• Medically fit patients with newly diagnosed adverse-risk AML may benefit from treatment with aza-ven (IC, 1.4 QALYs vs aza-ven, 2.0 QALYs).

  • IC remains the preferred induction regimen for patients with intermediate-risk AML.

Abstract

Although induction chemotherapy (IC) is the standard of care in medically fit patients with newly diagnosed acute myeloid leukemia (AML), limited retrospective data indicate that patients at adverse-risk may benefit from azacytidine and venetoclax (aza-ven). Our goal was to perform a Markov decision analysis to determine whether IC or aza-ven is the optimal induction regimen in this population. Using the TreeAge software, Markov models were created for adverse-risk and intermediate-risk cohorts. A systematic review of the literature informed the transition probabilities and utilities included in the analyses. Our primary outcome was quality-adjusted life years (QALYs) gained over 5 years after diagnosis. Overall, patients at adverse risk treated with IC gained 1.4 QALYs, compared with 2.0 QALYs in patients treated with aza-ven. Patients at adverse risk treated with IC and allogeneic stem cell transplantation (allo-SCT), IC, aza-ven and allo-SCT, or aza-ven gained 2.1, 1.5, 3.0, and 1.9 QALYs, respectively. Meanwhile, patients at intermediate risk treated with IC gained 2.0 QALY, compared with 1.7 QALY in patients treated with aza-ven. Patients at intermediate risk treated with IC and allo-SCT, IC, aza-ven and allo-SCT, and aza-ven gained 2.7, 2.3, 2.6, and 1.8 QALYs, respectively. We have demonstrated that medically fit patients with newly diagnosed adverse-risk AML may benefit from treatment with aza-ven over those treated with IC, whereas IC remains the preferred approach for patients at intermediate risk. Our work challenges the use of the European LeukemiaNet risk classification for patients treated with aza-ven and highlights the need for prospective investigation into aza-ven as induction therapy for medically fit patients.

Induction chemotherapy (IC) is the standard of care for younger, medically fit patients with newly diagnosed acute myeloid leukemia (AML).1 It is well established that IC is an intensive chemotherapeutic regimen that requires prolonged hospitalization and carries a significant risk of morbidity and mortality. Age, comorbidities, performance status, and frailty are all assessed to determine whether a patient would benefit from IC.2,3 Typically, IC is not offered to those aged >75 years, which presents a significant challenge, given that the median age at diagnosis of AML is 69 years.4 

Historically, older or unfit patients with AML were treated with hypomethylating agents (eg, azacitidine [aza]), other nonintensive therapies such as low-dose cytarabine, or best supportive care.5 VIALE-A, a recent phase 3, multicenter randomized controlled trial compared aza combined with the B-cell lymphoma 2 protein inhibitor venetoclax (aza-ven) with aza alone.6 Aza-ven was found to have a significantly greater composite complete remission (CR) rate (66.4% vs 28.3%) and a significant improvement in overall survival (OS; (14.7 months vs 9.6 months).

The median age of the VIALE-A population was 76 years, and, by trial design, these patients were ineligible for treatment with IC. Whether IC or aza-ven is a superior approach to achieving CR and proceeding to allogeneic stem cell transplantation (allo-SCT) in medically fit patients remains unknown. A retrospective cohort study comparing IC and aza-ven found an unadjusted median OS of 884 days vs 483 days, respectively.7 In the unadjusted analysis, the patients treated with aza-ven were unsurprisingly older and more comorbid. However, in a propensity-matched analysis, patients with adverse-risk AML were found to have a preferential benefit when treated with aza-ven.7 

Randomized controlled trials comparing these regimens are ongoing, with results expected in 2026.8 In the interim, an evidence-based decision tool that enables risk-stratified comparisons between IC and aza-ven would be valuable. The objective of this study was to perform a Markov decision analysis to determine the optimal induction regimen (IC vs aza-ven) for a theoretical cohort of medically fit IC-eligible patients with newly diagnosed AML.

Selection of a Markov model

A Markov model was selected for our analysis because this approach effectively accounts for risk that varies over the study period (eg, risk of relapse is present throughout our 5-year study period but is different at year 1 vs year 5).9 This analysis is particularly useful for AML, in which relapse within year 1 is prognostically different from relapse at year 5, and the timing of relapse has a major impact on the quality-adjusted life years (QALYs) accrued. Additionally, Markov models effectively capture events that can occur multiple times within the study period (eg, relapse).9 Please see supplemental Materials for further details regarding the construction of our Markov model.

There is precedent for the use of Markov analyses in hematology. Cutler et al10 examined the optimal timing of allo-SCT for patients with myelodysplastic syndrome, and Kurosawa et al11 studied allo-SCT vs chemotherapy for patients with AML who had achieved first remission. Markov analyses have also been used to assess the cost-effectiveness of different treatment approaches. Yamamoto et al12 compared the incremental cost-effectiveness ratio of various tyrosine kinase inhibitors for the treatment of CML, and Slot et el13 compared the incremental cost-effectiveness ratio of ven-obinutuzumab vs ibrutinib for the treatment of chronic lymphocytic leukemia.

Patients and interventions

A systematic review of the literature was conducted to inform the probabilities of each outcome within the Markov analyses. Specifically, PubMed, Medline, and Embase were searched for prospective or retrospective studies reporting the outcomes of patients with newly diagnosed AML treated with IC or aza-ven, with or without allo-SCT. Reference lists of relevant studies as well as abstracts from the most recent meeting of the American Society of Hematology were manually reviewed to identify studies not extracted in the systematic search. Given the target population of patients eligible for IC, emphasis was placed on studies with a median age of between 60 and 70 years.

Model design

Using the TreeAge Pro Healthcare modeling software (version 2023 R1.2), 2 Markov models were generated: 1 for patients at adverse risk and 1 for patients at intermediate risk. Patients were stratified based on the 2017 European Leukemia Network classification system.14,15 A model for favorable-risk disease was omitted, given a paucity of data on the use of aza-ven in this group and the superior outcomes with IC in younger patients with favorable-risk disease.

A simplified schematic of the Markov model is shown in Figure 1. Each model examines 2 theoretical cohorts of 1000 patients with newly diagnosed AML: 1 cohort comprising patients treated with IC and the other comprising patients treated with aza-ven. Disease states after induction included composite CR (CR, or CR with incomplete hematologic recovery), primary refractory disease (no CR after 2 courses of intensive induction treatment), or early death (from any cause within 30 days).14-16 Those who achieved CR either did or did not receive allo-SCT, and both groups either remained in disease-free survival (DFS), transitioned to relapse, or proceeded to death (from any cause after 30 days). Patients who relapsed either transitioned to a second CR (CR2) or died. Patients with primary refractory disease either transitioned to CR or died. Our primary outcome was the QALYs gained over a time horizon of 5 years after initial diagnosis.

Figure 1.

A simplified schematic of the Markov model design. This figure outlines the health states included in the analysis and the possible transitions over time. CR, composite complete remission; RD, refractory disease.

Figure 1.

A simplified schematic of the Markov model design. This figure outlines the health states included in the analysis and the possible transitions over time. CR, composite complete remission; RD, refractory disease.

Close modal

Transition probabilities

Five-year OS and relapse-free survival curves extracted from the literature search were digitized to allow for accurate year-to-year variability in risk of death and relapse, respectively. Other discrete events, such as the proportion of patients who achieve CR proceeding allo-SCT, were reported as a single value. A final weighted average accounting for the sample sizes from the referenced studies was calculated for each variable and used in the model. The final weighted data used for the adverse-risk and intermediate-risk models can be found in Table 1.

Table 1.

Patient demographics by weighted averages

Adverse-risk modelIntermediate-risk model
VariableAza-venICVariableAza-venIC
Age, y 66.9 58.3 Age, y 72.4 55.2 
Male (%) 0.517 0.528 Male (%) 0.57 0.486 
Female (%) 0.483 0.472 Female (%) 0.43 0.514 
Bone marrow blasts (%) 0.502 0.66 Bone marrow blasts (%) 0.5 0.809 
Secondary AML (%) 0.459 0.082 Secondary AML (%) 0.3 0.09 
Treatment-related AML (%) 0.212 0.07 Treatment-related AML (%) 0.264 0.06 
Adverse-risk modelIntermediate-risk model
VariableAza-venICVariableAza-venIC
Age, y 66.9 58.3 Age, y 72.4 55.2 
Male (%) 0.517 0.528 Male (%) 0.57 0.486 
Female (%) 0.483 0.472 Female (%) 0.43 0.514 
Bone marrow blasts (%) 0.502 0.66 Bone marrow blasts (%) 0.5 0.809 
Secondary AML (%) 0.459 0.082 Secondary AML (%) 0.3 0.09 
Treatment-related AML (%) 0.212 0.07 Treatment-related AML (%) 0.264 0.06 

Health utilities

Each disease state was assigned a QALY utility derived from the literature (eg, adverse-risk AML DFS vs relapsed disease). The Markov model determined how many patients were in a particular disease state and for how long. Thus, the QALYs gained in the IC arm was compared with that in the aza-ven arm, for both adverse- and intermediate-risk disease. The QALY utilities for patients treated with IC and aza-ven were identified as part of our systematic review and can be found in Table 2.

Table 2.

Health state transition probabilities for the adverse- and intermediate-risk Markov decision analyses

Health state probabilityWeighted averageReferences
Adverse-risk model   
Relapse with AZ and SCT (year-by-year data) 0.227, 0.030, 0, 0, and 0 Pollyea et al17 and Winters et al18  
Death after relapse with AZ and SCT 0.62 Pollyea et al17 and Salhotra et al19  
Death within the first year of AZ and SCT 0.152 Pollyea et al,17 Winters et al,18 and Salhotra et al 19  
SCT after induction with AZ 0.343 Cherry et al,7 Pollyea et al,17 and Salhotra et al 19  
No SCT after induction with AZ 0.425 Cherry et al,7 Pollyea et al,17 and Salhotra et al19  
Early death with AZ 0.049 Cherry et al7  
Relapse with AZ and no SCT (year-by-year data) 0.51, 0.28, 0.2, 0.18, and 0 Cherry et al7  
Death after relapse with AZ and no SCT 0.784 Garciaz et al20 and Johnson et al21  
Death within the first year of AZ and no SCT 0.42 Cherry et al7  
Composite CR with AZ after primary refractory disease 0.216 Garciaz et al20 and Johnson et al21  
Relapse with IC and SCT (year-by-year data) 0.507, 0.147, 0.106, 0, and 0.158 Bataller et al,22 Herold et al,23 and Hansen et al24  
Death after relapse with IC and SCT 0.727 Burnett et al25 and Herold et al23  
Death within the first year of IC and SCT 0.423 Bataller et al,22 Herold et al,23 Hansen et al24  
SCT after induction with IC 0.351 Bataller et al,22 Burnett et al,25 and Lo et al26  
No SCT after induction with IC 0.355 Bataller et al,22 Burnett et al,25 and Lo et al26  
Early death with IC 0.102 Bataller et al22  
Relapse with IC and no SCT (year-by-year data) 0.652, 0.352, 0.110, 0.120, and 0 Herold et al23 and Burnett et al25  
Death after relapse with IC and no SCT 0.794 Herold et al23 and Burnett et al25  
Death within the first year of IC and no SCT 0.564 Herold et al23 and Rausch et al27  
Composite CR with IC after primary refractory disease 0.66 Bataller et al22  
Intermediate-risk model   
Relapse with AZ and SCT (year-by-year data) 0.42, 0, 0, 0, 0 Pasvolsky et al28  
Death after relapse with AZ and SCT 0.421 Pasvolsky et al28  
Death within the first year of AZ and SCT 0.368 Pasvolsky et al28  
SCT after induction with AZ 0.317 Pollyea et al,29 Pollyea et al,17 and Salhotra et al19  
No SCT after induction with AZ 0.392 Pollyea et al,29 Pollyea et al,17 and Salhotra et al19  
Early death with AZ 0.049 Cherry et al7  
Relapse with AZ and no SCT (year-by-year data) 0.507, 0.432, 0.129, 0, 0 Pollyea et al29 and DiNardo et al5  
Death after relapse with AZ and no SCT 0.763 Garciaz et al20 and Johnson et al21  
Death within the first year of AZ and no SCT 0.444 Pollyea et al29 and DiNardo et al5  
Composite CR with AZ after primary refractory disease 0.237 Garciaz et al20 and Johnson et al21  
Relapse with IC and SCT (year-by-year data) 0.359, 0.111, 0.088, 0.046, and 0.018 Batalleret al,22 Herold et al,23 and Hansen et al 24  
Death after relapse with IC and SCT 0.576 Burnett et al25 and Herold et al23  
Death within the first year of IC and SCT 0.283 Bataller et al,22 Herold et al,23 and Hansen et al24  
SCT after induction with IC 0.263 Bataller et al,22 Burnett et al,25 and Lo26 ASH 2022 
No SCT after induction with IC 0.535 Bataller et al,22 Burnett et al,25 and Lo26 ASH 2022 
Early death with IC 0.119 Bataller et al22  
Relapse with IC and no SCT (year-by-year data) 0.394, 0.210, 0.083, 0.065, 0.030 Herold et al23 and Burnett et al25  
Death after relapse with IC and no SCT 0.674 Herold et al23 and Burnett et al25  
Death within the first year of IC and no SCT 0.392 Herold et al23 and Rausch et al27  
Composite CR with IC after primary refractory disease 0.667 Bataller et al22  
Health state probabilityWeighted averageReferences
Adverse-risk model   
Relapse with AZ and SCT (year-by-year data) 0.227, 0.030, 0, 0, and 0 Pollyea et al17 and Winters et al18  
Death after relapse with AZ and SCT 0.62 Pollyea et al17 and Salhotra et al19  
Death within the first year of AZ and SCT 0.152 Pollyea et al,17 Winters et al,18 and Salhotra et al 19  
SCT after induction with AZ 0.343 Cherry et al,7 Pollyea et al,17 and Salhotra et al 19  
No SCT after induction with AZ 0.425 Cherry et al,7 Pollyea et al,17 and Salhotra et al19  
Early death with AZ 0.049 Cherry et al7  
Relapse with AZ and no SCT (year-by-year data) 0.51, 0.28, 0.2, 0.18, and 0 Cherry et al7  
Death after relapse with AZ and no SCT 0.784 Garciaz et al20 and Johnson et al21  
Death within the first year of AZ and no SCT 0.42 Cherry et al7  
Composite CR with AZ after primary refractory disease 0.216 Garciaz et al20 and Johnson et al21  
Relapse with IC and SCT (year-by-year data) 0.507, 0.147, 0.106, 0, and 0.158 Bataller et al,22 Herold et al,23 and Hansen et al24  
Death after relapse with IC and SCT 0.727 Burnett et al25 and Herold et al23  
Death within the first year of IC and SCT 0.423 Bataller et al,22 Herold et al,23 Hansen et al24  
SCT after induction with IC 0.351 Bataller et al,22 Burnett et al,25 and Lo et al26  
No SCT after induction with IC 0.355 Bataller et al,22 Burnett et al,25 and Lo et al26  
Early death with IC 0.102 Bataller et al22  
Relapse with IC and no SCT (year-by-year data) 0.652, 0.352, 0.110, 0.120, and 0 Herold et al23 and Burnett et al25  
Death after relapse with IC and no SCT 0.794 Herold et al23 and Burnett et al25  
Death within the first year of IC and no SCT 0.564 Herold et al23 and Rausch et al27  
Composite CR with IC after primary refractory disease 0.66 Bataller et al22  
Intermediate-risk model   
Relapse with AZ and SCT (year-by-year data) 0.42, 0, 0, 0, 0 Pasvolsky et al28  
Death after relapse with AZ and SCT 0.421 Pasvolsky et al28  
Death within the first year of AZ and SCT 0.368 Pasvolsky et al28  
SCT after induction with AZ 0.317 Pollyea et al,29 Pollyea et al,17 and Salhotra et al19  
No SCT after induction with AZ 0.392 Pollyea et al,29 Pollyea et al,17 and Salhotra et al19  
Early death with AZ 0.049 Cherry et al7  
Relapse with AZ and no SCT (year-by-year data) 0.507, 0.432, 0.129, 0, 0 Pollyea et al29 and DiNardo et al5  
Death after relapse with AZ and no SCT 0.763 Garciaz et al20 and Johnson et al21  
Death within the first year of AZ and no SCT 0.444 Pollyea et al29 and DiNardo et al5  
Composite CR with AZ after primary refractory disease 0.237 Garciaz et al20 and Johnson et al21  
Relapse with IC and SCT (year-by-year data) 0.359, 0.111, 0.088, 0.046, and 0.018 Batalleret al,22 Herold et al,23 and Hansen et al 24  
Death after relapse with IC and SCT 0.576 Burnett et al25 and Herold et al23  
Death within the first year of IC and SCT 0.283 Bataller et al,22 Herold et al,23 and Hansen et al24  
SCT after induction with IC 0.263 Bataller et al,22 Burnett et al,25 and Lo26 ASH 2022 
No SCT after induction with IC 0.535 Bataller et al,22 Burnett et al,25 and Lo26 ASH 2022 
Early death with IC 0.119 Bataller et al22  
Relapse with IC and no SCT (year-by-year data) 0.394, 0.210, 0.083, 0.065, 0.030 Herold et al23 and Burnett et al25  
Death after relapse with IC and no SCT 0.674 Herold et al23 and Burnett et al25  
Death within the first year of IC and no SCT 0.392 Herold et al23 and Rausch et al27  
Composite CR with IC after primary refractory disease 0.667 Bataller et al22  

AZ, azacitidine and venetoclax

Sensitivity analyses

One-way sensitivity analyses were generated for each variable included in the model, and threshold values were reported. If the threshold value was not clinically plausible, then the analysis was considered insensitive to that variable.

Patient characteristics

Based on weighted averages from the studies included in our analysis, in the adverse-risk model, the median age of patients treated with aza-ven or IC was 67 and 58 years, respectively. In the intermediate-risk group, the median age of patients treated with aza-ven or IC was 72 and 55 years, respectively.

Adverse-risk analysis

Among patients with adverse-risk AML, aza-ven was superior to IC for the primary outcome. Overall, patients treated with IC gained 1.42 QALYs over 5 years from the time of diagnosis, compared with 1.97 QALYs among patients treated with aza-ven. On subgroup analysis, patients who achieved CR with IC gained 2.15 QALYs after proceeding to allo-SCT vs 1.50 QALYs without allo-SCT. Those treated with aza-ven in CR gained 3.00 QALYs after allo-SCT compared with 1.92 QALYs without allo-SCT.

Figures 2A-D shows how the proportions of patients with adverse-risk disease in states of DFS, relapse, CR2, or death changed over 5 years. For patients treated with IC who underwent allo-SCT, mortality was 42.3% and 59.0% at 1 and 5 years, respectively (Figure 2A). For patients treated with IC without allo-SCT, mortality was 56.4% and 79.8% at 1 and 5 years, respectively (Figure 2B). For patients treated with aza-ven who underwent allo-SCT, mortality was 15.2% and 32.0% at 1 and 5 years, respectively (Figure 2C), whereas for those who did not undergo allo-SCT, mortality was 42% and 72.4% at 1 and 5 years, respectively (Figure 2D).

Figure 2.

Rates of DFS, relapse, CR2, and death over 5 years from the time of AML diagnosis for patients with adverse-risk disease. (A) IC and allo-SCT, (B) IC without allo-SCT, (C) aza-ven and allo-SCT, and (D) aza-ven without allo-SCT.

Figure 2.

Rates of DFS, relapse, CR2, and death over 5 years from the time of AML diagnosis for patients with adverse-risk disease. (A) IC and allo-SCT, (B) IC without allo-SCT, (C) aza-ven and allo-SCT, and (D) aza-ven without allo-SCT.

Close modal

Intermediate-risk analysis

Among patients with intermediate-risk AML, IC was superior to aza-ven for the primary outcome. Overall, patients treated with IC gained 2.05 QALYs, compared with 1.69 QALYs among patients treated with aza-ven. Patients who achieved CR with IC followed by allo-SCT gained 2.74 QALYs, whereas those who did not receive allo-SCT gained 2.35 QALYs. Patients who achieved CR with aza-ven followed by allo-SCT gained 2.59 QALYs, whereas those who did not receive allo-SCT gained 1.84 QALYs. Health utilities values for both the adverse and intermediate risk models can be found in Table 3.

Table 3.

Health utilities for the adverse- and intermediate-risk Markov decision analyses

UtilityValueReferences
Adverse- and intermediate-risk model   
DFS if treated with AZ 0.8 Patel et al30  
Relapse if treated with AZ 0.67 Patel et al30  
DFS if treated with IC 0.83 Tremblay et al31  
Relapse if treated with IC 0.53 Tremblay et al31  
UtilityValueReferences
Adverse- and intermediate-risk model   
DFS if treated with AZ 0.8 Patel et al30  
Relapse if treated with AZ 0.67 Patel et al30  
DFS if treated with IC 0.83 Tremblay et al31  
Relapse if treated with IC 0.53 Tremblay et al31  

AZ, azacitidine and venetoclax

Figure 3A-D shows how the proportions of patient with intermediate-risk AML in states of DFS, relapse, CR2, or death changed over 5 years. For patients treated with IC who underwent allo-SCT, mortality was 28.3% and 43.5% at 1 and 5 years, respectively (Figure 3A). For patients treated with IC without allo-SCT, mortality was 39.2% and 55.5% at 1 and 5 years, respectively (Figure 3B). For patients treated with aza-ven who underwent allo-SCT, mortality was 36.8% and 40.0% at 1 and 5 years, respectively (Figure 3C), whereas in those who did not proceed to allo-SCT, mortality was 44.4% and 72.0% at 1 and 5 years, respectively (Figure 3D).

Figure 3.

Rates of DFS, relapse, CR2, and death over 5 years from the time of AML diagnosis for patients with intermediate-risk disease. (A) IC and allo-SCT, (B) IC without allo-SCT, (C) aza-ven and allo-SCT, and (D) aza-ven without allo-SCT.

Figure 3.

Rates of DFS, relapse, CR2, and death over 5 years from the time of AML diagnosis for patients with intermediate-risk disease. (A) IC and allo-SCT, (B) IC without allo-SCT, (C) aza-ven and allo-SCT, and (D) aza-ven without allo-SCT.

Close modal

Sensitivity analyses

The 1-way sensitivity analysis threshold values for each of the variables included in both the adverse and intermediate-risk analyses are shown in Table 4. Although the base-case analysis favored aza-ven in the adverse-risk group, IC was favored if >44% of patients treated with aza-ven and allo-SCT died within the first year or if the aza-ven DFS health utility was <0.56. Similarly, although the base-case analysis favored IC in the intermediate-risk group, aza-ven was favored if the proportion of patients treated with IC and allo-SCT who died within the first year was >39% or if the IC DFS health utility was <0.68.

Table 4.

Threshold values for the 1-way sensitivity analyses from the adverse- and intermediate-risk Markov models

Health state probability or utilityAdverse-risk favors ICIntermediate-risk favors AZ
Relapse with AZ and SCT — — 
Death after relapse with AZ and SCT — — 
Death within the first year of AZ and SCT >0.44 <0.05 
SCT after induction with AZ <0.11 >0.5 
Relapse with AZ and no SCT — — 
Death after relapse with AZ and no SCT — — 
Death within the first year of AZ and no SCT — <0.05 
Composite CR with AZ after primary refractory disease — >0.8 
Relapse with IC and SCT — >0.89 
Death after relapse with IC and SCT — — 
Death within the first year of IC and SCT <0.01 >0.39 
SCT after induction with IC — <0.07 
Relapse with IC and no SCT — >0.82 
Death after relapse with IC and no SCT — — 
Death within the first year with IC and no SCT <0.02 — 
Composite CR with IC after primary refractory disease — — 
Health utility of DFS if treated with AZ <0.56 >0.98 
Health utility of relapse if treated with AZ — — 
Health utility of DFS if treated with IC — <0.68 
Health utility of relapse if treated with IC — — 
Health state probability or utilityAdverse-risk favors ICIntermediate-risk favors AZ
Relapse with AZ and SCT — — 
Death after relapse with AZ and SCT — — 
Death within the first year of AZ and SCT >0.44 <0.05 
SCT after induction with AZ <0.11 >0.5 
Relapse with AZ and no SCT — — 
Death after relapse with AZ and no SCT — — 
Death within the first year of AZ and no SCT — <0.05 
Composite CR with AZ after primary refractory disease — >0.8 
Relapse with IC and SCT — >0.89 
Death after relapse with IC and SCT — — 
Death within the first year of IC and SCT <0.01 >0.39 
SCT after induction with IC — <0.07 
Relapse with IC and no SCT — >0.82 
Death after relapse with IC and no SCT — — 
Death within the first year with IC and no SCT <0.02 — 
Composite CR with IC after primary refractory disease — — 
Health utility of DFS if treated with AZ <0.56 >0.98 
Health utility of relapse if treated with AZ — — 
Health utility of DFS if treated with IC — <0.68 
Health utility of relapse if treated with IC — — 

AZ, azacitidine and venetoclax

Using a Markov analysis, we demonstrate that induction-eligible patients with newly diagnosed adverse-risk AML may benefit from treatment with aza-ven over IC. Conversely, our model indicates that IC remains the preferred approach for patients with intermediate-risk AML.

Our results are aligned with previously reported data from a propensity-matched analysis with a robust sample size (IC, n = 149; aza-ven, n = 143).7 The authors demonstrated improved OS among patients with adverse-risk disease treated with aza-ven, whereas those with intermediate-risk disease had improved OS if treated with IC. Our results mirror these data with regard to QALYs. Over 5 years, patients with adverse-risk disease treated with aza-ven gained 1.97 QALYs compared with 1.42 QALYs among those treated with IC. Conversely, patients with intermediate-risk disease treated with IC gained 2.05 QALYs compared with 1.69 QALYs among those treated with aza-ven.

In another recent retrospective, propensity-matched analysis comparing OS and DFS of patients (median age, 69 years) treated with IC vs aza-ven, intermediate-risk disease favored IC, whereas TP53-mutated AML favored aza-ven.32 This study was omitted from our model because the majority of patients in both the IC and aza-ven arms (58.7% and 63.0%, respectively) had unknown cytogenetics. The authors of that study reported no statistically significant differences in OS and DFS between IC and aza-ven, despite a higher rate of CR in the IC group. Although it is challenging to interpret these results with substantial missing cytogenetic data, the findings support a subgroup of patients at higher risk who may benefit from treatment with aza-ven.

We selected a Markov design rather than a propensity score matched analysis because the former enables a sophisticated estimation of QALY accrued. Markov models are inherently capable of generating DFS and OS data (Figure 2A-D); however, improved QALYs is the standard primary outcome for both clinically oriented and cost-effectiveness Markov analyses.10,11 Markov analyses are uniquely positioned to evaluate the length of time a patient remains in a given health state and apply the relevant QALY for that duration. This provides an additional layer of clinical relevance by incorporating patient experience and comorbidity that may not be captured by the limited variables incorporated in the model. For example, QALY better captures the experience of a prolonged hospital admission for IC, compared with outpatient treatment for aza-ven. Additionally, propensity score matched analyses reporting DFS and OS alone would fail to capture sequential relapses within the same patient, and the differences in quality of life associated with transitions between these health states. One limitation of a Markov analysis compared with a propensity score matched analysis is that the latter would create matched cohorts of patients treated with either IC or aza-ven by virtue of similar propensity scores.33 This allows for a balanced distribution of measured covariates in an observational study.

Our model produces results that are reflective of real-world data. For example, comparing the results of the adverse-risk cohort treated with IC and allo-SCT from Bataller et al with our results revealed a 1-year DFS of 48.0% vs 49.3%, 1-year mortality of 43.8% vs 42.3%, 5-year DFS of 33.0% vs 33.4%, and 5-year mortality 62% vs 59%, respectively.22 This association is strong despite using weighted averages from multiple sources. There was generally good agreement between different sources for the probability of the same variable, as can be seen in Table 2.

Although there are recent but limited QALY utility data available for aza-ven, we elected to incorporate the single agent aza QALY utility values. The aza QALY utility data have been published in the literature for longer, so there is more clinical experience with these values, and they have been used in cost-effectiveness analyses.30 This extrapolation is based upon results from VIALE-A, which reported no differences between the aza and aza-ven in terms of quality of life.6,30 The aza-ven QALY values reported in 2 studies were challenging to reconcile with our clinical experience, particularly because the high-utility values (0.733 and 0.723) for the relapse state were so similar to the utility values of 0.815 and 0.796 for DFS in the same studies.34,35 

Our models carry several limitations. As an analysis of largely retrospective data, it was not possible to create IC and aza-ven cohorts with an equal distribution of key demographic and disease-specific characteristics. Given that IC is the standard of care in medically fit individuals, there are more robust data available for younger patients. In the current treatment landscape, aza-ven is primarily offered to patients with comorbidities that preclude them from receiving IC, so data are predominantly only available for older patients. The average age of patients receiving IC in our analysis was less than that of patients receiving aza-ven (adverse-risk IC, 58 years vs aza-ven, 67 years; intermediate-risk IC, 55 years vs aza-ven, 72 years), so these patients were likely more fit. Thus, the QALYs gained by patients in both the adverse- and intermediate-risk IC-treated cohort are likely inflated relative to those of their counterparts treated with aza-ven. Furthermore, among patients who underwent SCT, there was a strong selection bias in favor of those who were medically fit, so the improved survival and QALY outcomes they experienced were not due to this intervention alone.

Another limitation of our analysis is that few studies reported Eastern Cooperative Oncology Group performance status; the Charlson Comorbidity Index; or AML-specific comorbidity indices, such as the AML comorbidity index, the hematopoietic cell transplantation comorbidity index, or the AML composite model (AML-CM).36-38 This introduces bias into our results because older but fit patients might have received aza-ven on the basis of age alone, and younger but unfit patients might have received IC because of discomfort using aza-ven in a currently off-label manner. An example of this would be the effect of comorbidity data on our intermediate-risk model. The 1-way sensitivity analysis for death within the first year in patients treated with IC followed by SCT identified a threshold value of 39%. Thus, if 1-year mortality in these patients exceeded 40%, the intermediate model would actually favor treatment with aza-ven over IC. Given that patients with high comorbidity had much greater mortality within the first year (15% among those with an AML-CM score of 1-4 vs 80% among those with a score > 10), our intermediate-risk model is indeed sensitive to differences in comorbidity as captured by the AML-CM. The impact of formally reported AML-specific comorbidity measures on our QALY outcomes would be an important area for further research, as much of these data predate the aza-ven era.

Finally, most of the patients included in our analysis were from the United States and Europe, which may not capture the global experience. Although there was generally a strong agreement between different sources for the probability of a particular variable in our data, this is likely not the case in all jurisdictions.

Our results contribute to a growing body of literature indicating that the 2017 European Leukemia Network risk stratification is not necessarily prognostic in patients treated with a combination of hypomethylating agent and ven.16,39 An area of particular interest is the efficacy of aza-ven in isocitrate dehydrogenase 1/2 (IDH1/2)-mutated AML (∼8% and ∼12% of AML cases, respectively). The prognostic implications of IDH1/2 mutations have yet to be fully elucidated, with either favorable40,41 adverse,42-44 or uncertain prognosis.45,46 Current literature would suggest that these different prognoses arise on the basis of submutations or comutations.42 Pollyea et al found that patients with IDH1/2 mutations treated with aza and ven had a composite CR rate of 79% as well as a median duration of remission of 29.5 months and a median OS of 24.5 months.47 Notably, patients with IDH1/2-mutated disease had superior OS compared with the wild-type cohort. VIALE-A also reported favorable hazard ratios for patients treated with aza-ven with respect to death of 0.28, 0.34, and 0.34 for IDH1, IDH2, and IDH1/2, respectively, compared with patients treated with aza alone.6 Recent work demonstrated that the combination of the IDH1 inhibitor ivosidenib and aza resulted in a statistically significant increase in DFS and OS compared with aza alone.48 Although the literature on aza-ven in patients with IDH1/2 mutations are limited, this represents an important future application of our model.

Another AML subgroup of interest is patients with FMS-like tyrosine kinase 3 (FLT3) mutations, particularly those with FLT3–internal tandem duplication mutations, given their poor prognosis and high risk of relapse.49 Konopleva et al, however, did not find a significant difference in outcomes between patients with FLT3-mutated disease and those with wild-type disease treated with aza-ven.50 Similarly, although VIALE-A did report a hazard ratio of 0.66 of death for FLT3 in favor of aza-ven, the 95% confidence interval was not significant at 0.35 to 1.26. That being said, recent data have reported that patients with FLT3-mutated disease had superior OS when treated with hypomethylating agent, ven, and FLT3 inhibitor (triplet therapy) compared with hypomethylating agent and FLT3 inhibitor alone (doublet therapy).51 Taken together, further research into less-intensive cocktails personalized to mutational status will provide important opportunities to refine our model.

Our results, although derived from a modeled analysis of largely retrospective data, raise important questions about the use of aza-ven in medically fit patients. Patients with adverse-risk AML may preferentially gain a QALY benefit from treatment with aza-ven. The comparison in the intermediate-risk cohort is more limited given that IC remains the standard of care in this population, and, thus, the aza-ven data are derived from older, less medically fit patients. Future work should be aimed at prospectively evaluating the efficacy of aza-ven in medically fit patients with newly diagnosed AML.

Contribution: R.B. conceptualized the study; M.R., L.M., M.C., R.B., and J.T. were responsible for the study methodology; M.R. and J.T. were responsible for formal analysis and investigation, and writing the original manuscript draft; M.R., L.M., M.C., R.B., and J.T. were responsible for reviewing and editing the manuscript; and R.B. supervised the study.

Conflict-of-interest disclosure: L.M. served on an advisory board for AbbVie. The remaining authors declare no competing financial interests.

Correspondence: Rena Buckstein, Division of Medical Oncology and Hematology, Odette Cancer Center, Sunnybrook Health Sciences Center, 2075 Bayview Ave, T2-042, Toronto, ON M4N 3M5, Canada; email: rena.buckstein@sunnybrook.ca.

1.
DiNardo
CD
,
Wei
AH
.
How I treat acute myeloid leukemia in the era of new drugs
.
Blood
.
2020
;
135
(
2
):
85
-
96
.
2.
Ossenkoppele
G
,
Löwenberg
B
.
How I treat the older patient with acute myeloid leukemia
.
Blood
.
2015
;
125
(
5
):
767
-
774
.
3.
Abel
GA
,
Klepin
HD
.
Frailty and the management of hematologic malignancies
.
Blood
.
2018
;
131
(
5
):
515
-
524
.
4.
Surveillance, Epidemiology, and End Results (SEER) Program
.
Cancer Stat Facts: Leukemia — Acute Myeloid Leukemia (AML). National Cancer Institute
. Accessed 20 December 2023. https://seer.cancer.gov/statfacts/html/amyl.html.
5.
DiNardo
CD
,
Pratz
K
,
Pullarkat
V
, et al
.
Venetoclax combined with decitabine or azacitidine in treatment-naive, elderly patients with acute myeloid leukemia
.
Blood
.
2019
;
133
(
1
):
7
-
17
.
6.
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
.
7.
Cherry
EM
,
Abbott
D
,
Amaya
M
, et al
.
Venetoclax and azacitidine compared with induction chemotherapy for newly diagnosed patients with acute myeloid leukemia
.
Blood Adv
.
2021
;
5
(
24
):
5565
-
5573
.
8.
Fathi
AT
,
Fell
GG
,
El-Jawahri
A
, et al
.
A phase 2 randomized study comparing venetoclax and azacitidine to conventional induction chemotherapy for newly diagnosed fit adults with acute myeloid leukemia
.
Blood
.
2022
;
140
(
suppl 1
):
3284
-
3286
.
9.
Sonnenberg
FA
,
Beck
JR
.
Markov models in medical decision making: a practical guide
.
Med Decis Making
.
1993
;
13
(
4
):
322
-
338
.
10.
Cutler
CS
,
Lee
SJ
,
Greenberg
P
, et al
.
A decision analysis of allogeneic bone marrow transplantation for the myelodysplastic syndromes: delayed transplantation for low-risk myelodysplasia is associated with improved outcome
.
Blood
.
2004
;
104
(
2
):
579
-
585
.
11.
Kurosawa
S
,
Yamaguchi
T
,
Miyawaki
S
, et al
.
A Markov decision analysis of allogeneic hematopoietic cell transplantation versus chemotherapy in patients with acute myeloid leukemia in first remission
.
Blood
.
2011
;
117
(
7
):
2113
-
2120
.
12.
Yamamoto
C
,
Nakashima
H
,
Ikeda
T
, et al
.
Analysis of the cost-effectiveness of treatment strategies for CML with incorporation of treatment discontinuation
.
Blood Adv
.
2019
;
3
(
21
):
3266
-
3277
.
13.
Slot
M
,
Niemann
CU
,
Ehlers
LH
,
Rotbain
EC
.
Cost-effectiveness of targeted treatment vs chemoimmunotherapy in treatment-naive unfit CLL without TP53 aberrations
.
Blood Adv
.
2023
;
7
(
15
):
4186
-
4196
.
14.
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
.
15.
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
.
16.
El Chaer
F
,
Hourigan
CS
,
Zeidan
AM
.
How I treat AML incorporating the updated classifications and guidelines
.
Blood
.
2023
;
141
(
23
):
2813
-
2823
.
17.
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
.
18.
Winters
AC
,
Bosma
G
,
Abbott
D
, et al
.
Outcomes are similar after allogeneic hematopoietic stem cell transplant for newly diagnosed acute myeloid leukemia patients who received venetoclax + azacitidine versus intensive chemotherapy
.
Transplant Cell Ther
.
2022
;
28
(
10
):
694.e1
-
694.e9
.
19.
Salhotra
A
,
Tsai
N-C
,
Zhang
J
, et al
.
Venetoclax and hypomethylating agents yield high response rates and favourable transplant outcomes in patients with newly diagnosed acute myeloid leukaemia
.
Br J Haematol
.
2022
;
196
(
6
):
e71
-
e74
.
20.
Garciaz
S
,
Hospital
M-A
,
Alary
A-S
, et al
.
Azacitidine plus venetoclax for the treatment of relapsed and newly diagnosed acute myeloid leukemia patients
.
Cancers
.
2022
;
14
(
8
):
2025
.
21.
Johnson
IM
,
Ilyas
R
,
McCullough
K
, et al
.
Molecular predictors of response and survival in patients with relapsed/refractory acute myeloid leukemia following venetoclax plus hypomethylating agent therapy
.
Blood
.
2022
;
140
(
suppl 1
):
3233
-
3234
.
22.
Bataller
A
,
Garrido
A
,
Guijarro
F
, et al
.
European LeukemiaNet 2017 risk stratification for acute myeloid leukemia: validation in a risk-adapted protocol
.
Blood Adv
.
2022
;
6
(
4
):
1193
-
1206
.
23.
Herold
T
,
Rothenberg-Thurley
M
,
Grunwald
VV
, et al
.
Validation and refinement of the revised 2017 European LeukemiaNet genetic risk stratification of acute myeloid leukemia
.
Leukemia
.
2020
;
34
(
12
):
3161
-
3172
.
24.
Hansen
DK
,
Kim
J
,
Thompson
Z
, et al
.
ELN 2017 Genetic Risk Stratification predicts survival of acute myeloid leukemia patients receiving allogeneic hematopoietic stem cell transplantation
.
Transplant Cell Ther
.
2021
;
27
(
3
):
256.e1
-
256.e7
.
25.
Burnett
AK
,
Goldstone
A
,
Hills
RK
, et al
.
Curability of patients with acute myeloid leukemia who did not undergo transplantation in first remission
.
J Clin Oncol
.
2013
;
31
(
10
):
1293
-
1301
.
26.
Lo
MY
,
Tsai
C-H
,
Kuo
Y-Y
, et al
.
Prognostic relevance of adult acute myeloid leukemia patients according to the 2022 European Leukemianet Risk Stratification
.
Blood
.
2022
;
140
(
suppl 1
):
130
-
131
.
27.
Rausch
C
,
Rothenberg-Thurley
M
,
Dufour
AM
, et al
.
Validation of the 2022 European Leukemianet Genetic Risk Stratification of Acute Myeloid Leukemia
.
Blood
.
2022
;
140
(
suppl 1
):
3408
-
3409
.
28.
Pasvolsky
O
,
Shimony
S
,
Ram
R
, et al
.
Allogeneic hematopoietic cell transplantation for acute myeloid leukemia in first complete remission after 5-azacitidine and venetoclax: a multicenter retrospective study
.
Ann Hematol
.
2022
;
101
(
2
):
379
-
387
.
29.
Pollyea
DA
,
Pratz
K
,
Letai
A
, et al
.
Venetoclax with azacitidine or decitabine in patients with newly diagnosed acute myeloid leukemia: Long term follow-up from a phase 1b study
.
Am J Hematol
.
2021
;
96
(
2
):
208
-
217
.
30.
Patel
KK
,
Zeidan
AM
,
Shallis
RM
, et al
.
Cost-effectiveness of azacitidine and venetoclax in unfit patients with previously untreated acute myeloid leukemia
.
Blood Adv
.
2021
;
5
(
4
):
994
-
1002
.
31.
Tremblay
G
,
Dolph
M
,
Patel
S
,
Brandt
P
,
Forsythe
A
.
Cost-effectiveness analysis for midostaurin versus standard of care in acute myeloid leukemia in the United Kingdom
.
Cost Eff Resour Alloc
.
2018
;
16
:
33
.
32.
Zeidan
AM
,
Pollyea
DA
,
Borate
U
, et al
.
Venetoclax plus azacitidine compared with intensive chemotherapy as induction for patients with acute myeloid leukemia: retrospective analysis of an electronic medical record database in the United States
.
Ann Hematol
.
2023
;
102
(
4
):
749
-
754
.
33.
Austin
PC
.
An introduction to propensity score methods for reducing the effects of confounding in observational studies
.
Multivariate Behav Res
.
2011
;
46
(
3
):
399
-
424
.
34.
Pratz
KW
,
Chai
X
,
Xie
J
, et al
.
Cost-effectiveness analysis of venetoclax in combination with azacitidine versus azacitidine monotherapy in patients with acute myeloid leukemia who are ineligible for intensive chemotherapy: from a US third party payer perspective
.
Pharmacoeconomics
.
2022
;
40
(
8
):
777
-
790
.
35.
Guinan
K
,
Mathurin
K
,
Au
Y
, et al
.
Venetoclax in combination with azacitidine for the treatment of newly diagnosed acute myeloid leukemia: a canadian cost-utility analysis
.
Curr Oncol
.
2022
;
29
(
10
):
7524
-
7536
.
36.
Sorror
ML
,
Storer
BE
,
Fathi
AT
, et al
.
Development and validation of a novel acute myeloid leukemia-composite model to estimate risks of mortality
.
JAMA Oncol
.
2017
;
3
(
12
):
1675
-
1682
.
37.
Sorror
ML
,
Storer
BE
,
Nyland
J
,
Estey
EH
.
Revised acute myeloid leukemia composite model using the 2017 European LeukemiaNet risk classification
.
JAMA Oncol
.
2019
;
5
(
7
):
1062
-
1064
.
38.
Sorror
ML
,
Storer
BE
,
Fathi
AT
, et al
.
Multisite 11-year experience of less-intensive vs intensive therapies in acute myeloid leukemia
.
Blood
.
2021
;
138
(
5
):
387
-
400
.
39.
Döhner
H
,
Pratz
KW
,
DiNardo
CD
, et al
.
ELN risk stratification is not predictive of outcomes for treatment-naïve patients with acute myeloid leukemia treated with venetoclax and azacitidine
.
Blood
.
2022
;
140
(
suppl 1
):
1441
-
1444
.
40.
Patel
JP
,
Gönen
M
,
Figueroa
ME
, et al
.
Prognostic relevance of integrated genetic profiling in acute myeloid leukemia
.
N Engl J Med
.
2012
;
366
(
12
):
1079
-
1089
.
41.
Chou
W-C
,
Lei
W-C
,
Ko
B-S
, et al
.
The prognostic impact and stability of Isocitrate dehydrogenase 2 mutation in adult patients with acute myeloid leukemia
.
Leukemia
.
2011
;
25
(
2
):
246
-
253
.
42.
Kunadt
D
,
Stasik
S
,
Metzeler
KH
, et al
.
Impact of IDH1 and IDH2 mutational subgroups in AML patients after allogeneic stem cell transplantation
.
J Hematol Oncol
.
2022
;
15
(
1
):
126
.
43.
Paschka
P
,
Schlenk
RF
,
Gaidzik
VI
, et al
.
IDH1 and IDH2 mutations are frequent genetic alterations in acute myeloid leukemia and confer adverse prognosis in cytogenetically normal acute myeloid leukemia with NPM1 mutation without FLT3 internal tandem duplication
.
J Clin Oncol
.
2010
;
28
(
22
):
3636
-
3643
.
44.
Yamaguchi
S
,
Iwanaga
E
,
Tokunaga
K
, et al
.
IDH1 and IDH2 mutations confer an adverse effect in patients with acute myeloid leukemia lacking the NPM1 mutation
.
Eur J Haematol
.
2014
;
92
(
6
):
471
-
477
.
45.
Abbas
S
,
Lugthart
S
,
Kavelaars
FG
, et al
.
Acquired mutations in the genes encoding IDH1 and IDH2 both are recurrent aberrations in acute myeloid leukemia: prevalence and prognostic value
.
Blood
.
2010
;
116
(
12
):
2122
-
2126
.
46.
Chotirat
S
,
Thongnoppakhun
W
,
Promsuwicha
O
,
Boonthimat
C
,
Auewarakul
CU
.
Molecular alterations of isocitrate dehydrogenase 1 and 2 (IDH1 and IDH2) metabolic genes and additional genetic mutations in newly diagnosed acute myeloid leukemia patients
.
J Hematol Oncol
.
2012
;
5
:
5
.
47.
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
.
48.
Montesinos
P
,
Recher
C
,
Vives
S
, et al
.
Ivosidenib and azacitidine in -mutated acute myeloid leukemia
.
N Engl J Med
.
2022
;
386
(
16
):
1519
-
1531
.
49.
Stone
RM
,
Mandrekar
SJ
,
Sanford
BL
, et al
.
Midostaurin plus chemotherapy for acute myeloid leukemia with a FLT3 mutation
.
N Engl J Med
.
2017
;
377
(
5
):
454
-
464
.
50.
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
.
51.
Yilmaz
M
,
Kantarjian
H
,
Short
NJ
, et al
.
Hypomethylating agent and venetoclax with FLT3 inhibitor “triplet” therapy in older/unfit patients with FLT3 mutated AML
.
Blood Cancer J
.
2022
;
12
(
5
):
1
-
9
.

Author notes

R.B. and J.T. are joint senior authors.

Data are available on request from the corresponding author, Rena Buckstein (rena.buckstein@sunnybrook.ca).

The full-text version of this article contains a data supplement.

Supplemental data