Remission induction with chemotherapy has long been the frontline treatment of acute myeloid leukemia (AML). However, intensive therapy is limited in frail patients by its associated toxicity and higher rates of failure or relapse in patients with chemoresistant disease, such as secondary AML or poor-risk cytogenetics. Frailty and chemoresistance are more frequent in older adults with AML. In recent years, epigenetic therapies with the hypomethylating agents decitabine and azacitidine have been thoroughly explored in AML. The results of two pivotal studies carried out with these agents in older adults with newly diagnosed AML have challenged the role of intensive chemotherapy as the frontline treatment option in this high-risk population. Here, we review the results of treatment with intensive chemotherapy and hypomethylating agents in older patients with AML; discuss the patient- and disease-specific criteria to integrate into treatment decision making; and also, highlight the methodological limitations of cross-study comparison in this population.

Learning Objectives
  • To learn the modalities, clinical results, and limitations of intensive chemotherapy and hypomethylating agents as frontline treatment in patients with AML

  • To learn the patient- and disease-specific criteria that must be integrated into the individualized therapeutic decision in high-risk AML

Currently, treatment decision making in patients with newly diagnosed acute myeloid leukemia (AML) is a stepwise process. Early diagnosis is critical for the minority of patients with acute promyelocytic leukemia, for whom long-term cure may be achieved using specific therapy based on retinoic acid and arsenic trioxide. The overall approach is that AML cytogenetics and genomics are then determined as precisely as possible to assess individual prognosis; orient risk-adapted therapeutic strategies, notably allogeneic hematopoietic stem cell transplantation (HSCT) in first remission; and enroll patients with specific molecular targets (FLT3 and IDH1/2 mutations) into dedicated clinical trials evaluating new targeted agents.1  Patients’ general health status is also concomitantly evaluated, and potential comorbidities are explored for each patient to adjust treatment intensity to the patient’s ability to cope with treatment-associated toxicities dominated by myelosuppression and cardiotoxicity. In older patients, a population that is generally defined as patients older than 60 to 65 years old, estimating benefit/risk associated with treatments of various intensities is of utmost importance.

The poor prognosis of older adults with AML is an intricate consequence of patient-related factors (such as comorbidities) and disease-related chemoresistance (notably because of more frequent secondary AML), both of which can be seen, albeit at lower frequencies, in adults below the age of 60 years old. In patients over the age of 60 years old, except in those with very proliferative disease, delaying AML treatment does not seem to affect prognosis.2,3  In older patients, such as in those with relapsed/refractory (R/R) AML, the choice between an intensive chemotherapy (ICT) regimen or a lower-intensity treatment is crucial, because these two options are not associated with equivalent outcomes, at least in the short term. To date, lower-intensity AML therapy is mostly represented by epigenetic therapy, initially developed for patients with higher-risk myelodysplastic syndrome (MDS). Both hypomethylating agents (HMAs) decitabine (DAC) and azacitidine (AZA) have shown their ability to improve median and short-term overall survival (OS) compared with low-dose cytarabine (LDAC) or best supportive care (BSC) only, even if this has not translated into improved cure rates. In this review, we will discuss how and when to decide between HMAs and ICT, focusing on older patients with AML.

In older patients with AML, standard ICT allows 45% to 65% of older adults with newly diagnosed AML to achieve a complete remission (CR). Overall, because of worse tolerance of ICT and higher intrinsic AML resistance, median OS does not usually exceed 12 to 18 months, and 5-year OS remains <15%. Standard induction chemotherapy is a combination of intermediate-dose cytarabine and an anthracycline administered for 7 and 3 days (7 + 3), respectively. Cytarabine is given as continuous IV infusion at the daily dose of 100 or 200 mg/m2 per day. The anthracycline can be either daunorubicin or idarubicin, usually given at 60 and 12 mg/m2 per day, respectively, with a reduced dose of 45 mg/m2 per day daunorubicin being considered suboptimal.4  It has been reported that idarubicin might be associated with more rapid CR achievement and a higher cure rate in the long term.5  Despite progress made in terms of supportive measures, notably in antibiotic and antifungal therapy, early mortality remains high. Early death in induction occurs before response evaluation in ∼10% to 20% of patients selected to receive ICT, and the 60-day mortality rate ranges from 15% to 30%. After CR has been achieved, it is acknowledged that, in patients capable of tolerating additional chemotherapy, it is preferable to administer some form of postremission therapy, although there is no standard consolidation or maintenance regimen.

Prospective randomized studies comparing different postremission chemotherapy regimens are sparse. In the Cancer and Leukemia Group B (CALGB) 8525 Study, which compared three different dose-intensity schedules of cytarabine, older patients did not benefit from dose intensification.6  In the CALGB 8923 Study, addition of two intensive consolidation courses with intermediate-dose cytarabine and mitoxantrone yielded similar survival as four LDAC courses.7  Moreover, in the Acute Leukemia French Association (ALFA) 9803 Study, six outpatient mild-intensity postremission courses yielded superior survival compared with one intensive consolidation course.8  In addition, no survival advantage was observed in patients ages 65 to 70 years old who received two intensive consolidation courses rather than one single intensive consolidation course in the concomitant ALFA 9801 Trial.9  Finally, in the German AML Intergroup Study, which randomized older patients between one common standard arm and two other chemotherapy strategies used by distinct German cooperative groups, no differences in relapse-free and OS were observed.10  As in younger adults, allogeneic HSCT is considered to be probably the best option to prevent AML recurrence in selected patients with intermediate- or adverse-risk AML, even if no formal evidence for this assertion has been provided to date. While awaiting the results of an ongoing European randomized trial (NCT00766779) that is prospectively comparing reduced-intensity conditioning (RIC) transplantation with additional chemotherapy, with OS as the primary end point, HSCT is offered to an increasing proportion of older patients in first remission. The best therapy before HSCT in this population has been an open issue for years.

Drugs that could potentially improve outcome over current ICT in the near future include the antibody-drug conjugate (ADC) gemtuzumab ozogamicin and the liposomal CPX-351 chemotherapy as well as FLT3 and IDH1/2 inhibitors.11,12  A recent French study also suggested that the addition of norethandrolone during postremission chemotherapy may improve long-term outcome.13  The rationale for this latter study is based on the potential ability of androgens to block proliferation and induce differentiation of AML cells.

Genetic factors with prognostic impact

In intensively treated AML patients, pretreatment prognostic factors are usually separated into two groups: AML-related genetic factors governing the risk of AML resistance or recurrence and patient-related factors governing the risk of treatment-associated toxicity and mortality. The former are much better standardized than the latter. As summarized in the recently updated recommendations of the European LeukemiaNet panel,1  cytogenetics and molecular genetics allow classification of AML cases into the three categories: favorable, intermediate, and adverse risk (Table 1). Among the adverse-risk subset, particularly worrisome are AML with a monosomal karyotype, TP53 gene mutation, and abnormalities involving the MECOM (EVI1) gene. Importantly, the better outcome associated with entities considered as favorable per European LeukemiaNet criteria (Table 1) has been validated in older patients treated intensively and translates into higher cure rates.5,14,15  More recently, the complexity and prognostic significance of individual gene mutation patterns as revealed by next generation sequencing has been specifically explored in older patients. Early studies confirmed the favorable impact of NPM1 mutations. However, the presence of FLT3 internal tandem duplication and TP53 mutations was associated with shorter OS. Interestingly, some driver gene mutations, such as DNA-methyltransferase 3A (DNMT3A), TET2, SRSF2, and ASXL1, may be detected in seemingly healthy patients before overt AML development.16,17  Such preexisting, MDS-like18  clonal hematopoiesis is more frequent in older patients, and the persistence of preleukemic clones during CR seems to represent a poor prognostic factor associated with higher relapse incidence in these patients.19  A mutation signature mostly including spliceosome and cohesion genes has been proposed to define secondary AML. Although this signature does not impart poor prognosis compared with de novo AML,20  additional work is required to determine whether these molecularly defined secondary AML patients benefit from epigenetic therapy. In a sense, these novel observations enlarge the field of AML secondary to a prior MDS, a setting reported as being of poor prognosis for years. Finally, a so-called leukemic stem cell gene expression signature, which is more frequently observed in AML with an adverse karyotype but can also be seen in the intermediate-risk subset, has been recently shown to be associated with poor outcomes.21 

Table 1.

European LeukemiaNet risk stratification by genetics

Risk categoryGenetic abnormality
Favorable t(8,21)(q22,q22.1), RUNX1-RUNX1T1 
Favorable inv(16)(p13.1q22) or t(16,16)(p13.1,q22), CBFB-MYH11 
Favorable Mutated NPM1 without FLT3-ITD or with FLT3-ITDlow* 
Favorable Biallelic mutated CEBPA 
Intermediate Mutated NPM1 and FLT3-ITDhigh* 
Intermediate Wild-type NPM1 without FLT3-ITD or with FLT3-ITDlow* (without adverse-risk genetic lesions) 
Intermediate t(9,11)(p21.3,q23.3), MLLT3-KMT2A 
Intermediate Cytogenetic abnormalities not classified as favorable or adverse 
Adverse t(6,9)(p23,q34.1), DEK-NUP214 
Adverse t(v,11q23.3), KMT2A rearranged 
Adverse t(9,22)(q34.1,q11.2), BCR-ABL1 
Adverse inv(3)(q21.3q26.2) or t(3,3)(q21.3,q26.2), GATA2, MECOM(EVI1) 
Adverse −5 or del(5q), −7, −17/abn(17p) 
Adverse Complex karyotype, monosomal karyotype§ 
Adverse Wild-type NPM1 and FLT3-ITDhigh(a) 
Adverse Mutated RUNX1 
Adverse Mutated ASXL1 
Adverse Mutated TP53 
Risk categoryGenetic abnormality
Favorable t(8,21)(q22,q22.1), RUNX1-RUNX1T1 
Favorable inv(16)(p13.1q22) or t(16,16)(p13.1,q22), CBFB-MYH11 
Favorable Mutated NPM1 without FLT3-ITD or with FLT3-ITDlow* 
Favorable Biallelic mutated CEBPA 
Intermediate Mutated NPM1 and FLT3-ITDhigh* 
Intermediate Wild-type NPM1 without FLT3-ITD or with FLT3-ITDlow* (without adverse-risk genetic lesions) 
Intermediate t(9,11)(p21.3,q23.3), MLLT3-KMT2A 
Intermediate Cytogenetic abnormalities not classified as favorable or adverse 
Adverse t(6,9)(p23,q34.1), DEK-NUP214 
Adverse t(v,11q23.3), KMT2A rearranged 
Adverse t(9,22)(q34.1,q11.2), BCR-ABL1 
Adverse inv(3)(q21.3q26.2) or t(3,3)(q21.3,q26.2), GATA2, MECOM(EVI1) 
Adverse −5 or del(5q), −7, −17/abn(17p) 
Adverse Complex karyotype, monosomal karyotype§ 
Adverse Wild-type NPM1 and FLT3-ITDhigh(a) 
Adverse Mutated RUNX1 
Adverse Mutated ASXL1 
Adverse Mutated TP53 

ITD, internal tandem duplication.

*

Low indicates low allelic ratio (<0.5), high indicates high allelic ratio (>0.5), and semiquantitative assessment of FLT3-ITD allelic ratio (using DNA fragment analysis) is determined as the ratio of the area under the curve FLT3-ITD divided by the area under the curve FLT3-wild type.

The presence of t(9,11)(p21.3,q23.3) takes precedence over rare, concurrent adverse-risk gene mutations.

Three or more unrelated chromosome abnormalities in the absence of one of the WHO-designated recurring translocations or inversions [ie, t(8,21), inv(16); t(16,16), t(9,11), t(v,11)(v,q23.3), t(6,9), inv(3); or t(3,3), AML with BCR-ABL1].

§

Defined by the presence of one single monosomy (excluding loss of X or Y) in association with at least one additional monosomy or structural chromosome abnormality (excluding core binding factor AML).

These markers should not be used as an adverse prognostic marker if they cooccur with favorable-risk AML subtypes.

TP53 mutations are significantly associated with AML with complex and monosomal karyotypes.

Assessment of eligibility for standard induction therapy

Except for very old patients over the age of 80 to 85 years old, chronologic age alone is not a good criterion for fitness assessment. Beyond subjective physical function scales, such as Eastern Cooperative Oncology Group (ECOG) performance status (PS), there is, however, no consensus definition of patients unfit for intensive therapy. Although PS is a good tool to detect frail patients, it does not detect subclinical impairments that could make a patient more vulnerable to therapy.22  Interestingly, poor PS is significantly associated with higher white blood cell count (WBC) at diagnosis, suggesting that PS at diagnosis captures both the patients’ durable intrinsic frailty and transient disease–related vulnerability. Geriatric assessment may be of help but is limited to studies of small sample size to date, maybe because of the very small proportion of patients over the age of 80 to 85 years old in most current older AML intensive trials. In current practice, eligibility for ICT is mostly defined in clinical trials on the basis of PS and specific markers for cardiac, renal, and liver functions, which incidentally, do not always represent formal contraindications for ICT.23  Comorbidity burden measurement using, for instance, the Charlson Comorbidity Index or the Hematopoietic Cell Transplantation Comorbidity Index24  tends to be more frequently prospectively evaluated. Several scores mixing disease-related and patient-related factors have been developed but are rarely used prospectively.25  In the real-life setting (ie, outside of clinical trials), treatment intensity mostly relies on the physician’s subjective assessment of a patient’s vulnerability and expectations, which may frequently differ from the patient’s viewpoint.26  A direct consequence of this lack of standardization is the extreme difficulty of capturing the extent to which a trial’s study population has been truly selected and thus, comparing one study with another. This remains a major concern for evidence-based treatment decision-making recommendations between ICT and lower-intensity therapy in AML patients.

The role of epigenetic deregulation in the pathogenesis of AML has been a matter of intense research during recent years. As mentioned above, recurrent somatic mutations in key genes involved in the epigenetic machinery (DNMT3A, TET2, IDH1, IDH2, and ASXL1), also present in MDS, have been identified in AML and preleukemic clones. Although aberrant methylation in AML goes beyond the role of these mutations,27  therapies targeting DNMTs have been investigated in MDS and AML. Both DAC and AZA are pyrimidine analogs acting as DNMT inhibitors, leading to global hypomethylation of cytosine residues at not only cytosine guanine dinucleotide–rich gene promoters but also, distal enhancers critical for gene expression regulation.28  More recently, an oral formulation of AZA (CC-486) and a second generation HMA, guadecitabine (SGI-110),29  have entered clinical development.

Prospective studies evaluating DAC or AZA in AML patients are summarized in Table 2.30-38  These studies focused on either World Health Organization (WHO) AML patients or MDS patients, including WHO AML patients with 20% to 30% bone marrow (BM) blasts. In these studies, AZA was generally administered subcutaneously according to the MDS schedule (75 mg/m2 per day for 7 days per cycle), whereas DAC was administered IV according to different dose schedules. Initially, DAC was given at 15 mg/m2 per 8 hours IV for 3 days every 6 weeks.35,37  An alternative outpatient dose schedule (20 mg/m2 per day for 5 days every 4 weeks) was also developed.32,36  An even more prolonged 10-day administration schedule has been proposed as potentially associated with a higher response rate.33  With the exception of the most recent AZA-AML-001 Study phase 3 trial, AML studies recruited patients considered unsuitable for standard intensive AML therapy. Median ages were thus relatively high, ranging from 65 to 75 years old. With HMAs, the best hematologic responses are frequently reached after a median of two to four cycles and may even be observed later with continuous therapy in patients classified early with only stable disease. Continuous HMA administration is thus recommended until disease progression or limiting toxicities, the latter being relatively rare, because these agents are, in general, well tolerated. Such a strategy is clearly at odds with standard ICT. This potential ability of HMAs to prolong survival, despite the lack of remission achievement through the maintenance of stable diseases and/or improvement of cytopenias, represents a novel paradigm in AML therapy.39,40  These differences in HMA vs chemotherapy explain why OS must remain the primary end point when trying to compare the two strategies, although OS might be influenced by subsequent uncontrolled crossovers, HSCT, or enrolment in other investigational trials.

Table 2.

Prospective DAC or AZA studies in AML patients

ReferenceAML typeMedian age (range), yDrug dose and schedulePatients, NCR/PR (%)ORR* (%)Median OS, mo
31         
 CALGB 8421 AML (20%-30% BM blasts) 65 (35-81) AZA 75 mg/m2 per day for 7 d, IV 25 4 (12) 12 (48) — 
 CALG 8921 AML (20%-30% BM blasts) 66 (23-82) AZA 75 mg/m2 per day for 7 d, SC 26 3 (12) 9 (35) — 
 CALGB 9221 AML (20%-30% BM blasts) 69 (1-82) Two randomization arms     
AZA 75 mg/m2 per day for 7 d, SC 27 2 (7) 10 (37%) 19.3 
BSC 25 2 (8) 12.9 
32  WHO-AML 74 (61-87) DAC 20 mg/m2 per day for 5 d, IV 55 13 (24) 14 (25) 7.7 
33  WHO-AML 74 (60-85) DAC 20 mg/m2 per day for 10 d, IV 53 25 (47) 34 (64) 12.6 
34  AML (20%-30% BM blasts) 70 (62-80) Two randomization arms     
AZA 75 mg/m2 per day for 7 d, SC 55 10 (18) — 24.5 
BSC, LDAC, or ICT 58 9 (16) — 16 
35  WHO-AML 72 (56-86) DAC 15 mg/m2 per 8 hours for 3 d, IV 227 59 (26)§ — 5.5 
36  WHO-AML 73 (64-91) Two randomization arms     
DAC 20 mg/m2 per day for 5 d, IV 242 44 (18) 73 (30) 7.7 
BSC or LDAC 243 27 (11) 34 (14) 
37  AML (20%-30% BM blasts) 69.5 (61-90) Two randomization arms     
DAC 15 mg/m2 per 8 hours for 3 d, IV 40 6 (15) 12 (30) 
BSC 35 
38  WHO-AML 75 (64-91) Two randomization arms     
AZA, 75 mg/m2 per day for 7 d, SC 231 50 (22) 70 (30) 10.4 
BSC, LDAC, or ICT 247 57 (23) 65 (26) 6.5 
ReferenceAML typeMedian age (range), yDrug dose and schedulePatients, NCR/PR (%)ORR* (%)Median OS, mo
31         
 CALGB 8421 AML (20%-30% BM blasts) 65 (35-81) AZA 75 mg/m2 per day for 7 d, IV 25 4 (12) 12 (48) — 
 CALG 8921 AML (20%-30% BM blasts) 66 (23-82) AZA 75 mg/m2 per day for 7 d, SC 26 3 (12) 9 (35) — 
 CALGB 9221 AML (20%-30% BM blasts) 69 (1-82) Two randomization arms     
AZA 75 mg/m2 per day for 7 d, SC 27 2 (7) 10 (37%) 19.3 
BSC 25 2 (8) 12.9 
32  WHO-AML 74 (61-87) DAC 20 mg/m2 per day for 5 d, IV 55 13 (24) 14 (25) 7.7 
33  WHO-AML 74 (60-85) DAC 20 mg/m2 per day for 10 d, IV 53 25 (47) 34 (64) 12.6 
34  AML (20%-30% BM blasts) 70 (62-80) Two randomization arms     
AZA 75 mg/m2 per day for 7 d, SC 55 10 (18) — 24.5 
BSC, LDAC, or ICT 58 9 (16) — 16 
35  WHO-AML 72 (56-86) DAC 15 mg/m2 per 8 hours for 3 d, IV 227 59 (26)§ — 5.5 
36  WHO-AML 73 (64-91) Two randomization arms     
DAC 20 mg/m2 per day for 5 d, IV 242 44 (18) 73 (30) 7.7 
BSC or LDAC 243 27 (11) 34 (14) 
37  AML (20%-30% BM blasts) 69.5 (61-90) Two randomization arms     
DAC 15 mg/m2 per 8 hours for 3 d, IV 40 6 (15) 12 (30) 
BSC 35 
38  WHO-AML 75 (64-91) Two randomization arms     
AZA, 75 mg/m2 per day for 7 d, SC 231 50 (22) 70 (30) 10.4 
BSC, LDAC, or ICT 247 57 (23) 65 (26) 6.5 

ORR, overall response rate; PR, partial remission; SC, subcutaneous.

*

ORR including CR/PR, CR with incomplete blood count recovery, and/or hematologic improvements.

Thirteen patients received study drug after crossover.

CR only.

§

PR criteria were used in this study, and it included patients with persistent cytopenia.

If >30% marrow blasts and WBC < 15 G/L.

To date, the two largest HMA studies are randomized phase 3 studies of HMA vs patient/physician choice among various conventional care regimen (CCR) options.36,38  The DACO-16 Study compared DAC with LDAC or BSC, whereas the AZA-AML-001 Study compared AZA with LDAC, BSC, or ICT. Eligibility criteria were not identical in the two studies, with more proliferative disease allowed in the DAC trial.41  In addition, the LDAC dose schedule differed: 20 mg/m2 per day for 10 days every 4 weeks in the DAC study and 20 mg twice per day for 10 days every 4 weeks in the AZA study. Both studies showed significant improvement in median OS in the HMA arm (7.7 months with DAC and 10.4 months with AZA) compared with the CCR arm (5.0 months in the DAC study and 6.5 months in the AZA study). However, because of the convergence of survival curves in the longer term, cure rates were not improved by HMA therapy. Rates of CR and CR + CR with incomplete blood count recovery were 15.7% and 25.6% with DAC, respectively; 19.5% and 27.8% with AZA, respectively; 7.9% and 10.7% with LDAC in the DACO-16 Study, respectively; and 24.1% and 25.9% with LDAC in the AZA-AML-001 Study, respectively. Because these trials were designed to compare HMA with CCR as a whole, it is difficult to extrapolate the results in any specific CCR subgroup analysis. It seemed, nevertheless, that clinically meaningful gains in median OS were still observed when comparing either DAC or AZA with LDAC. In addition, in the AZA-AML-001 Study, AZA therapy was associated with lower incidence rates of treatment-emergent anemia, neutropenia, febrile neutropenia, and thrombocytopenia compared with LDAC.38  Real-life studies of AZA therapy in patients with previously untreated or R/R AML are summarized in Table 3.42-51  Overall, these real-life data, collected through national/regional registries or compassionate use programs, confirm that the outcome described in AZA trials could be reproduced in fewer selected patients and across different AML subtypes.

Table 3.

Real-life studies with AZA in AML patients

ReferenceDrugPatients, NAML typePrior AML therapyMedian age (range), yMedian OS, mo
42  AZA 54 Post-MPN AML (48%) or MDS None 69.5 (37-89) 8* 
43  AZA 82 Any WHO-AML subtype 57% 72 (29-87) 
44  AZA ± VPA or ATRA 149 Any WHO-AML subtype None 74 (31-91) 9.4 
45  AZA 302 Any WHO-AML subtype 54% 73 (30-93) 9.6 
46  AZA 95 Any WHO-AML subtype None 76 (71-81) 11.3 
47        
 Training set AZA 110 Any WHO-AML subtype None 75 (56-89) 8.1 
 Validation set AZA 261 Any WHO-AML subtype None 75 (31-93) 10.0 
48  AZA 95 WHO-AML None 77 (23-93) 10.8 
49  AZA 107 Post-MDS, post-MPN, and therapy-related AML None 76.5 (71-81) 10.8 
50  AZA 47 Any WHO-AML subtype All R/R AML 63 (29-79) 
51  AZA ± VPA or ATRA 130 Any WHO-AML subtype All R/R AML 67 (50-80) 8.4 
ReferenceDrugPatients, NAML typePrior AML therapyMedian age (range), yMedian OS, mo
42  AZA 54 Post-MPN AML (48%) or MDS None 69.5 (37-89) 8* 
43  AZA 82 Any WHO-AML subtype 57% 72 (29-87) 
44  AZA ± VPA or ATRA 149 Any WHO-AML subtype None 74 (31-91) 9.4 
45  AZA 302 Any WHO-AML subtype 54% 73 (30-93) 9.6 
46  AZA 95 Any WHO-AML subtype None 76 (71-81) 11.3 
47        
 Training set AZA 110 Any WHO-AML subtype None 75 (56-89) 8.1 
 Validation set AZA 261 Any WHO-AML subtype None 75 (31-93) 10.0 
48  AZA 95 WHO-AML None 77 (23-93) 10.8 
49  AZA 107 Post-MDS, post-MPN, and therapy-related AML None 76.5 (71-81) 10.8 
50  AZA 47 Any WHO-AML subtype All R/R AML 63 (29-79) 
51  AZA ± VPA or ATRA 130 Any WHO-AML subtype All R/R AML 67 (50-80) 8.4 

ATRA, all-trans retinoic acid; MPN, myeloproliferative neoplasm; VPA, valproic acid.

*

In patients fulfilling WHO-AML diagnostic criteria.

With >30% marrow blasts and WBC < 15 G/L (AZA-AML-001 Study entry criteria).

Clinical parameters as predictors of HMA response

Significant HMA activity has been observed across most AML subgroups, including those with intermediate- or adverse-risk cytogenetics, >30% or 50% BM blasts, post-MDS AML, or AML with myelodysplastic-related changes as defined by the WHO. Although adverse-risk genetics remains a poor prognostic factor in patients receiving frontline HMA therapy,42,45-48  greater benefits have been associated with AZA therapy in patients with poor-prognostic karyotypes and/or WHO myelodysplastic-related changes AML in the comparative AZA-AML-001 Study (John F. Seymour, Hartmut Döhner, Aleksandra Butrym, et al, manuscript submitted, 2017).52  AZA has also been reported as a relevant frontline option in patients with AML or MDS with chromosome 3q abnormalities.53  Probably because of preferential selection of older patients, the prognostic impact of age in HMA studies remains unclear. Age was not significantly associated with OS in a multivariate analysis of outcomes in some AZA real-life cohorts44,45  but not in all.46  Age was, however, significantly associated with OS of the whole population of the DAC vs treatment choice trial54  but not in the AZA-AML-001 Study.38  As with intensive therapy of AML, an ECOG PS higher than two remains of strong prognostic value when older AML patients are treated front line with HMAs.44,45,47,48  The prognostic value of WBC at diagnosis remains uncertain but was associated with worse long-term outcome in the DACO-16 Study and most real-life AZA reports. Because of their mechanisms of action, HMAs are generally not considered as a good frontline option in high WBC patients with rapidly proliferative AML.44-47,51,54  Based on previous observations by Gardin et al,55  15 × 109/L was retained as the upper WBC limit for eligibility in the AZA-AML-001 Study.

Genetic predictors of HMA response

The precise mechanisms of action of HMAs remain unclear at both the molecular level, where reexpression of tumor suppressor genes on HMA exposure has long been sought without success,56  and the cellular level, where it is increasingly apparent that HMAs do not allow clonal eradication,57  although transient clonal reduction has been reported.58  As a consequence, validated biomarkers of HMA activity are yet to emerge. A number of reports have investigated the predictive and prognostic role of gene mutations in AML and MDS treated with HMAs. Results were limited by the small size of most cohorts and their retrospective nature. Convergent results nevertheless suggest that mutations in epigenetic regulators, such as TET2 or DNMT3A, could predict a superior response rate, although this does not translate into a survival benefit.59-62  More recently, in a series of 116 patients with AML or MDS treated with 10-day courses of DAC, higher response rates were observed in patients with adverse-risk cytogenetics and/or TP53 gene mutations compared with other patients. This improved response rate also did not translate into a survival benefit.58  The favorable role of TP53 mutations has not been observed in other cohorts of AML patients treated with HMA at conventional and thus, less intensive schedules.63  These observations raise the intriguing possibility that specific HMAs and schedules may differently affect leukemic clones and warrant additional prospective validation given the poor prognosis of this patient population, even after allogeneic transplant. Finally, in the AZA-AML-001 Study, patients with TP53 and/or NRAS gene mutation treated with AZA had longer OS than similar patients treated with CCR, whereas OS was similar in both the AZA and CCR arms for patients with a mutation in any of the DNMT3A, IDH1/2, or TET2 genes.64  In the same study, FLT3 gene mutations had a negative impact on OS in patients treated with AZA but did not have a negative impact on those in the CCR arm.

Methylation changes as a predictor of HMA response

Global hypomethylation has been consistently reported after in vitro and in vivo exposure to HMAs in MDS or AML,65,66  and some reports have suggested more profound hypomethylation in responders.29,57  Several studies have reported reversal of methylation or gene reexpression at specific loci on HMA treatment33,67,68 ; however, few reports have identified patterns of methylation at baseline that could predict response and/or survival of patients treated with HMA.69,70  This likely reflects the technical limitations of older methylome strategies focusing on gene promoter cytosine guanine dinucleotide islands69  as well the intratumoral heterogeneity of methylation patterns in AML.71  The heterogeneity of patients analyzed and the early analysis of posttreatment methylation changes, not to mention the uncertainties regarding the most relevant mechanisms of actions of these drugs in AML, may also explain the limited role of methylation studies in predicting HMA activity. Authors have also evaluated the expression or polymorphisms in genes involved in the metabolism of HMA, including the transporter hENT1, or the enzymes uridine cytidine kinase, deoxycytidine kinase, and cytidine deaminase, without reproducible results.69,72  Taken together, identification of relevant biomarkers guiding the allocation of HMA in AML will require a better understanding of the target cells and the mechanism of action of these drugs that can also act in a noncellular intrinsic manner on the immune response.

Given the significant unmet need, current developments of almost all new antileukemic agents include combination trials with AZA, DAC, or even LDAC in patients with newly diagnosed or R/R AML deemed unfit for ICT. Early results have been reported with agents, such as the kinase inhibitors sorafenib and midostaurin, BCL2 inhibitor venetoclax, MDM2 inhibitor idasanutlin, or SMO inhibitor glasdegib, or even with chemotherapeutic agents or ADCs, such as vosaroxin, gemtuzumab ozogamicin, or vadastuximab talirine. Combining HMAs with histone deacetylase inhibitors, another class of epigenetic drugs, has yielded disappointing results to date. Studies with new antibodies, ADCs, immune checkpoint inhibitors, or other epigenetic agents similar to IDH inhibitors are ongoing. Unfortunately, the efficacy of such combinations is too frequently hindered by their poor tolerance in older unfit patients, raising the issue of their evaluation in fitter patients, potentially against ICT.

The two treatment options described above, ICT and epigenetic therapy, are very different by nature. Intensive therapy systematically induces transient but prolonged myelosuppression requiring hospitalization because of worsening neutropenia and thrombocytopenia, with a risk of infections and/or hemorrhages that may be associated with a higher early mortality rate. However, the CR rate is higher after ICT than after HMA therapy, potentially offering a better quality of life to those patients who may reach CR, at least until eventual AML recurrence. Epigenetic therapy is less toxic, may be administered in the outpatient setting, and might thus be associated with a better quality of life during therapy and lower early mortality rate. The CR rate is significantly lower than after ICT, but HMAs seem to be capable of maintaining stable diseases or incomplete responses, leading to a median OS that might eventually be similar to that observed after ICT.

Summarizing this, HMAs could represent an option retained by default in patients “unlikely to benefit from ICT” as defined by their physicians through a global evaluation based on age, ECOG PS, fitness assessment, history of prior hematologic disorders or chemotherapy, WBC, AML genetic feature, comorbidities, and polypharmacy. This is, in fact, what happens in real life. According to real-life reports and one recent population-based study from the United States,46,49,55,73-75  it can be estimated that approximately one-half of all older patients treated for AML receive HMA, whereas one-half receive ICT, with variations in proportions according to countries and centers. In these reports, patient characteristics differed markedly between the HMA and ICT cohorts, underlying major and reproducible selection biases. Patients treated with HMAs were significantly older, had fewer comorbid diseases, had more frequent secondary AML, had lower WBC and/or BM blast percentage at diagnosis, and had higher rates of adverse-risk karyotypes. Resulting observations were similar early mortality rates in HMA vs ICT cohorts46,55,73,74  and longer OS in ICT vs HMA in some cohorts,46,73  but these retrospective comparisons were clearly confounded by patient selection issues. To address this problem, two French groups performed propensity score matching analyses, which identified 63 and 66 pairs of higher-risk patients, in whom OS was ultimately similar after ICT or AZA therapy.46,55  To date, no large randomized studies have been initiated aiming to compare epigenetic therapy with ICT in older AML patients, and any such study would probably face major recruitment and/or selection problems. ICT was offered as a CCR option in the randomized phase 3 AZA-AML-001 Study. However, most patients in the CCR arm received LDAC, and only a minority of the whole study population was randomized between AZA and ICT. There were thus not enough patients to make any definitive statements about the relative benefit of AZA and chemotherapy in patients who were deemed ICT candidates. The lower than expected rate of patients randomized against ICT in the study (18% when 50% could have been anticipated) shows how the experimental AZA arm skewed the study population in this trial, which does not encompass the full spectrum of older patients with AML.

In conclusion, there is definitely a subgroup of poor-risk AML patients in whom epigenetic therapy may yield similar or maybe even better outcomes than chemotherapy. This subgroup cannot be readily defined today by universal standardized criteria, because final decisions are highly individualized, taking into account balanced benefit/risk as well as subjective assessments, not to mention the patients’ opinion. Factors that could be used to guide the individual treatment decision are summarized in Table 4.

Table 4.

Deciding between chemotherapy and epigenetic therapy in patients with AML

Factors in favor of ICTFactors in favor of epigenetic therapy
Age <70 y Age ≥80 y 
ECOG PS < 2 ECOG PS ≥ 2 
Low CCI, HCT-CI (<3) High CCI, HCT-CI (≥3) 
Higher WBC* Lower WBC* 
De novo AML Secondary AML (post-MDS, post-MPN) 
AML not classified as WHO MRC-AML AML classified as WHO MRC-AML 
Favorable genetics: favorable ELN category (Table 1Unfavorable genetics: monosomy 5 or 7, del(5q); complex, monosomal karyotype; TP53 gene mutation 
FLT3-ITD No FLT3-ITD 
No epigenetic gene mutation? Epigenetic gene mutations (TET2, DNMT3A)? 
Factors in favor of ICTFactors in favor of epigenetic therapy
Age <70 y Age ≥80 y 
ECOG PS < 2 ECOG PS ≥ 2 
Low CCI, HCT-CI (<3) High CCI, HCT-CI (≥3) 
Higher WBC* Lower WBC* 
De novo AML Secondary AML (post-MDS, post-MPN) 
AML not classified as WHO MRC-AML AML classified as WHO MRC-AML 
Favorable genetics: favorable ELN category (Table 1Unfavorable genetics: monosomy 5 or 7, del(5q); complex, monosomal karyotype; TP53 gene mutation 
FLT3-ITD No FLT3-ITD 
No epigenetic gene mutation? Epigenetic gene mutations (TET2, DNMT3A)? 

CCI, Charlson Comorbidity Index; ELN, European LeukemiaNet; HCT-CI, Hematopoietic Cell Transplantation Comorbidity Index; ITD, internal tandem duplication; MPN, myeloproliferative neoplasm; MRC, myelodysplastic-related change.

*

Higher WBC is associated with a worse prognosis after ICT; nevertheless, the prognosis of patients with WBC > 15 000 G9/L might be even worse after HMA therapy, at least when treated with AZA55  and maybe less when treated with DAC.41 

High WBC AML

How to manage older AML patients with high WBC at diagnosis remains an open issue, because these patients appear to poorly respond to both chemotherapy and HMAs. Because they frequently present activating kinase mutations, such as FLT3 internal tandem duplications, combination with kinase inhibitors represents an interesting approach.

Secondary AML in patients previously treated with HMAs

A proportion of higher-risk MDS patients failing HMA quickly progress to secondary AML. The outcome of higher-risk MDS patients after failure of HMAs is dismal, with a median OS around 3 months,76  and there are no approved second-line regimens. Notably, ICT77  or administration of other first generation HMAs seems of limited efficacy in this context,78  although newer HMAs, such as guadecitabine, seem to retain some activity in this setting.29  Of note, guadecitabine is currently prospectively under evaluation vs treatment of choice (LDAC, AZA, and DAC) in the frontline setting (NCT02920008).

Allogeneic HSCT after HMA vs ICT

Several retrospective studies have reported comparable outcome of higher-risk MDS patients who received allogeneic HSCT after ICT or HMA.79  These reports were likely biased because of their retrospective nature. A recent prospective Italian trial showed the feasibility of HSCT in the majority (74%) of higher-risk MDS or AML patients with a donor who were treated with AZA.80  This figure compares favorably with a previous prospective report with ICT as a frontline option, where only a minority (7%) of patients ultimately received HSCT,81  although the latter included patients with much higher-risk disease, and both RIC availability and supportive care have made significant progress in the decade separating these two reports. The European Organization for Research and Treatment of Cancer (EORTC) is currently conducting a prospective randomized trial comparing 10-day DAC with ICT before allogeneic HSCT in patients ages 60 years old or older (NCT02172872). According to the current dogma that lowering leukemic burden is of tantamount importance before HSCT, especially with RIC, one can expect that the higher CR rate associated with ICT is to be preferred to HMAs in this context. However, there is, to date, no formal demonstration of this concept in AML. Furthermore, although ICT and HMAs differ by their CR rate in the global AML population, this may not be the case in all disease categories, especially in patients with poor-risk cytogenetics. Finally, not only the CR rate but also, the quality of these CRs are also likely to impact outcome after HSCT. Indeed, clonal remissions are frequent in this patient population after ICT82  and may obviate prognosis after CR. Measurable residual disease as assessed by flow cytometry relying on Leukemia Aberrant Immunophenotype or leukemic stem cell markers83,84  may also help compare these two strategies in a retrospective, nonrandomized fashion. Finally, one cannot neglect the different consequences that ICT vs AZA may have on the gut microbiome85  and host immune system,86  both of which may alter post-HSCT outcome.

Sequential combinations of chemotherapy and epigenetic therapy

ICT and HMAs are not mutually exclusive. To combine HMAs with standard induction chemotherapy seemed to be too toxic.87  Using HMAs as postremission or maintenance therapy in patients in remission after ICT is attractive, because one may hypothesize a reduction in relapse incidence through the efficacy of epigenetic agents on residual MDS-like clonal hematopoiesis. This has been investigated,88,89  and a British group has reported prolonged survival in older patients receiving AZA maintenance after achieving MRD-negative remission after ICT.89  This contrasts with other reports not stratified on MRD, where DAC or low-dose AZA maintenance provided no obvious benefit compared with historical controls in AML patients in first CR.90,91  A randomized trial is currently testing the role of oral AZA in this setting (NCT01757535).

Epigenetic therapy at relapse

Activity of HMA in R/R AML has mostly been reported retrospectively, with overall response rate in the 15% to 35% range, including 10% to 15% CRs, and median OS around 7 to 9 months, although prolonged survival, but not cure, was seen in patients reaching CR.45,50,51,92  Prognostic factors in the R/R setting vary between reports, highlighting the differences in study populations. Some studies stressed the role of CR duration and BM blast percentage,50,92  whereas others stressed the importance of cytogenetics and the presence of peripheral blasts.51  Despite differences in study populations, the convergent median survival values seen in these series, which are comparable with other approaches of intermediate-intensity chemotherapy in R/R AML, nevertheless provide a coherent benchmark for the wealth of ongoing phase 2 trials in this setting, where single-agent HMA is often included as a reference arm.

Hervé Dombret, Department of Hematology, University Hospital Saint-Louis, 1 Avenue Claude Vellefaux, 75010 Paris, France; e-mail: herve.dombret@aphp.fr.

1.
Döhner
H
,
Estey
E
,
Grimwade
D
, et al
.
Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel
.
Blood
.
2017
;
129
(
4
):
424
-
447
.
2.
Sekeres
MA
,
Elson
P
,
Kalaycio
ME
, et al
.
Time from diagnosis to treatment initiation predicts survival in younger, but not older, acute myeloid leukemia patients
.
Blood
.
2009
;
113
(
1
):
28
-
36
.
3.
Bertoli
S
,
Bérard
E
,
Huguet
F
, et al
.
Time from diagnosis to intensive chemotherapy initiation does not adversely impact the outcome of patients with acute myeloid leukemia
.
Blood
.
2013
;
121
(
14
):
2618
-
2626
.
4.
Löwenberg
B
,
Ossenkoppele
GJ
,
van Putten
W
, et al
;
Dutch-Belgian Cooperative Trial Group for Hemato-Oncology (HOVON); German AML Study Group (AMLSG); Swiss Group for Clinical Cancer Research (SAKK) Collaborative Group
.
High-dose daunorubicin in older patients with acute myeloid leukemia [publisched correction appears in N Engl J Med. 2010;362(12);1155]
.
N Engl J Med
.
2009
;
361
(
13
):
1235
-
1248
.
5.
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
;
31
(
3
):
321
-
327
.
6.
Mayer
RJ
,
Davis
RB
,
Schiffer
CA
, et al
;
Cancer and Leukemia Group B
.
Intensive postremission chemotherapy in adults with acute myeloid leukemia
.
N Engl J Med
.
1994
;
331
(
14
):
896
-
903
.
7.
Stone
RM
,
Berg
DT
,
George
SL
, et al
.
Postremission therapy in older patients with de novo acute myeloid leukemia: a randomized trial comparing mitoxantrone and intermediate-dose cytarabine with standard-dose cytarabine
.
Blood
.
2001
;
98
(
3
):
548
-
553
.
8.
Gardin
C
,
Turlure
P
,
Fagot
T
, et al
.
Postremission treatment of elderly patients with acute myeloid leukemia in first complete remission after intensive induction chemotherapy: results of the multicenter randomized Acute Leukemia French Association (ALFA) 9803 trial
.
Blood
.
2007
;
109
(
12
):
5129
-
5135
.
9.
Itzykson
R
,
Gardin
C
,
Pautas
C
, et al
;
Acute Leukemia French Association (ALFA)
.
Impact of post-remission therapy in patients aged 65-70 years with de novo acute myeloid leukemia: a comparison of two concomitant randomized ALFA trials with overlapping age inclusion criteria
.
Haematologica
.
2011
;
96
(
6
):
837
-
844
.
10.
Niederwieser
D
,
Hoffmann
VS
,
Pfirrmann
M
, et al
. Comparison of treatment strategies in patients over 60 years with AML: final analysis of a prospective randomized German AML Intergroup study. In: Proceedings from the American Society of Hematology Annual Meeting; 3-6 December
2016
; San Diego, CA. Abstract 1066.
11.
Dombret
H
,
Gardin
C
.
An update of current treatments for adult acute myeloid leukemia
.
Blood
.
2016
;
127
(
1
):
53
-
61
.
12.
Stein
EM
,
Tallman
MS
.
Emerging therapeutic drugs for AML
.
Blood
.
2016
;
127
(
1
):
71
-
78
.
13.
Pigneux
A
,
Béné
MC
,
Guardiola
P
, et al
.
Addition of androgens improves survival in elderly patients with acute myeloid leukemia: a GOELAMS study
.
J Clin Oncol
.
2017
;
35
(
4
):
387
-
393
.
14.
Prébet
T
,
Boissel
N
,
Reutenauer
S
, et al
;
Acute Leukemia French Association; Groupe Ouest-Est des leucémies et autres maladies du sang (GOELAMS); Core Binding Factor Acute Myeloid Leukemia (CBF AML) intergroup
.
Acute myeloid leukemia with translocation (8;21) or inversion (16) in elderly patients treated with conventional chemotherapy: a collaborative study of the French CBF-AML intergroup
.
J Clin Oncol
.
2009
;
27
(
28
):
4747
-
4753
.
15.
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
;
28
(
4
):
596
-
604
.
16.
Genovese
G
,
Kähler
AK
,
Handsaker
RE
, et al
.
Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence
.
N Engl J Med
.
2014
;
371
(
26
):
2477
-
2487
.
17.
Jaiswal
S
,
Fontanillas
P
,
Flannick
J
, et al
.
Age-related clonal hematopoiesis associated with adverse outcomes
.
N Engl J Med
.
2014
;
371
(
26
):
2488
-
2498
.
18.
Haferlach
T
,
Nagata
Y
,
Grossmann
V
, et al
.
Landscape of genetic lesions in 944 patients with myelodysplastic syndromes
.
Leukemia
.
2014
;
28
(
2
):
241
-
247
.
19.
Rothenberg-Thurley
M
,
Amler
S
,
Goerlich
D
, et al
. Persistence of driver mutations during complete remission associates with shorter survival and contributes to the inferior outcomes of elderly patients with acute myeloid leukemia. In: Proceedings from the European Hematology Association Annual Meeting; 9-12 June
2016
; Copenhagen, Denmark. Abstract S146.
20.
Lindsley
RC
,
Mar
BG
,
Mazzola
E
, et al
.
Acute myeloid leukemia ontogeny is defined by distinct somatic mutations
.
Blood
.
2015
;
125
(
9
):
1367
-
1376
.
21.
Ng
SWK
,
Mitchell
A
,
Kennedy
JA
, et al
.
A 17-gene stemness score for rapid determination of risk in acute leukaemia
.
Nature
.
2016
;
540
(
7633
):
433
-
437
.
22.
Klepin
HD
.
Definition of unfit for standard acute myeloid leukemia therapy
.
Curr Hematol Malig Rep
.
2016
;
11
(
6
):
537
-
544
.
23.
Ofran
Y
,
Tallman
MS
,
Rowe
JM
.
How I treat acute myeloid leukemia presenting with preexisting comorbidities
.
Blood
.
2016
;
128
(
4
):
488
-
496
.
24.
Sorror
ML
,
Maris
MB
,
Storb
R
, et al
.
Hematopoietic cell transplantation (HCT)-specific comorbidity index: a new tool for risk assessment before allogeneic HCT
.
Blood
.
2005
;
106
(
8
):
2912
-
2919
.
25.
Almeida
AM
,
Ramos
F
.
Acute myeloid leukemia in the older adults
.
Leuk Res Rep
.
2016
;
6
:
1
-
7
.
26.
Sekeres
MA
,
Gerds
AT
.
Mitigating fear and loathing in managing acute myeloid leukemia
.
Semin Hematol
.
2015
;
52
(
3
):
249
-
255
.
27.
Spencer
DH
,
Russler-Germain
DA
,
Ketkar
S
, et al
.
CpG island hypermethylation mediated by DNMT3A is a consequence of AML progression
.
Cell
.
2017
;
168
(
5
):
801
-
816.e13
.
28.
Glass
J
,
Hassane
DC
,
Wouters
B
, et al
.
Epigenetic identity in AML depends on disruption of non-promoter regulatory elements and is affected by antagonistic effects of mutations in epigenetic modifiers [published online ahead of print 1 April 2017]
.
Cancer Discov
.
29.
Issa
JJ
,
Roboz
G
,
Rizzieri
D
, et al
.
Safety and tolerability of guadecitabine (SGI-110) in patients with myelodysplastic syndrome and acute myeloid leukaemia: a multicentre, randomised, dose-escalation phase 1 study
.
Lancet Oncol
.
2015
;
16
(
9
):
1099
-
1110
.
30.
Gardin
C
,
Dombret
H
.
Hypomethylating agents as a therapy for AML
.
Curr Hematol Malig Rep
.
2017
;
12
(
1
):
1
-
10
.
31.
Silverman
LR
,
McKenzie
DR
,
Peterson
BL
, et al
;
Cancer and Leukemia Group B
.
Further analysis of trials with azacitidine in patients with myelodysplastic syndrome: studies 8421, 8921, and 9221 by the Cancer and Leukemia Group B
.
J Clin Oncol
.
2006
;
24
(
24
):
3895
-
3903
.
32.
Cashen
AF
,
Schiller
GJ
,
O’Donnell
MR
,
DiPersio
JF
.
Multicenter, phase II study of decitabine for the first-line treatment of older patients with acute myeloid leukemia
.
J Clin Oncol
.
2010
;
28
(
4
):
556
-
561
.
33.
Blum
W
,
Garzon
R
,
Klisovic
RB
, et al
.
Clinical response and miR-29b predictive significance in older AML patients treated with a 10-day schedule of decitabine
.
Proc Natl Acad Sci USA
.
2010
;
107
(
16
):
7473
-
7478
.
34.
Fenaux
P
,
Mufti
GJ
,
Hellström-Lindberg
E
, et al
.
Azacitidine prolongs overall survival compared with conventional care regimens in elderly patients with low bone marrow blast count acute myeloid leukemia
.
J Clin Oncol
.
2010
;
28
(
4
):
562
-
569
.
35.
Lübbert
M
,
Rüter
BH
,
Claus
R
, et al
.
A multicenter phase II trial of decitabine as first-line treatment for older patients with acute myeloid leukemia judged unfit for induction chemotherapy
.
Haematologica
.
2012
;
97
(
3
):
393
-
401
.
36.
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
;
30
(
21
):
2670
-
2677
.
37.
Becker
H
,
Suciu
S
,
Rüter
BH
, et al
.
Decitabine versus best supportive care in older patients with refractory anemia with excess blasts in transformation (RAEBt) - results of a subgroup analysis of the randomized phase III study 06011 of the EORTC Leukemia Cooperative Group and German MDS Study Group (GMDSSG)
.
Ann Hematol
.
2015
;
94
(
12
):
2003
-
2013
.
38.
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
;
126
(
3
):
291
-
299
.
39.
Schuh
A
,
Dombret
H
,
Sandhu
I
, et al
. Overall survival without complete remission in older patients with acute myeloid leukemia: azacitidine versus conventional care regimens in the AZA-AML-001 study. In: Proceedings from the European Hematology Association Annual Meeting; 11-14 June
2015
; Vienna, Austria. Abstract S.
40.
Schuh
A
,
Döhner
H
,
Seymour
JF
, et al
. Stable disease with hematologic improvement is clinically meaningful for older patients with acute myeloid leukemia (AML) treated with azacitidine. In: Proceedings from the European Hematology Association Annual Meeting; 22-25 June
2017
; Madrid, Spain. Abstract P208.
41.
Kadia
TM
,
Thomas
XG
,
Dmoszynska
A
, et al
.
Decitabine improves outcomes in older patients with acute myeloid leukemia and higher blast counts
.
Am J Hematol
.
2015
;
90
(
7
):
E139
-
E141
.
42.
Thepot
S
,
Itzykson
R
,
Seegers
V
, et al
;
Groupe Francophone des Myelodysplasies (GFM)
.
Treatment of progression of Philadelphia-negative myeloproliferative neoplasms to myelodysplastic syndrome or acute myeloid leukemia by azacitidine: a report on 54 cases on the behalf of the Groupe Francophone des Myelodysplasies (GFM)
.
Blood
.
2010
;
116
(
19
):
3735
-
3742
.
43.
Maurillo
L
,
Venditti
A
,
Spagnoli
A
, et al
.
Azacitidine for the treatment of patients with acute myeloid leukemia: report of 82 patients enrolled in an Italian Compassionate Program
.
Cancer
.
2012
;
118
(
4
):
1014
-
1022
.
44.
Thépot
S
,
Itzykson
R
,
Seegers
V
, et al
;
Groupe Francophone des Myélodysplasies (GFM), Acute Leukemia French Association (ALFA); Groupe Ouest-Est des Leucémies Aiguës; Maladies du Sang (GOELAMS)
.
Azacitidine in untreated acute myeloid leukemia: a report on 149 patients
.
Am J Hematol
.
2014
;
89
(
4
):
410
-
416
.
45.
Pleyer
L
,
Burgstaller
S
,
Girschikofsky
M
, et al
.
Azacitidine in 302 patients with WHO-defined acute myeloid leukemia: results from the Austrian Azacitidine Registry of the AGMT-Study Group
.
Ann Hematol
.
2014
;
93
(
11
):
1825
-
1838
.
46.
Bories
P
,
Bertoli
S
,
Bérard
E
, et al
.
Intensive chemotherapy, azacitidine, or supportive care in older acute myeloid leukemia patients: an analysis from a regional healthcare network
.
Am J Hematol
.
2014
;
89
(
12
):
E244
-
E252
.
47.
Ramos
F
,
Thépot
S
,
Pleyer
L
, et al
;
European ALMA Investigators
.
Azacitidine frontline therapy for unfit acute myeloid leukemia patients: clinical use and outcome prediction
.
Leuk Res
.
2015
;
39
(
3
):
296
-
306
.
48.
Pleyer
L
,
Döhner
H
,
Dombret
H
, et al
.
Azacitidine for front-line therapy of patients with AML: reproducible efficacy established by direct comparison of international phase 3 trial data with registry data from the Austrian azacitidine registry of the AGMT Study Group
.
Int J Mol Sci
.
2017
;
18
(
2
):
415
-
433
.
49.
Dumas
PY
,
Bertoli
S
,
Bérard
E
, et al
.
Azacitidine or intensive chemotherapy for older patients with secondary or therapy-related acute myeloid leukemia [published online ahead of print 7 March 2017]
.
Oncotarget
.
50.
Ivanoff
S
,
Gruson
B
,
Chantepie
SP
, et al
.
5-Azacytidine treatment for relapsed or refractory acute myeloid leukemia after intensive chemotherapy
.
Am J Hematol
.
2013
;
88
(
7
):
601
-
605
.
51.
Itzykson
R
,
Thépot
S
,
Berthon
C
, et al
.
Azacitidine for the treatment of relapsed and refractory AML in older patients
.
Leuk Res
.
2015
;
39
(
2
):
124
-
130
.
52
Döhner
H
,
Vyas
P
,
Seymour
JF
, et al
.
Azacitidine (AZA) prolongs overall survival in older patients with acute myeloid leukemia (AML) with poor prognostic karyotypes compared to conventional care regimens (CCR)
. In: Proceedings from the American Society of Hematology Annual Meeting; 3-6 December
2016
; San Diego, CA. Abstract 1638.
53.
Wanquet
A
,
Prebet
T
,
Berthon
C
, et al
.
Azacitidine treatment for patients with myelodysplastic syndrome and acute myeloid leukemia with chromosome 3q abnormalities
.
Am J Hematol
.
2015
;
90
(
10
):
859
-
863
.
54.
Mayer
J
,
Arthur
C
,
Delaunay
J
, et al
.
Multivariate and subgroup analyses of a randomized, multinational, phase 3 trial of decitabine vs treatment choice of supportive care or cytarabine in older patients with newly diagnosed acute myeloid leukemia and poor- or intermediate-risk cytogenetics
.
BMC Cancer
.
2014
;
14
:
69
-
77
.
55.
Gardin
C
,
Itzykson
R
,
Thépot
S
, et al
.
Frontline azacitidine (AZA) or intensive chemotherapy (ICTx) in older AML patients [abstract]
.
J Clin Oncol
.
2010
;
26
(
suppl
):
15s
. Abstract 6530.
56.
Voso
MT
,
Santini
V
,
Fabiani
E
, et al
.
Why methylation is not a marker predictive of response to hypomethylating agents
.
Haematologica
.
2014
;
99
(
4
):
613
-
619
.
57.
Merledeve
J
,
Droin
N
,
Qin
T
, et al
.
Mutation allele burden remains unchanged in chronic myelomonocytic leukaemia responding to hypomethylating agents
.
Nat Commun
.
2016
;
7
:
10767
.
58.
Welch
JS
,
Petti
AA
,
Miller
CA
, et al
.
TP53 and decitabine in acute myeloid leukemia and myelodysplastic syndromes
.
N Engl J Med
.
2016
;
375
(
21
):
2023
-
2036
.
59.
Itzykson
R
,
Kosmider
O
,
Cluzeau
T
, et al
;
Groupe Francophone des Myelodysplasies (GFM)
.
Impact of TET2 mutations on response rate to azacitidine in myelodysplastic syndromes and low blast count acute myeloid leukemias
.
Leukemia
.
2011
;
25
(
7
):
1147
-
1152
.
60.
Metzeler
KH
,
Walker
A
,
Geyer
S
, et al
.
DNMT3A mutations and response to the hypomethylating agent decitabine in acute myeloid leukemia
.
Leukemia
.
2012
;
26
(
5
):
1106
-
1107
.
61.
Traina
F
,
Visconte
V
,
Elson
P
, et al
.
Impact of molecular mutations on treatment response to DNMT inhibitors in myelodysplasia and related neoplasms
.
Leukemia
.
2014
;
28
(
1
):
78
-
87
.
62.
Bejar
R
,
Lord
A
,
Stevenson
K
, et al
.
TET2 mutations predict response to hypomethylating agents in myelodysplastic syndrome patients
.
Blood
.
2014
;
124
(
17
):
2705
-
2712
.
63.
Desoutter
J
,
Gay
J
,
Berthon
C
, et al
.
Molecular prognostic factors in acute myeloid leukemia receiving first-line therapy with azacitidine
.
Leukemia
.
2016
;
30
(
6
):
1416
-
1418
.
64.
Tang
L
,
Dolnik
A
,
MacBeth
KJ
, et al
. Impact of gene mutations on overall survival in older patients with acute myeloid leukemia (AML) treated with azacitidine (AZA) or conventional care regimens (CCR). In: Proceedings from the American Society of Hematology Annual Meeting; 3-6 December
2016
; San Diego, CA. Abstract 2859.
65.
Kantarjian
H
,
Oki
Y
,
Garcia-Manero
G
, et al
.
Results of a randomized study of 3 schedules of low-dose decitabine in higher-risk myelodysplastic syndrome and chronic myelomonocytic leukemia
.
Blood
.
2007
;
109
(
1
):
52
-
57
.
66.
Blum
W
,
Klisovic
RB
,
Hackanson
B
, et al
.
Phase I study of decitabine alone or in combination with valproic acid in acute myeloid leukemia
.
J Clin Oncol
.
2007
;
25
(
25
):
3884
-
3891
.
67.
Daskalakis
M
,
Nguyen
TT
,
Nguyen
C
, et al
.
Demethylation of a hypermethylated P15/INK4B gene in patients with myelodysplastic syndrome by 5-Aza-2′-deoxycytidine (decitabine) treatment
.
Blood
.
2002
;
100
(
8
):
2957
-
2964
.
68.
Follo
MY
,
Finelli
C
,
Mongiorgi
S
, et al
.
Reduction of phosphoinositide-phospholipase C beta1 methylation predicts the responsiveness to azacitidine in high-risk MDS
.
Proc Natl Acad Sci USA
.
2009
;
106
(
39
):
16811
-
16816
.
69.
Meldi
K
,
Qin
T
,
Buchi
F
, et al
.
Specific molecular signatures predict decitabine response in chronic myelomonocytic leukemia
.
J Clin Invest
.
2015
;
125
(
5
):
1857
-
1872
.
70.
Shen
L
,
Kantarjian
H
,
Guo
Y
, et al
.
DNA methylation predicts survival and response to therapy in patients with myelodysplastic syndromes
.
J Clin Oncol
.
2010
;
28
(
4
):
605
-
613
.
71.
Li
S
,
Garrett-Bakelman
FE
,
Chung
SS
, et al
.
Distinct evolution and dynamics of epigenetic and genetic heterogeneity in acute myeloid leukemia
.
Nat Med
.
2016
;
22
(
7
):
792
-
799
.
72.
Valencia
A
,
Masala
E
,
Rossi
A
, et al
.
Expression of nucleoside-metabolizing enzymes in myelodysplastic syndromes and modulation of response to azacitidine
.
Leukemia
.
2014
;
28
(
3
):
621
-
628
.
73.
Quintás-Cardama
A
,
Ravandi
F
,
Liu-Dumlao
T
, et al
.
Epigenetic therapy is associated with similar survival compared with intensive chemotherapy in older patients with newly diagnosed acute myeloid leukemia
.
Blood
.
2012
;
120
(
24
):
4840
-
4845
.
74.
Gupta
N
,
Miller
A
,
Gandhi
S
, et al
.
Comparison of epigenetic versus standard induction chemotherapy for newly diagnosed acute myeloid leukemia patients ≥60 years old
.
Am J Hematol
.
2015
;
90
(
7
):
639
-
646
.
75.
Medeiros
BC
,
Satram-Hoang
S
,
Hurst
D
,
Hoang
KQ
,
Momin
F
,
Reyes
C
.
Big data analysis of treatment patterns and outcomes among elderly acute myeloid leukemia patients in the United States
.
Ann Hematol
.
2015
;
94
(
7
):
1127
-
1138
.
76.
Prébet
T
,
Gore
SD
,
Thépot
S
, et al
.
Outcome of acute myeloid leukaemia following myelodysplastic syndrome after azacitidine treatment failure
.
Br J Haematol
.
2012
;
157
(
6
):
764
-
766
.
77.
Prébet
T
,
Gore
SD
,
Esterni
B
, et al
.
Outcome of high-risk myelodysplastic syndrome after azacitidine treatment failure
.
J Clin Oncol
.
2011
;
29
(
24
):
3322
-
3327
.
78.
Harel
S
,
Cherait
A
,
Berthon
C
, et al
.
Outcome of patients with high risk Myelodysplastic Syndrome (MDS) and advanced Chronic Myelomonocytic Leukemia (CMML) treated with decitabine after azacitidine failure
.
Leuk Res
.
2015
;
39
(
5
):
501
-
504
.
79.
Damaj
G
,
Duhamel
A
,
Robin
M
, et al
.
Impact of azacitidine before allogeneic stem-cell transplantation for myelodysplastic syndromes: a study by the Société Française de Greffe de Moelle et de Thérapie-Cellulaire and the Groupe-Francophone des Myélodysplasies
.
J Clin Oncol
.
2012
;
30
(
36
):
4533
-
4540
.
80.
Voso
MT
,
Leone
G
,
Piciocchi
A
, et al
.
Feasibility of allogeneic stem-cell transplantation after azacitidine bridge in higher-risk myelodysplastic syndromes and low blast count acute myeloid leukemia: results of the BMT-AZA prospective study
.
Ann Oncol
.
2017
;
28
(
7
):
1547
-
1553
.
81.
Estey
E
,
de Lima
M
,
Tibes
R
, et al
.
Prospective feasibility analysis of reduced-intensity conditioning (RIC) regimens for hematopoietic stem cell transplantation (HSCT) in elderly patients with acute myeloid leukemia (AML) and high-risk myelodysplastic syndrome (MDS)
.
Blood
.
2007
;
109
(
4
):
1395
-
1400
.
82.
Wong
TN
,
Miller
CA
,
Klco
JM
, et al
.
Rapid expansion of preexisting nonleukemic hematopoietic clones frequently follows induction therapy for de novo AML
.
Blood
.
2016
;
127
(
7
):
893
-
897
.
83.
Araki
D
,
Wood
BL
,
Othus
M
, et al
.
Allogeneic hematopoietic cell transplantation for acute myeloid leukemia: time to move toward a minimal residual disease-based definition of complete remission?
J Clin Oncol
.
2016
;
34
(
4
):
329
-
336
.
84.
Craddock
C
,
Quek
L
,
Goardon
N
, et al
.
Azacitidine fails to eradicate leukemic stem/progenitor cell populations in patients with acute myeloid leukemia and myelodysplasia
.
Leukemia
.
2013
;
27
(
5
):
1028
-
1036
.
85.
Galloway-Peña
JR
,
Smith
DP
,
Sahasrabhojane
P
, et al
.
The role of the gastrointestinal microbiome in infectious complications during induction chemotherapy for acute myeloid leukemia
.
Cancer
.
2016
;
122
(
14
):
2186
-
2196
.
86.
Cooper
ML
,
Choi
J
,
Karpova
D
, et al
.
Azacitidine mitigates graft-versus-host disease via differential effects on the proliferation of T effectors and natural regulatory T cells in vivo
.
J Immunol
.
2017
;
198
(
9
):
3746
-
3754
.
87.
Müller-Tidow
C
,
Tschanter
P
,
Röllig
C
, et al
;
Study Alliance Leukemia Group
.
Azacitidine in combination with intensive induction chemotherapy in older patients with acute myeloid leukemia: the AML-AZA trial of the Study Alliance Leukemia
.
Leukemia
.
2016
;
30
(
3
):
555
-
561
.
88.
Gardin
C
,
Prébet
T
,
Bouabdallah
K
, et al
. A phase II study of post–remission therapy with azacitidine (AZA) in patients with AML post-MDS and high-risk MDS: a GFM Group study. In: Proceedings from the American Society of Hematology Annual Meeting; 5-8 December
2009
; New Orleans, LA. Abstract 844.
89.
Burnett
AK
,
Russell
NH
,
Kell
J
, et al
. A comparison of daunorubicin/Ara-C (DA) versus daunorubicin/clofarabine (DClo) and two versus three courses of total treatment for older patients with AML and high risk MDS: results of the UK NCRI AML16 trial. In: Proceedings from the American Society of Hematology Annual Meeting; 8-11 December
2012
; Atlanta, GA. Abstract 892.
90.
Blum
W
,
Sanford
BL
,
Klisovic
R
, et al
;
Alliance for Clinical Trials in Oncology
.
Maintenance therapy with decitabine in younger adults with acute myeloid leukemia in first remission: a phase 2 Cancer and Leukemia Group B Study (CALGB 10503)
.
Leukemia
.
2017
;
31
(
1
):
34
-
39
.
91.
Griffin
PT
,
Komrokji
RS
,
De Castro
CM
, et al
.
A multicenter, phase II study of maintenance azacitidine in older patients with acute myeloid leukemia in complete remission after induction chemotherapy
.
Am J Hematol
.
2015
;
90
(
9
):
796
-
799
.
92.
Craddock
C
,
Labopin
M
,
Robin
M
, et al
.
Clinical activity of azacitidine in patients who relapse after allogeneic stem cell transplantation for acute myeloid leukemia
.
Haematologica
.
2016
;
101
(
7
):
879
-
883
.

Competing Interests

Conflict-of-interest disclosure: H.D. is on the Board of Directors or an advisory committee for Menarini, Servier, Seattle Genetics, Cellectis, Janssen, Astellas, Karyopharm, Sunesis, Daiichi Sankyo, Agios, Kite Pharma, Jazz Pharmaceuticals, Novartis, Incyte, Pfizer, Amgen, and Celgene; has received research funding from Servier, Kite Pharma, Jazz Pharmaceuticals, Incyte, Pfizer, and Amgen; has consulted for Menarini, Jazz Pharmaceuticals, Celgene, Chugai Pharmaceutical Co., and Roche; has received honoraria from Menarini, Servier, Seattle Genetics, Cellectis, Janssen, Astellas, Karyopharm, Sunesis, Daiichi Sankyo, Agios, Kite Pharma, Jazz Pharmaceuticals, Novartis, Incyte, Pfizer, Amgen, and Celgene; and has been affilitated with the Speaker’s Bureau for Incyte, Pfizer, Amgen, and Celgene. R.I. has received research funding from Janssen and honoraria from Celgene.

Author notes

Off-label drug use: None disclosed.