Graux
 
C
, et al. 
Gemtuzumab ozogamicin as post-remission treatment in AML at 60 years of age or more: results of a multicenter phase 3 study.
Blood
2010
, vol. 
115
 
13
(pg. 
2586
-
2591
)
27
Hills
 
RK
Castaigne
 
S
Appelbaum
 
FR
, et al. 
Addition of gemtuzumab ozogamicin to induction chemotherapy in adult patients with acute myeloid leukaemia: a meta-analysis of individual patient data from randomised controlled trials.
Lancet Oncol
2014
, vol. 
15
 
9
(pg. 
986
-
996
)
28
Holowiecki
 
J
Grosicki
 
S
Giebel
 
S
, et al. 
Cladribine, but not fludarabine, added to daunorubicin and cytarabine during induction prolongs survival of patients with acute myeloid leukemia: a multicenter, randomized phase III study.
J Clin Oncol
2012
, vol. 
30
 
20
(pg. 
2441
-
2448
)
29
Russell
 
N
Burnett
 
A
Kjeldsen
 
L
, et al. 
A comparison of daunorubicin/ara-c versus daunorubicin/clofarabine as induction treatment in older patients with AML and high-risk MDS: Long term results of the UK NCRI AML16 trial in 806 patients [abstract].
Haematologica
2015
, vol. 
100
 
s1
 
Abstract 514
30
Burnett
 
AK
Russell
 
NH
Hills
 
RK
, et al. 
Optimization of chemotherapy for younger patients with acute myeloid leukemia: results of the medical research council AML15 trial.
J Clin Oncol
2013
, vol. 
31
 
27
(pg. 
3360
-
3368
)
31
Thomas
 
X
de Botton
 
S
Chevret
 
S
, et al. 
Clofarabine-based consolidation improves relapse-free survival of younger adults with non-favorable acute myeloid leukemia (AML) in first remission: results of the randomized ALFA-0702/CLARA study [abstract].
 
Blood. 2015;126(23). Abstract 218
32
Faderl
 
S
Wetzler
 
M
Rizzieri
 
D
, et al. 
Clofarabine plus cytarabine compared with cytarabine alone in older patients with relapsed or refractory acute myelogenous leukemia: results from the CLASSIC I Trial.
J Clin Oncol
2012
, vol. 
30
 
20
(pg. 
2492
-
2499
)
33
Faderl
 
S
Verstovsek
 
S
Cortes
 
J
, et al. 
Clofarabine and cytarabine combination as induction therapy for acute myeloid leukemia (AML) in patients 50 years of age or older.
Blood
2006
, vol. 
108
 
1
(pg. 
45
-
51
)
34
Kantarjian
 
HM
Erba
 
HP
Claxton
 
D
, et al. 
Phase II study of clofarabine monotherapy in previously untreated older adults with acute myeloid leukemia and unfavorable prognostic factors.
J Clin Oncol
2010
, vol. 
28
 
4
(pg. 
549
-
555
)
35
Willemze
 
R
Suciu
 
S
Muus
 
P
, et al. 
Clofarabine in combination with a standard remission induction regimen (cytosine arabinoside and idarubicin) in patients with previously untreated intermediate and bad-risk acute myelogenous leukemia (AML) or high-risk myelodysplastic syndrome (HR-MDS): phase I results of an ongoing phase I/II study of the leukemia groups of EORTC and GIMEMA (EORTC GIMEMA 06061/AML-14A trial).
Ann Hematol
2014
, vol. 
93
 
6
(pg. 
965
-
975
)
36
Serve
 
H
Krug
 
U
Wagner
 
R
, et al. 
Sorafenib in combination with intensive chemotherapy in elderly patients with acute myeloid leukemia: results from a randomized, placebo-controlled trial.
J Clin Oncol
2013
, vol. 
31
 
25
(pg. 
3110
-
3118
)
37
Röllig
 
C
Müller-Tidow
 
C
Hüttmann
 
A
, et al. 
Sorafenib versus placebo in addition to standard therapy in younger patients with newly diagnosed acute myeloid leukemia: results from 267 patients treated in the randomized placebo-controlled SAL-SORAML trial [abstract].
Blood
2014
, vol. 
124
 
21
 
Abstract 6
38
Mayer
 
RJ
Davis
 
RB
Schiffer
 
CA
, et al. 
Intensive postremission chemotherapy in adults with acute myeloid leukemia.
N Engl J Med
1994
, vol. 
331
 
14
(pg. 
896
-
903
)
39
Löwenberg
 
B
Sense and nonsense of high-dose cytarabine for acute myeloid leukemia.
Blood
2013
, vol. 
121
 
1
(pg. 
26
-
28
)
40
Schlenk
 
RF
Post-remission therapy for acute myeloid leukemia.
Haematologica
2014
, vol. 
99
 
11
(pg. 
1663
-
1670
)
41
Cornelissen
 
JJ
van Putten
 
WL
Verdonck
 
LF
, et al. 
Results of a HOVON/SAKK donor versus no-donor analysis of myeloablative HLA-identical sibling stem cell transplantation in first remission acute myeloid leukemia in young and middle-aged adults: benefits for whom?
Blood
2007
, vol. 
109
 
9
(pg. 
3658
-
3666
)
42
Koreth
 
J
Schlenk
 
R
Kopecky
 
KJ
, et al. 
Allogeneic stem cell transplantation for acute myeloid leukemia in first complete remission: systematic review and meta-analysis of prospective clinical trials.
JAMA
2009
, vol. 
301
 
22
(pg. 
2349
-
2361
)
43
Schlenk
 
RF
Döhner
 
K
Krauter
 
J
, et al. 
Mutations and treatment outcome in cytogenetically normal acute myeloid leukemia.
N Engl J Med
2008
, vol. 
358
 
18
(pg. 
1909
-
1918
)
44
Gooley
 
TA
Chien
 
JW
Pegam
 
SA
, et al. 
Reduced mortality after allogeneic hematopoietic-cell transplantation.
N Engl J Med
2010
, vol. 
363
 
22
(pg. 
2091
-
2101
)
45
Luznik
 
L
O’Donnell
 
PV
Fuchs
 
EJ
Post-transplantation cyclophosphamide for tolerance induction in HLA-haploidentical bone marrow transplantation.
Semin Oncol
2012
, vol. 
39
 
6
(pg. 
683
-
693
)
46
Ciurea
 
SO
Zhang
 
MJ
Bacigalupo
 
AA
, et al. 
Haploidentical transplant with post-transplant cyclophosphamide versus matched unrelated donor transplant for acute myeloid leukemia.
Blood
2015
, vol. 
126
 
8
(pg. 
1033
-
1040
)
47
Russell
 
NH
Kjeldsen
 
L
Craddock
 
C
, et al. 
A comparative assessment of the curative potential of reduced intensity allografts in acute myeloid leukaemia.
Leukemia
2015
, vol. 
29
 
7
(pg. 
1478
-
1484
)
48
Lioure
 
B
Béné
 
MC
Pigneux
 
A
, et al. 
Early matched sibling hematopoietic cell transplantation for adult AML in first remission using an age-adapted strategy: long-term results of a prospective GOELAMS study.
Blood
2012
, vol. 
119
 
12
(pg. 
2943
-
2948
)
49
Cornelissen
 
JJ
Versluis
 
J
Passweg
 
JR
, et al. 
Comparative therapeutic value of post-remission approaches in patients with acute myeloid leukemia aged 40-60 years.
Leukemia
2015
, vol. 
29
 
5
(pg. 
1041
-
1050
)
50
Passweg
 
JR
Labopin
 
M
Cornelissen
 
J
, et al. 
Conditioning intensity in middle-aged patients with AML in first CR: no advantage for myeloablative regimens irrespective of the risk group-an observational analysis by the Acute Leukemia Working Party of the EBMT.
Bone Marrow Transplant
2015
, vol. 
50
 
8
(pg. 
1063
-
1068
)
51
Brunet
 
S
Martino
 
R
Sierra
 
J
Hematopoietic transplantation for acute myeloid leukemia with internal tandem duplication of FLT3 gene (FLT3/ITD).
Curr Opin Oncol
2013
, vol. 
25
 
2
(pg. 
195
-
204
)
52
Pratcorona
 
M
Brunet
 
S
Nomdedéu
 
J
, et al. 
Favorable outcome of patients with acute myeloid leukemia harboring a low-allelic burden FLT3-ITD mutation and concomitant NPM1 mutation: relevance to post-remission therapy.
Blood
2013
, vol. 
121
 
14
(pg. 
2734
-
2738
)
53
Linch
 
DC
Hills
 
RK
Burnett
 
AK
Khwaja
 
A
Gale
 
RE
Impact of FLT3(ITD) mutant allele level on relapse risk in intermediate-risk acute myeloid leukemia.
Blood
2014
, vol. 
124
 
2
(pg. 
273
-
276
)
54
Schlenk
 
RF
Kayser
 
S
Bullinger
 
L
, et al. 
Differential impact of allelic ratio and insertion site in FLT3-ITD-positive AML with respect to allogeneic transplantation.
Blood
2014
, vol. 
124
 
23
(pg. 
3441
-
3449
)
55
Grimwade
 
D
Freeman
 
SD
Defining minimal residual disease in acute myeloid leukemia: which platforms are ready for “prime time”?
Blood
2014
, vol. 
124
 
23
(pg. 
3345
-
3355
)
56
Kayser
 
S
Walter
 
RB
Stock
 
W
Schlenk
 
RF
Minimal residual disease in acute myeloid leukemia–current status and future perspectives.
Curr Hematol Malig Rep
2015
, vol. 
10
 
2
(pg. 
132
-
144
)
57
Chen
 
X
Xie
 
H
Wood
 
BL
, et al. 
Relation of clinical response and minimal residual disease and their prognostic impact on outcome in acute myeloid leukemia.
J Clin Oncol
2015
, vol. 
33
 
11
(pg. 
1258
-
1264
)
58
Jourdan
 
E
Boissel
 
N
Chevret
 
S
, et al. 
Prospective evaluation of gene mutations and minimal residual disease in patients with core binding factor acute myeloid leukemia.
Blood
2013
, vol. 
121
 
12
(pg. 
2213
-
2223
)
59
Walter
 
RB
Buckley
 
SA
Pagel
 
JM
, et al. 
Significance of minimal residual disease before myeloablative allogeneic hematopoietic cell transplantation for AML in first and second complete remission.
Blood
2013
, vol. 
122
 
10
(pg. 
1813
-
1821
)
60
Boissel
 
N
Renneville
 
A
Leguay
 
T
, et al. 
Dasatinib in high-risk core binding factor acute myeloid leukemia in first complete remission: a French Acute Myeloid Leukemia Intergroup trial.
Haematologica
2015
, vol. 
100
 
6
(pg. 
780
-
785
)
61
Paschka
 
P
Schlenk
 
RF
Weber
 
D
, et al. 
Dasatinib (DAS) in combination with chemotherapy and as maintenance in core-binding factor (CBF) acute myeloid leukemia (AML): a phase Ib/IIa study of the German-Austrian AML Study Group (AMLSG) [abstract].
Haematologica
2015
, vol. 
100
 
s1
 
Abstract 515
62
Marcucci
 
G
Geyer
 
S
Zhao
 
W
, et al. 
Adding KIT inhibitor dasatinib (DAS) to chemotherapy overcomes the negative impact of KIT mutation/over-expression in core binding factor (CBF) acute myeloid leukemia (AML): results from CALGB 10801 (Alliance) [abstract].
Blood
2014
, vol. 
124
 
21
 
Abstract 8
63
Levis
 
M
Ravandi
 
F
Wang
 
ES
, et al. 
Results from a randomized trial of salvage chemotherapy followed by lestaurtinib for patients with FLT3 mutant AML in first relapse.
Blood
2011
, vol. 
117
 
12
(pg. 
3294
-
3301
)
64
Knapper
 
S
Hills
 
RK
Cavenagh
 
JD
, et al. 
A randomised comparison of the sequential addition of the FLT3 inhibitor lestaurtinib (CEP701) to standard first line chemotherapy for FLT3-mutated acute myeloid leukemia: The UK experience [abstract].
Blood
2014
, vol. 
124
 
21
 
Abstract 3736
65
Chen
 
YB
Shuli
 
L
Andrew
 
LA
, et al. 
Phase I trial of maintenance sorafenib after allogeneic hematopoietic stem cell transplantation for patients with FLT3-ITD AML [abstract].
Blood
2014
, vol. 
124
 
21
 
Abstract 671
66
Daver
 
N
Cortes
 
J
Ravandi
 
F
, et al. 
Secondary mutations as mediators of resistance to targeted therapy in leukemia.
Blood
2015
, vol. 
125
 
21
(pg. 
3236
-
3245
)
67
Burnett
 
AK
Hills
 
RK
Green
 
C
, et al. 
The impact on outcome of the addition of all-trans retinoic acid to intensive chemotherapy in younger patients with nonacute promyelocytic acute myeloid leukemia: overall results and results in genotypic subgroups defined by mutations in NPM1, FLT3, and CEBPA.
Blood
2010
, vol. 
115
 
5
(pg. 
948
-
956
)
68
Schlenk
 
RF
Döhner
 
K
Kneba
 
M
, et al. 
Gene mutations and response to treatment with all-trans retinoic acid in elderly patients with acute myeloid leukemia. Results from the AMLSG trial AML HD98B.
Haematologica
2009
, vol. 
94
 
1
(pg. 
54
-
60
)
69
Schlenk
 
RF
Döhner
 
K
Krauter
 
J
, et al. 
All-trans retinoic acid improves outcome in younger patients with nucleophosmin-1 mutated acute myeloid leukemia. Results of the AMLSG 07-04 randomized treatment trial [abstract].
Blood
2011
, vol. 
118
 
21
 
Abstract 80
70
El Hajj
 
H
Dassouki
 
Z
Berthier
 
C
, et al. 
Retinoic acid and arsenic trioxide trigger degradation of mutated NPM1, resulting in apoptosis of AML cells.
Blood
2015
, vol. 
125
 
22
(pg. 
3447
-
3454
)
71
Martelli
 
MP
Gionfriddo
 
I
Mezzasoma
 
F
, et al. 
Arsenic trioxide and all-trans retinoic acid target NPM1 mutant oncoprotein levels and induce apoptosis in NPM1-mutated AML cells.
Blood
2015
, vol. 
125
 
22
(pg. 
3455
-
3465
)
72
Jaglal
 
MV
Duong
 
VH
Bello
 
CM
, et al. 
Cladribine, cytarabine, filgrastim, and mitoxantrone (CLAG-M) compared to standard induction in acute myeloid leukemia from myelodysplastic syndrome after azanucleoside failure.
Leuk Res
2014
, vol. 
38
 
4
(pg. 
443
-
446
)
73
Stone
 
RM
Mazzola
 
E
Neuberg
 
D
, et al. 
Phase III open-label randomized study of cytarabine in combination with amonafide L-malate or daunorubicin as induction therapy for patients with secondary acute myeloid leukemia.
J Clin Oncol
2015
, vol. 
33
 
11
(pg. 
1252
-
1257
)
74
Genovese
 
G
Kähler
 
AK
Handsaker
 
RE
, et al. 
Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence.
N Engl J Med
2014
, vol. 
371
 
26
(pg. 
2477
-
2487
)
75
Jaiswal
 
S
Fontanillas
 
P
Flannick
 
J
, et al. 
Age-related clonal hematopoiesis associated with adverse outcomes.
N Engl J Med
2014
, vol. 
371
 
26
(pg. 
2488
-
2498
)
76
Becker
 
H
Marcucci
 
G
Maharry
 
K
, et al. 
Favorable prognostic impact of NPM1 mutations in older patients with cytogenetically normal de novo acute myeloid leukemia and associated gene- and microRNA-expression signatures: a Cancer and Leukemia Group B study.
J Clin Oncol
2010
, vol. 
28
 
4
(pg. 
596
-
604
)
77
Gardin
 
C
Chevret
 
S
Pautas
 
C
, et al. 
Superior long-term outcome with idarubicin compared with high-dose daunorubicin in patients with acute myeloid leukemia age 50 years and older.
J Clin Oncol
2013
, vol. 
31
 
3
(pg. 
321
-
327
)
78
Burnett
 
AK
Milligan
 
D
Prentice
 
AG
, et al. 
A comparison of low-dose cytarabine and hydoxyurea with or without all-trans retinoic acid for acute myeloid leukemia and high-risk myelodysplastic syndrome in patients not considered fit for intensive treatment.
Cancer
2007
, vol. 
109
 
6
(pg. 
1114
-
1124
)
79
Kantarjian
 
HM
Thomas
 
XG
Dmoszynska
 
A
, et al. 
Multicenter, randomized, open-label, phase III trial of decitabine versus patient choice, with physician advice, of either supportive care or low-dose cytarabine for the treatment of older patients with newly diagnosed acute myeloid leukemia.
J Clin Oncol
2012
, vol. 
30
 
21
(pg. 
2670
-
2677
)
80
Dombret
 
H
Seymour
 
JF
Butrym
 
A
, et al. 
International phase 3 study of azacitidine vs conventional care regimens in older patients with newly diagnosed AML with >30% blasts.
Blood
2015
, vol. 
126
 
3
(pg. 
291
-
299
)
81
Burnett
 
AK
Russell
 
NH
Culligan
 
D
, et al. 
The addition of the farnesyl transferase inhibitor, tipifarnib, to low dose cytarabine does not improve outcome for older patients with AML.
Br J Haematol
2012
, vol. 
158
 
4
(pg. 
519
-
522
)
82
Burnett
 
AK
Hills
 
RK
Hunter
 
A
, et al. 
The addition of arsenic trioxide to low-dose Ara-C in older patients with AML does not improve outcome.
Leukemia
2011
, vol. 
25
 
7
(pg. 
1122
-
1127
)
83
Burnett
 
AK
Hills
 
RK
Hunter
 
AE
, et al. 
The addition of gemtuzumab ozogamicin to low-dose Ara-C improves remission rate but does not significantly prolong survival in older patients with acute myeloid leukaemia: results from the LRF AML14 and NCRI AML16 pick-a-winner comparison.
Leukemia
2013
, vol. 
27
 
1
(pg. 
75
-
81
)
84
Dennis
 
M
Russell
 
N
Hills
 
RK
, et al. 
Vosaroxin and vosaroxin plus low-dose Ara-C (LDAC) vs low-dose Ara-C alone in older patients with acute myeloid leukemia.
Blood
2015
, vol. 
125
 
19
(pg. 
2923
-
2932
)
85
Burnett
 
AK
Russell
 
N
Hills
 
RK
, et al. 
A randomised comparison of the novel nucleoside analogue sapacitabine with low-dose cytarabine in older patients with acute myeloid leukaemia.
Leukemia
2015
, vol. 
29
 
6
(pg. 
1312
-
1319
)
86
Burnett
 
AK
Russell
 
NH
Hunter
 
AE
, et al. 
Clofarabine doubles the response rate in older patients with acute myeloid leukemia but does not improve survival.
Blood
2013
, vol. 
122
 
8
(pg. 
1384
-
1394
)
87
Döhner
 
H
Lübbert
 
M
Fiedler
 
W
, et al. 
Randomized, phase 2 trial of low-dose cytarabine with or without volasertib in AML patients not suitable for induction therapy.
Blood
2014
, vol. 
124
 
9
(pg. 
1426
-
1433
)
88
Naval
 
D
Kantarjian
 
HM
Garcia-Manero
 
G
, et al. 
Phase I/II study of vosaroxin and decitabine in newly diagnosed older patients (pts) with acute myeloid leukemia (AML) and high risk myelodysplastic syndrome (MDS) [abstract].
Blood
2014
, vol. 
124
 
21
 
Abstract 385
89
Breems
 
DA
Van Putten
 
WL
Huijgens
 
PC
, et al. 
Prognostic index for adult patients with acute myeloid leukemia in first relapse.
J Clin Oncol
2005
, vol. 
23
 
9
(pg. 
1969
-
1978
)
90
Burnett
 
AK
Goldstone
 
A
Hills
 
RK
, et al. 
Curability of patients with acute myeloid leukemia who did not undergo transplantation in first remission.
J Clin Oncol
2013
, vol. 
31
 
10
(pg. 
1293
-
1301
)
91
Ravandi
 
F
Ritchie
 
E
Sayar
 
H
, et al. 
Improved survival in patients with first relapsed or refractory acute myeloid leukemia (AML) treated with vosaroxin plus cytarabine versus placebo plus cytarabine: results of a phase 3 double-blind randomized controlled multinational study (VALOR) [abstract].
Blood
2014
, vol. 
124
 
21
 
Abstract LBA 6
92
O’Donnell
 
MR
Abboud
 
CN
Altman
 
J
, et al. 
Acute myeloid leukemia.
J Natl Compr Canc Netw
2012
, vol. 
10
 
8
(pg. 
984
-
1021
)
Sign in via your Institution