The ongoing development of molecularly targeted therapies in addition to the new standard of care combination of azacitidine and venetoclax (AZA-VEN) has transformed the prognostic outlook for older, transplant-ineligible patients with acute myeloid leukemia (AML). While conventional treatments, such as standard anthracycline and cytarabine- based chemotherapy or hypomethylating agent (HMA) monotherapy, are associated with a generally poor prognosis in this patient population, the use of these novel regimens can result in long-lasting, durable remissions in select patient subgroups. Furthermore, the simultaneous discovery of resistance mechanisms to targeted therapies and AZA-VEN has enabled the identification of patient subgroups with inferior outcomes, leading to the development, of new risk-stratification models and clinical investigations incorporating targeted therapies using an HMA-VEN–based platform. Treatments inclusive of IDH1, IDH2, FLT3, and menin inhibitors combined with HMA-VEN have additionally demonstrated safety and high rates of efficacy in early-phase clinical trials, suggesting these regimens may further improve outcomes within select subgroups of patients with AML in the near future. Additional studies defining the prognostic role of measurable residual disease following VEN-based treatment have further advanced prognostication capabilities and increased the ability for close disease monitoring and early targeted intervention prior to morphologic relapse. This review summarizes these recent developments and their impact on the treatment and survival of transplant-ineligible patients living with AML.

Learning Objectives

  • Evaluate the impact of baseline genomics on outcomes for patients with AML treated with lower-intensity regimens

  • Recognize patient subgroups that may benefit from novel treatment approaches combining targeted therapy with hypomethylating agents

A 73-year-old man with a history of head and neck cancer, chronic obstructive pulmonary disease, and congestive heart failure presents with anemia, neutropenia, and circulating blasts. A bone marrow biopsy confirms a diagnosis of AML. Additional testing reveals a diploid karyotype and mutations in DNMT3A and IDH1. Given his comorbidities, he is not considered a candidate for induction chemotherapy.

Acute myeloid leukemia (AML) is a life-threatening hematologic malignancy primarily occurring in older adults (median age at diagnosis: 69 years).1 Historically, older patients with AML experience poor survival due to increased treatment-related mortality, adverse-risk molecular characteristics, and persistent measurable residual disease (MRD) resulting in disease relapse.2-5 

Long-term survival rates of 30% to 60% are realized following allogeneic hematopoietic cell transplantation (HCT), yet only approximately 30% of patients successfully proceed with HCT in remission.6,7 Increasing age, concurrent comorbidities, and primary refractory disease limit the applicability of both intensive chemotherapy (IC) and HCT in older patients.8 In this setting, combination therapies with azacitidine plus venetoclax (AZA-VEN) and azacitidine plus ivosidenib (AZA-IVO; approved for newly diagnosed AML with a concurrent IDH1 mutation) are now standard treatments that have transformed outcomes in this difficult to treat population.9,10 

Translational studies developed alongside the rapid clinical adoption of AZA-VEN characterized key resistance mechanisms and patient subgroups with differing prognoses.11-20 New treatment-specific risk models, molecular and/or flow-cytometric (MFC)–based MRD monitoring and intervention, and emerging targeted therapy combinations are enabling treatment individualization and further improving clinical outcomes.21-28 

With these advancements, the treatment outlook for older, unfit patients with AML has simultaneously become more optimistic while becoming increasingly complex and individualized. This review summarizes current and emerging lower-intensity treatment perspectives for older, unfit patients with AML and provides a practical framework to guide clinical decisions across genetically defined AML subgroups (Figure 1).

Figure 1.

Contemporary treatment approach for transplant-ineligible patients with AML. The initial evaluation should consist of a comprehensive history and physical exam to ascertain the patient's fitness and ability to undergo leukemia-directed therapy, followed by a bone marrow examination with special attention to cytogenetic and molecular abnormalities that can inform treatment selection and prognosis. Risk-stratification using either the 4-gene prognostic risk signature or Mayo Clinic risk-prognostication model (after initiation of therapy) may provide important prognostic information. For patients with IDH1-mutated AML, AZA-IVO may be preferred over HMA-VEN. For patients with TP53-mutated AML, a clinical trial option is preferred over HMA-VEN if available. Ongoing clinical trials (highlighted in yellow boxes) are actively investigating targeted therapies in combination with HMA-VEN in molecularly informed patient subgroups.

Figure 1.

Contemporary treatment approach for transplant-ineligible patients with AML. The initial evaluation should consist of a comprehensive history and physical exam to ascertain the patient's fitness and ability to undergo leukemia-directed therapy, followed by a bone marrow examination with special attention to cytogenetic and molecular abnormalities that can inform treatment selection and prognosis. Risk-stratification using either the 4-gene prognostic risk signature or Mayo Clinic risk-prognostication model (after initiation of therapy) may provide important prognostic information. For patients with IDH1-mutated AML, AZA-IVO may be preferred over HMA-VEN. For patients with TP53-mutated AML, a clinical trial option is preferred over HMA-VEN if available. Ongoing clinical trials (highlighted in yellow boxes) are actively investigating targeted therapies in combination with HMA-VEN in molecularly informed patient subgroups.

Close modal

The European LeukemiaNet (ELN) 2022 guidelines, developed from younger patients with AML treated with standard IC, inadequately risk stratify patients receiving HMA-VEN therapy.21,29,30 Two newer treatment-specific risk-stratification models for HMA-VEN are now available (Figure 2).21,22 

Figure 2.

Risk-stratification models for patients receiving HMA-VEN therapies. Two models available for use include the molecular prognostic risk signature (mPRS) and Mayo Clinic risk-prognostication model. The mPRS utilizes 4 key genes (N/KRAS, FLT3-ITD, and TP53) to characterize patients into higher-benefit (ie, low-risk), intermediate-benefit, and lower-benefit (ie, high-risk) groups. Of note, using this 4-gene model, approximately 40% to 50% of patients will be characterized as higher benefit. The Mayo Clinic risk- prognostication model classifies patients into low, intermediate, and high-risk groups based on the incorporation of molecular and response features. Similar to the mPRS model, approximately 40% of patients are categorized as low risk.

Figure 2.

Risk-stratification models for patients receiving HMA-VEN therapies. Two models available for use include the molecular prognostic risk signature (mPRS) and Mayo Clinic risk-prognostication model. The mPRS utilizes 4 key genes (N/KRAS, FLT3-ITD, and TP53) to characterize patients into higher-benefit (ie, low-risk), intermediate-benefit, and lower-benefit (ie, high-risk) groups. Of note, using this 4-gene model, approximately 40% to 50% of patients will be characterized as higher benefit. The Mayo Clinic risk- prognostication model classifies patients into low, intermediate, and high-risk groups based on the incorporation of molecular and response features. Similar to the mPRS model, approximately 40% of patients are categorized as low risk.

Close modal

The 4-gene molecular prognostic-risk signature (mPRS) proposed by Döhner et al defines 3 groups based on the phase 3 VIALE-A treatment population: higher benefit (wild-type FLT3-ITD, N/KRAS, and TP53), intermediate (a mutation in either FLT3-ITD and/or N/KRAS and wild-type TP53), and lower benefit (presence of a TP53 mutation) corresponding to a median overall survival (OS) of 24, 12, and less than 6 months, respectively.30 The lower-benefit group was enriched for patients with complex cytogenetics (89%), which, while not predictive of VEN resistance in isolation, are frequently associated with TP53 mutations.13,31 

A validation study supports the utility of the mPRS compared to ELN 2022.21 Across the higher, intermediate, and lower- benefit groups, the CR/CRi rates were 86%, 54%, and 59%, and the median OS was 30, 12, and 5 months with a 2-year cumulative incidence of relapse of 35%, 70%, and 60%, respectively. A minority (12%) of patients received HCT in remission. Thirty-five percent of patients classified as ELN adverse risk were reallocated into the mPRS higher-benefit group, likely a reflection of ascribing myelodysplasia-related mutations (ASXL1, RUNX1, SRSF2, SF3B1, U2AF1, ZRSR2, EHZ2, BCOR, STAG2) as adverse risk in ELN 2022 guidelines.32 This latter point highlights that molecular- based prognostication models are treatment-specific. Prior studies demonstrated that VEN-based therapies attenuate the inferior survival associated with myelodysplasia-related mutations in the setting of IC.33-35 

The Mayo Clinic risk-prognostication model incorporates cytogenetics, mutational status, and clinical response.22 Favorable mutations predictive of an increased likelihood of CR/CRi included NPM1, IDH2, and/or DDX41, while unfavorable cytogenetics, mutated FLT3-ITD, or RUNX1 corresponded with lower CR/CRi rates with HMA-VEN regimens. Multivariate analysis identified the failure to attain CR/CRi, ELN 2022 adverse-risk cytogenetics, the presence of a TP53-mutation, and the lack of an IDH2 mutation as significant predictors of inferior survival, permitting development of a response-based prognostic score allocating patients into low-, intermediate-, and high-risk groups.22 The 3-year OS (censored for HCT) across these groups was 42%, 5%, and 0%, respectively. Like the mPRS population, only 13% of patients received HCT.

An expanded set of mutations now predict favorable responses and survival following VEN-based therapy, including mutated NPM1, IDH2, or DDX41, particularly in the absence of signaling mutations.9-14,19,20,30 The higher-benefit group from the mPRS (median OS, 30 months; 2-year cumulative incidence of relapse, 35%) and the low-risk group from the Mayo Clinic model (3-year OS, 42%) identify patients with improved long-term survival with HMA-VEN regimens. These outcomes compare favorably to reported survival in transplant-eligible patients over age 60 undergoing HCT.8,36 Some patients may additionally experience improved fitness following treatment enabling HCT candidacy, which should remain an important consideration for all patients receiving HMA-VEN therapy as this therapy is not considered curative in the majority of patients.37 

The mPRS and Mayo Clinic models differ slightly in prognostication, likely related to the inclusion of CR/CRi as a variable within the Mayo Clinic model and different frequencies in prognostic gene mutations and co-occurring mutations between cohorts.

Long-term survival data from the phase 3 VIALE-A trial identified differing outcomes between patients with IDH1 vs IDH2 mutations (median OS of 10.2 vs 27.5 months), with IDH2-mutated AML experiencing particularly favorable outcomes following HMA-VEN (Table 1).38 

For IDH1-mutated AML, treatment with the IDH1 inhibitor ivosidenib in combination with azacitidine (IVO-AZA) is generally preferred based on data from the phase 3 AGILE trial, a placebo-controlled, randomized, double-blind study evaluating IVO-AZA vs AZA-placebo (Table 2).10 IVO-AZA improved event-free survival vs AZA-placebo (hazard ratio, 0.33; 95% CI, 0.16-0.69; P  =  .002).10 An updated OS analysis with a median follow-up of 28.6 months continues to favor IVO-AZA vs AZA-placebo (29.3 vs 7.9 months; P < .0001).39 Compared with the more cytotoxic activity of HMA-VEN (median time to CR/CRi, 1.2 months), responses occurred more slowly with IVO-AZA (median time to CR/CRh, 4 months); however, composite CR/CRi rates (54% vs 67%) and median duration of response (not reached vs 22 months) are comparable.10,18 Rates of grade 3 or greater febrile neutropenia (28% vs 42%) and infections of any grade (28% vs 59%) appear to favor IVO-AZA vs AZA-VEN,9,10 although QTc prolongation and IDH-inhibitor related differentiation syndrome (IDH-DS) are higher with AZA-IVO (Table 3).10 

Key questions remain concerning optimizing the use of these 2 effective doublets in IDH1-mutated AML and, more broadly, about sequencing, combining targeted therapies, and the role of MRD-monitoring in IDH1/2-mutated AML.

Prospective data on sequencing remain sparse. Incorporating an IDH inhibitor with an HMA-VEN backbone may improve response and survival compared to use following HMA-VEN therapy.40,41 In 1 retrospective analysis, salvage response rates using HMA-VEN after receipt of a frontline IDH inhibitor–based regimen were 88% vs 56% when using an IDH inhibitor following HMA-VEN.40 Most (60%) of these latter responses were observed when IDH inhibitors were integrated as triplet regimens with continuation of the HMA-VEN backbone. Few patients (8%) enrolled in VIALE-A with IDH1/2-mutated AML received IDH inhibitors at progression, and data on the use of HMA-VEN following IVO-AZA are not readily available.38 The I-DATA study (NCT05401097) will attempt to prospectively answer questions pertaining to the optimal sequencing of treatment in IDH1-mutated AML.

If ongoing studies confirm tolerability, “triplet” therapy combining IDH1/2 inhibitors with an HMA-VEN backbone may optimize therapy in patients with IDH1/2 mutations (Table 2).23,42 High composite CR rates were observed in patients with newly diagnosed AML treated with the triplet of IVO-VEN-AZA (93%; N  =  13/14), IVO in combination with oral decitabine (ASTX-727, decitabine/cedazuridine [C-DEC]) and VEN (90%; N  =  9/10), or the IDH2 inhibitor enasidenib with C-DEC–VEN (100%; N  =  14/14).23,42 This latter combination appears to further increase activity in patients with IDH2-mutated AML compared to the doublet of enasidenib-AZA, which improved CR/CRi rates compared with AZA monotherapy.43 

After a median follow-up of 24 months, the estimated 24-month OS of 67% in patients with newly diagnosed AML treated with IVO-VEN-AZA compares favorably to reported outcomes with AZA-VEN or IVO-AZA, respectively.23 

Additionally, triplet therapy incorporating IVO or ENA resulted in MRD-negative (measured using MFC) remissions in 80% to 83% and 93% of patients with newly diagnosed IDH1- or IDH2-mutated AML, which compares favorably to MRD-MFC negative rates observed with AZA-VEN (42% and 50%, respectively).23,42IDH1 mutation clearance by droplet digital polymerase chain reaction (PCR) occurred in 64% of patients receiving 5 or more cycles of IVO-VEN-AZA.23 While MFC was prognostic, molecular clearance did not correspond with survival, similar to recent reports that IDH1/2 mutations in remission following IC do not influence disease relapse post HCT.23,44 

Two planned studies inclusive of larger patient cohorts in the NCI MyeloMatch initiative will provide additional insight into the benefit of frontline triplet therapy in IDH1/2-mutated AML, prospectively randomizing patients to an all-oral combination of C-DEC-VEN and an IDH1 or IDH2 inhibitor vs C-DEC-VEN.

Responses in patients with FLT3-ITD–mutated AML are improved with AZA-VEN vs AZA monotherapy (CR/CRi, 63% vs 46%)12; however, survival remains suboptimal and is similar between AZA-VEN vs AZA (median OS, 9.9 vs 8.5 months) (Table 1).12 Fortunately, trials incorporating targeted therapy appear to improve response rates and survival in this higher-risk molecular subgroup; once combination dosing and schedules are established, these may soon represent a new standard of care.

More specifically, marked activity is observed when incorporating the type 2 FLT3 inhibitor gilteritinib with VEN as a doublet, or AZA-VEN-GILT as a triplet (Table 2).24,45 The frontline use of AZA-VEN-GILT resulted in a CR/CRi rate of 96% (N  =  29/30), including 93% of patients attaining MRD-MFC negative remissions and 65% attaining FLT3-ITD levels of less than 5  ×  10−5 following 4 cycles of therapy.24 After a median follow-up of 19.3 months, the estimated 18-month relapse-free survival and OS were 71% and 72%, respectively. Frontline use of the type 1 FLT3 inhibitor quizartinib with decitabine-VEN (DAC-VEN-QUIZ) demonstrated similarly impressive response rates.46 DAC-VEN-QUIZ resulted in a CR/CRi in 100% (N  =  10/10) of patients, including 75% with undetectable FLT3-ITD PCR.46 After a median follow-up of 11 months, a median OS was not reached.

Myelosuppression with triplet regimens incorporating FLT3 inhibitors is notable and is managed by attenuating the FLT3i dose (gilteritinib to 80  mg/d, quizartinib to 30  mg/d); the use of early disease assessment on cycle 1, day 14, with treatment interruption if blasts are fewer than 5% or marrow aplasia is observed; and the use of a 7-day course of venetoclax from cycle 2 onward.24 With this approach, rates febrile neutropenia and infection of grade 3 or higher were similar to HMA-VEN doublets (33% and 53%, respectively). A randomized dose-ranging and expansion study to identify the optimal dose of the GILT triplet combination (NCT05520567) is ongoing, and a randomized study comparing GILT-C-DEC-VEN to C-DEC-VEN is planned in the upcoming MyeloMatch Initiative.

VEN-GILT and AZA-VEN-GILT remain salvage options for patients with relapsed or refractory (R/R) FLT3-ITD-mutated AML, though with decreased efficacy if previously treated with HMA-VEN.24,45 In all patients with R/R AML, the composite CR rate (CR + CRi + CRp + morphologic leukemia-free state) to VEN-GILT and AZA-VEN-GILT was 75% and 68%, and the median OS was 10 and 5.8 months, respectively. Reduction of FLT3 transcript levels to less than 10−4 and less than 10−5 occurred in 20% and 27% of patients with R/R AML treated with VEN-GILT or AZA-VEN-GILT, respectively.

Serial monitoring of FLT3-ITD transcripts and the use of a FLT3 inhibitor in select patients with molecular persistence or rising MRD on HMA-VEN regimens may also preempt overt morphologic relapse and further improve survival, either as a bridge to HCT or in patients who remain MRD positive before or following HCT.47,48 

Mutations in N/KRAS genes correspond with inferior response and survival following HMA-VEN treatment (Table 1).11,21,49 Monocytic myeloid differentiation, particularly in the absence of co-occurring favorable mutations such as NPM1, also confers venetoclax resistance, often coexisting with mutations in active signaling genes.17,50 For this molecular subgroup, the incorporation of nucleoside analogs including fludarabine or cladribine (CLAD) is an increasingly recognized alternative to standard HMA-VEN combinations.25,50 

The triplet nucleoside regimen alternating cycles of CLAD, low-dose cytarabine (LDAC), and AZA-VEN resulted in CR/CRi rates of 93%, including 84% who were MRD negative measured using MFC (Table 2).25 After a follow-up of 22 months, the estimated 24-month OS rate was 72.9%. While inclusive of a younger patient population compared to VIALE-A (median age, 68 years) with high HCT utilization (34% proceeded to HCT in the first CR), these outcomes are similar to those observed in an IC-eligible population. A post-hoc analysis comparing CLAD/LDAC/VEN with HMA-VEN demonstrated a CR/CRi rate of 83% vs 45% in patients with N/KRAS-mutated AML.51 Updated phase 2 data demonstrated a median OS of 25 months for patients with intermediate- benefit AML based on the mPRS score, suggesting this regimen may improve survival compared to HMA-VEN in patients with active signaling mutations.52 A prospective study evaluating the efficacy of this combination specifically in active signaling and/or monocytic AML is planned (NCT06504459).

NPM1-mutated AML is characterized by cytarabine sensitivity, and thus older patients with NPM1-mutated AML historically experienced inferior outcomes compared to younger patients able to receive IC.9,11,19,53 However, two lower-intensity treatment options, LDAC-VEN (VIALE-C) and AZA-VEN (VIALE-A), resulted in CR/CRi rates of 78% and 66.7%, with the latter regimen associated with an MRD-negative (MFC) CR/CRi rate of 88%.9,54 Reported 2-year OS from a pooled analysis of patients receiving either LDAC-VEN or AZA-VEN was 72%, confirming the important role of venetoclax in older IC-ineligible patients with NPM1- mutated AML (Table 1).11 

As highlighted throughout this text, VEN-based regimens can effectively eradicate MRD, which has a demonstrable prognostic impact in patients receiving lower-intensity treatment.27,28,55 Using MRD-MFC (level of sensitivity, 10−3), 88% of evaluable patients enrolled in VIALE-A with NPM1-mutated AML attained MRD-negative CR/CRi; similarly high MRD-negative CR/CRi rates (58%-63%) using NPM1 MRD assays measured via quantitative reverse transcription PCR (RT-qPCR) (level of sensitivity, 10−4) have been reported.22,27,28 Undetectable MRD-MFC from bone marrow samples (from all included patients) following AZA-VEN corresponded with an estimated 12-month OS of 94%, while undetectable NPM1 MRD RT-qPCR following 4 cycles of either AZA-VEN or LDAC-VEN therapy corresponded to a 2-year OS of 84%.27,28 Of note, MRD responses with lower-intensity therapy may evolve slowly, with some patients becoming undetectable only after 6-7 cycles of therapy.27,28 

In venetoclax-naïve patients with persistent MRD-positive NPM1-mutated AML, MRD-targeted therapy using LDAC-VEN resulted in molecular clearance in 55% of patients with molecular relapse (defined as >1 log10 rise in MRD) and 75% of patients with oligoblastic relapse (ie, 5%-15% bone marrow myeloblasts).55 The corresponding 2-year OS of 63% compares favorably to reported outcomes following IC in this setting (2-year OS, 29%-61%).55 

Treatment-free remissions may even be a reality for select patients in this NPM1-mutated molecular subgroup who attain MRD negativity.28,56 After a median follow-up of 15 months after treatment cessation, treatment-free remissions in patients who had undetectable NPM1 MRD RT-qPCR from bone marrow samples was ongoing in 88%.28 Similar treatment-free remissions have been described for patients with co-occurring or isolated IDH2 mutations.56 

The inhibition of the menin-KMT2A axis represents an emergent class of therapeutics with activity in NPM1-mutated and KMT2A-rearranged (KMT2Ar) AML, molecular subclasses driven by shared HOX and MEIS1 gene upregulation.11,26 

In patients with R/R NPM1-mutated or KMT2Ar acute leukemias (60% of whom received prior venetoclax), revumenib treatment resulted in a CR plus CR with partial hematologic recovery rate of 30% (NPM1 mutated, 21%; KMT2Ar, 33%).26 Notably high rates of undetectable MRD-MFC were observed (78%) in responding patients, although responses to menin inhibitors are generally short-lived with a median DOR of 6 months, with emergent mutations in the MEN1 gene as one identified resistance mechanism limiting single-agent activity.26,57 Combining menin inhibitors with HMA-VEN in the frontline or RR setting may represent more effective treatment strategies (Table 2). While survival follow-up is premature, frontline therapy with AZA-VEN- revumenib resulted in an impressive composite CR rate of 100% (N  =  13/13), suggesting this regimen is highly active in NPM1 or KMT2A-rearranged AML.58 

The adverse prognostic impact of persistent MRD in AML provides a further rationale for the development of MRD-directed therapies, which can prevent morphologic relapse and improve survival.27,28,44,55 The ongoing INTERCEPT and MyeloMatch studies are investigating targeted MRD-directed interventions, while the development of antibody-based therapies such as bispecific CD3-CD123 T-cell engagers and CD123-targeting antibody-drug conjugates continues in this area.59–61 

IVO-AZA was selected as frontline therapy. A repeat bone marrow examination following 2 treatment cycles demonstrated a complete morphologic response, and after 4 cycles no IDH1 mutation was detectable. The patient remains in an MRD-negative remission to date.

AML treatment has evolved tremendously since the binary options of hypomethylating agent monotherapy vs induction chemotherapy. These emerging therapeutic options, along with the preemptive treatment of MRD, rationally designed treatment combinations and sequencing approaches, and increasingly individualized prognostication tools, will assuredly further improve the treatment and survival of older adults with AML.

Curtis A. Lachowiez is supported through an Oregon Clinical and Translational Research Institute KL2 award (KL2TR002370). Courtney D. DiNardo is supported by the LLS Scholar in Clinical Research Award.

Curtis A. Lachowiez: consultancy: AbbVie, Rigel, Servier, Bristol Myers Squibb, Syndax, COTA Healthcare; advisory board: AbbVie, Rigel, Servier, Bristol Myers Squibb, Syndax, COTA Healthcare.

Courtney D. DiNardo: research funding: AbbVie, Astex, Beigene, Bristol Myers Squibb, Foghorn, ImmuneOnc, Jazz, Rigel, Schrodinger, Servier; consultancy: AbbVie, AstraZeneca, Bristol Myers Squibb, Genentech, GenMab, GSK, Immunogen, Notable Labs, Rigel, Schrodinger, Servier; advisory board: AbbVie, AstraZeneca, Bristol Myers Squibb, Genentech, GenMab, GSK, Immunogen, Notable Labs, Rigel, Schrodinger, Servier.

Curtis A. Lachowiez: Off-label drug use will be discussed throughout the article (CAL and CDD).

Courtney D. DiNardo: Off-label drug use will be discussed throughout the article (CAL and CDD).

1.
Siegel
RL
,
Giaquinto
AN
,
Jemal
A.
Cancer statistics, 2024
.
CA Cancer J Clin
.
2024
;
74
(
1
):
12
-
49
.
2.
Juliusson
G
,
Antunovic
P
,
Derolf
A
, et al.
Age and acute myeloid leukemia: real world data on decision to treat and outcomes from the Swedish Acute Leukemia Registry
.
Blood
.
2009
;
113
(
18
):
4179
-
4187
.
3.
Kantarjian
H
,
Ravandi
F
,
O'Brien
S
, et al.
Intensive chemotherapy does not benefit most older patients (age 70 years or older) with acute myeloid leukemia
.
Blood
.
2010
;
116
(
22
):
4422
-
4429
.
4.
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
.
5.
Lindsley
RC
,
Mar
BG
,
Mazzola
E
, et al.
Acute myeloid leukemia ontogeny is defined by distinct somatic mutations
.
Blood
.
2015
;
125
(
9
):
1367
-
1376
.
6.
Murdock
HM
,
Kim
HT
,
Denlinger
N
, et al.
Impact of diagnostic genetics on remission MRD and transplantation outcomes in older patients with AML
.
Blood
.
2022
;
139
(
24
):
3546
-
3557
.
7.
Uy
GL
,
Newell
LF
,
Lin
TL
, et al.
Transplant outcomes after CPX-351 vs 7 + 3 in older adults with newly diagnosed high-risk and/or secondary AML
.
Blood Adv
.
2022
;
6
(
17
):
4989
-
4993
.
8.
Sorror
ML
,
Gooley
TA
,
Storer
BE
, et al.
An 8-year pragmatic observation evaluation of the benefits of allogeneic HCT in older and medically infirm patients with AML
.
Blood
.
2023
;
141
(
3
):
295
-
308
.
9.
DiNardo
CD
,
Jonas
BA
,
Pullarkat
V
, et al.
Azacitidine and venetoclax in previously untreated acute myeloid leukemia
.
N Engl J Med
.
2020
;
383
(
7
):
617
-
629
.
10.
Montesinos
P
,
Recher
C
,
Vives
S
, et al.
Ivosidenib and azacitidine in IDH1- mutated acute myeloid leukemia
.
N Engl J Med
.
2022
;
386
(
16
):
1519
-
1531
.
11.
DiNardo
CD
,
Tiong
IS
,
Quaglieri
A
, et al.
Molecular patterns of response and treatment failure after frontline venetoclax combinations in older patients with AML
.
Blood
.
2020
;
135
(
11
):
791
-
803
.
12.
Konopleva
M
,
Thirman
MJ
,
Pratz
KW
, et al.
Impact of FLT3 Mutation on outcomes after venetoclax and azacitidine for patients with treatment-naïve acute myeloid leukemia
.
Clin Cancer Res
.
2022
;
28
(
13
):
2744
-
2752
.
13.
Pollyea
DA
,
Pratz
KW
,
Wei
AH
, et al.
Outcomes in patients with poor-risk cytogenetics with or without TP53 mutations treated with venetoclax and azacitidine
.
Clin Cancer Res
.
2022
;
28
(
24
):
5272
-
5279
.
14.
Thijssen
R
,
Diepstraten
ST
,
Moujalled
D
, et al.
Intact TP-53 function is essential for sustaining durable responses to BH3-mimetic drugs in leukemias
.
Blood
.
2021
;
137
(
20
):
2721
-
2735
.
15.
Pei
S
,
Pollyea
DA
,
Gustafson
A
, et al.
Monocytic subclones confer resistance to venetoclax-based therapy in patients with acute myeloid leukemia
.
2020
;
10
(
4
):
536
-
551
.
16.
Zhang
H
,
Nakauchi
Y
,
Köhnke
T
, et al.
Integrated analysis of patient samples identifies biomarkers for venetoclax efficacy and combination strategies in acute myeloid leukemia
.
Nat Cancer
.
2020
;
1
(
8
):
826
-
839
.
17.
Waclawiczek
A
,
Leppä
AM
,
Renders
S
, et al.
Combinatorial BCL2 family expression in acute myeloid leukemia stem cells predicts clinical response to azacitidine/venetoclax
.
Cancer Discov
.
2023
;
13
(
6
):
1408
-
1427
.
18.
Pollyea
DA
,
DiNardo
CD
,
Arellano
ML
, et al.
Impact of venetoclax and azacitidine in treatment-naïve patients with acute myeloid leukemia and IDH1/2 mutations
.
Clin Cancer Res
.
2022
;
28
(
13
):
2753
-
2761
.
19.
Lachowiez
CA
,
Loghavi
S
,
Kadia
TM
, et al.
Outcomes of older patients with NPM1-mutated AML: current treatments and the promise of venetoclax-based regimens
.
Blood Adv
.
2020
;
4
(
7
):
1311
-
1320
.
20.
Bataller
A
,
Loghavi
S
,
Gerstein
Y
, et al.
Characteristics and clinical outcomes of patients with myeloid malignancies and DDX41 variants
.
Am J Hematol
.
2023
;
98
(
11
):
1780
-
1790
.
21.
Bataller
A
,
Bazinet
A
,
DiNardo
CD
, et al.
Prognostic risk signature in patients with acute myeloid leukemia treated with hypomethylating agents and venetoclax
.
Blood Adv
.
2024
;
8
(
4
):
927
-
935
.
22.
Gangat
N
,
Karrar
O
,
Iftikhar
M
, et al.
Venetoclax and hypomethylating agent combination therapy in newly diagnosed acute myeloid leukemia: genotype signatures for response and survival among 301 consecutive patients
.
Am J Hematol
.
2024
;
99
(
2
):
193
-
202
.
23.
Lachowiez
CA
,
Loghavi
S
,
Zeng
Z
, et al.
A phase Ib/II study of ivosidenib with venetoclax ± azacitidine in IDH1-mutated myeloid malignancies
.
Blood Cancer Discov
.
2023
;
4
(
4
):
276
-
293
.
24.
Short
NJ
,
Daver
N
,
Dinardo
CD
, et al.
Azacitidine, venetoclax, and gilteritinib in newly diagnosed and relapsed or refractory FLT3-mutated AML
.
J Clin Oncol
.
2024
;
42
(
13
):
1499
-
1508
.
25.
Kadia
TM
,
Reville
PK
,
Wang
X
, et al.
Phase II study of venetoclax added to cladribine plus low-dose cytarabine alternating with 5-azacitidine in older patients with newly diagnosed acute myeloid leukemia
.
J Clin Oncol
.
2022
;
40
(
33
):
3848
-
3857
.
26.
Issa
GC
,
Aldoss
I
,
DiPersio
J
, et al.
The menin inhibitor revumenib in KMT2A- rearranged or NPM1-mutant leukaemia
.
Nature
.
2023
;
615
(
7954
):
920
-
924
.
27.
Pratz
KW
,
Jonas
BA
,
Pullarkat
V
, et al.
Measurable residual disease response and prognosis in treatment-naïve acute myeloid leukemia with venetoclax and azacitidine
.
J Clin Oncol
.
2022
;
40
(
8
):
855
-
865
.
28.
Othman
J
,
Tiong
IS
,
O'Nions
J
, et al.
Molecular MRD is strongly prognostic in patients with NPM1-mutated AML receiving venetoclax-based nonintensive therapy
.
Blood
.
2024
;
143
(
4
):
336
-
341
.
29.
Lachowiez
CA
,
Long
N
,
Saultz
J
, et al.
Comparison and validation of the 2022 European LeukemiaNet guidelines in acute myeloid leukemia
.
Blood Adv
.
2023
;
7
(
9
):
1899
-
1909
.
30.
Döhner
H
,
Pratz
KW
,
DiNardo
CD
, et al.
Genetic risk stratification and outcomes among treatment-naive patients with AML treated with venetoclax and azacitidine
.
Blood
.
2024
.
31.
Grob
T
,
Al Hinai
ASA
,
Sanders
MA
, et al.
Molecular characterization of mutant TP53 acute myeloid leukemia and high-risk myelodysplastic syndrome
.
Blood
.
2022
;
139
(
15
):
2347
-
2354
.
32.
Döhner
H
,
Wei
AH
,
Appelbaum
FR
, et al.
Diagnosis and management of AML in adults: 2022 recommendations from an international expert panel on behalf of the ELN
.
Blood
.
2022
;
140
(
12
):
1345
-
1377
.
33.
Senapati
J
,
Urrutia
S
,
Loghavi
S
, et al.
Venetoclax abrogates the prognostic impact of splicing factor gene mutations in newly diagnosed acute myeloid leukemia
.
Blood
.
2023
;
142
(
19
):
1647
-
1657
.
34.
Lachowiez
CA
,
Loghavi
S
,
Furudate
K
, et al.
Impact of splicing mutations in acute myeloid leukemia treated with hypomethylating agents combined with venetoclax
.
Blood Adv
.
2021
;
5
(
8
):
2173
-
2183
.
35.
Shimony
S
,
Bewersdorf
JP
,
Shallis
RM
, et al.
Hypomethylating agents plus venetoclax compared with intensive induction chemotherapy regimens in molecularly defined secondary AML
.
Leukemia
.
2024
;
38
(
4
):
762
-
768
.
36.
Ustun
C
,
Le-Rademacher
J
,
Wang
HL
, et al.
Allogeneic hematopoietic cell transplantation compared to chemotherapy consolidation in older acute myeloid leukemia (AML) patients 60-75 years in first complete remission (CR1): an alliance (A151509), SWOG, ECOG-ACRIN, and CIBMTR study
.
Leukemia
.
2019
;
33
(
11
):
2599
-
2609
.
37.
Pollyea
DA
,
Winters
A
,
McMahon
C
, et al.
Venetoclax and azacitidine followed by allogeneic transplant results in excellent outcomes and may improve outcomes versus maintenance therapy among newly diagnosed AML patients older than 60
.
Bone Marrow Transplant
.
2022
;
57
(
2
):
160
-
166
.
38.
Pratz
KW
,
Jonas
BA
,
Pullarkat
V
, et al.
Long-term follow-up of VIALE-A: venetoclax and azacitidine in chemotherapy-ineligible untreated acute myeloid leukemia
.
Am J Hematol
.
2024
;
99
(
4
):
615
-
624
.
39.
De Botton
S
,
Montesinos
P
,
Vives Polo
S
, et al.
Updated efficacy and safety data from the AGILE study in patients with newly diagnosed acute myeloid leukemia treated with ivosidenib + azacitidine compared to placebo + azacitidine
.
J Clin Oncol
.
2023
;
41
(
suppl 16
):
7012
.
40.
DiNardo
CD
,
Stein
EM
,
de Botton
S
, et al.
Durable remissions with ivosidenib in IDH1-mutated relapsed or refractory AML
.
N Engl J Med
.
2018
;
378
(
25
):
2386
-
2398
.
40.
Hammond
D
,
Loghavi
S
,
Wang
SA
, et al.
Response patterns and impact of MRD in patients with IDH1/2-mutated AML treated with venetoclax and hypomethylating agents
.
Blood Cancer J
.
2023
;
13
(
1
):
148
.
41.
Bewersdorf
JP
,
Shallis
RM
,
Derkach
A
, et al.
Efficacy of FLT3 and IDH1/2 inhibitors in patients with acute myeloid leukemia previously treated with venetoclax
.
Leuk Res
.
2022
;
122
(
9
):
106942
.
42.
Atluri
H
,
Mullin
J
,
Takahashi
K
, et al.
Phase Ib/2 study of oral decitabine/ cedazuridine (ASTX727) and venetoclax in combination with the targeted mutant IDH1 inhibitor ivosidenib or the targeted mutant IDH2 inhibitor enasidenib: 2023 update
.
Blood
.
2023
;
142
:
968
.
43.
DiNardo
CD
,
Schuh
AC
,
Stein
EM
, et al.
Enasidenib plus azacitidine versus azacitidine alone in patients with newly diagnosed, mutant-IDH2 acute myeloid leukaemia (AG221-AML-005): a single-arm, phase 1b and randomised, phase 2 trial
.
Lancet Oncol
.
2021
;
22
(
11
):
1597
-
1608
.
44.
Dillon
LW
,
Gui
G
,
Page
KM
, et al.
DNA sequencing to detect residual disease in adults with acute myeloid leukemia prior to hematopoietic cell transplant
.
JAMA
.
2023
;
329
(
9
):
745
-
755
.
45.
Daver
N
,
Perl
AE
,
Maly
J
, et al.
Venetoclax plus gilteritinib for FLT3- mutated relapsed/refractory acute myeloid leukemia
.
J Clin Oncol
.
2022
;
40
(
35
):
4048
-
4059
.
46.
Yilmaz
M
,
Muftuoglu
M
,
DiNardo
CD
, et al.
Phase I/II study of quizartinib, venetoclax, and decitabine triple combination in FLT3-ITD mutated AML
.
Blood
.
2023
;
142
:
158
.
47.
Othman
J
,
Potter
N
,
Mokretar
K
, et al.
FLT3 inhibitors as MRD-guided salvage treatment for molecular failure in FLT3 mutated AML
.
Leukemia
.
2023
;
37
(
10
):
2066
-
2072
.
48.
Levis
MJ
,
Hamadani
M
,
Logan
BR
, et al.
Post-hoc analysis of measurable residual disease from BMT-CTN 1506/morpho: FLT3-ITD variant allele frequency and survival are highly correlated
.
Blood
.
2023
;
142
:
973
.
49.
Eide
CA
,
Kurtz
SE
,
Kaempf
A
, et al.
Clinical correlates of venetoclax-based combination sensitivities to augment acute myeloid leukemia therapy
.
Blood Cancer Discov
.
2023
;
4
(
6
):
452
-
467
.
50.
Pei
S
,
Shelton
IT
,
Gillen
AE
, et al.
A novel type of monocytic leukemia stem cell revealed by the clinical use of venetoclax-based therapy
.
Cancer Discov
.
2023
;
13
(
9
):
2032
-
2049
.
51.
Rivera
D
,
Kim
K
,
Kanagal-Shamanna
R
, et al.
Implications of RAS mutational status in subsets of patients with newly diagnosed acute myeloid leukemia across therapy subtypes
.
Am J Hematol
.
2022
;
97
(
12
):
1599
-
1606
.
52.
Bataller
A
,
Bazinet
A
,
Borthakur
G
, et al.
Cladribine with low dose cytarabine and venetoclax alternated with azacytidine and venetoclax for acute myeloid leukemia: prognostic analysis of a phase 2 clinical trial
.
Blood
.
2023
;
142
:
4256
.
53.
Ostronoff
F
,
Othus
M
,
Lazenby
M
, et al.
Prognostic significance of NPM1 mutations in the absence of FLT3-internal tandem duplication in older patients with acute myeloid leukemia: a SWOG and UK National Cancer Research Institute/Medical Research Council Report
.
J Clin Oncol
.
2015
;
33
(
10
):
1157
-
1164
.
54.
Wei
AH
,
Montesinos
P
,
Ivanov
V
, et al.
Venetoclax plus LDAC for newly diagnosed AML ineligible for intensive chemotherapy: a phase 3 randomized placebo-controlled trial
.
Blood
.
2020
;
135
(
24
):
2137
-
2145
.
55.
Tiong
IS
,
Hiwase
DK
,
Abro
E
, et al.
Targeting molecular measurable residual disease and low-blast relapse in AML with venetoclax and low-dose cytarabine: a prospective phase II study (VALDAC)
.
J Clin Oncol
.
2024
;
42
(
18
):
2161
-
2173
.
56.
Chua
CC
,
Hammond
D
,
Kent
A
, et al.
Treatment-free remission after ceasing venetoclax-based therapy in patients with acute myeloid leukemia
.
Blood Adv
.
2022
;
6
(
13
):
3879
-
3883
.
57.
Perner
F
,
Stein
EM
,
Wenge
DV
, et al.
MEN1 mutations mediate clinical resistance to menin inhibition
.
Nature
.
2023
;
615
(
7954
):
913
-
919
.
58.
Issa
GC
,
Cuglievan
B
,
DiNardo
CD
, et al.
Early results of the phase I/II study investigating the all-oral combination of the menin inhibitor revumenib (SNDX-5613) with decitabine/cedazuridine (ASTX727) and venetoclax in acute myeloid leukemia (SAVE)
.
Blood
.
2023
;
142
:
58
.
59.
Daver
NG
,
Montesinos
P
,
DeAngelo
DJ
, et al.
Pivekimab sunirine (IMGN632), a novel CD123-targeting antibody–drug conjugate, in relapsed or refractory acute myeloid leukaemia: a phase 1/2 study
.
Lancet Oncol
.
2024
;
25
(
3
):
388
-
399
.
60.
Ravandi
F
,
Bashey
A
,
Foran
J
, et al.
Phase 1 study of vibecotamab identifies an optimized dose for treatment of relapsed/refractory acute myeloid leukemia
.
Blood Adv
.
2023
;
7
(
21
):
6492
-
6505
.
61.
Wei
AH
,
Iland
HJ
,
Reynolds
J
, et al.
ALLG AMLM26 phase 1B/2 study investigating novel therapies to target early relapse and clonal evolution as pre-emptive therapy in AML (INTERCEPT): a multi-arm, precision-based, recursive, platform trial
.
Blood
.
2022
;
140
(
suppl 1
):
3341
-
3343
.
62.
Zeidner
J
,
Lin
TL
,
Welkie
R
, et al.
Phase 1b study of azacitidine, venetoclax and revumenib in newly diagnosed older adults with NPM1 mutated or KMT2A rearranged AML: interm results of dose escalation from the Beat AML Consortium
.
EHA Library
.
13
June
2024
;422238. Abstract S134.
63.
Norsworthy
KJ
,
Mulkey
F
,
Scott
EC
, et al.
Differentiation syndrome with ivosidenib and enasidenib treatment in patients with relapsed or refractory IDH-mutated AML: a US Food and Drug Administration systematic analysis
.
Clinical Cancer Research
.
2020
;
26
(
16
):
4280
88
.
64.
Fathi
AT
,
DiNardo
CD
,
Kline
I
, et al
.
AG221-C-001 Study Investigators. Differentiation syndrome associated with enasidenib, a selective inhibitor of mutant isocitrate dehydrogenase 2: analysis of a phase 1/2 study
.
JAMA Oncology
.
2018
;
4
(
8
):
1106
10
.
65.
de Botton
S
,
Fenaux
P
,
Yee
K
, et al
.
Olutasidenib (FT-2102) induces durable complete remissions in patients with relapsed or refractory IDH1-mutated AML
.
Blood Advances
.
2023
;
7
(
13
):
3117
27
.
66.
Fathi
AT
,
Stein
EM
,
DiNardo
CD
, et al.
Differentiation syndrome with lower- intensity treatments for acute myeloid leukemia
.
American Journal of Hematology
.
2021
;
96
(
6
):
735
46
.
67.
Roboz
GJ
,
DiNardo
CD
,
Stein
EM
, et al.
Ivosidenib induces deep durable remissions in patients with newly diagnosed IDH1-mutant acute myeloid leukemia
.
Blood, The Journal of the American Society of Hematology
.
2020
;
135
(
7
):
463
71
.
68.
Cortes
JE
,
Khaled
S
,
Martinelli
G
, et al
.
Quizartinib versus salvage chemotherapy in relapsed or refractory FLT3-ITD acute myeloid leukaemia (QuANTUM-R): a multicentre, randomised, controlled, open-label, phase 3 trial
.
The Lancet Oncology
.
2019
;
20
(
7
):
984
97
.
69.
Stein
EM
,
DiNardo
CD
,
Pollyea
DA
, et al.
Enasidenib in mutant IDH2 relapsed or refractory acute myeloid leukemia
.
Blood, The Journal of the American Society of Hematology
.
2017
;
130
(
6
):
722
31
.