CADTH Reimbursement Review

Venetoclax (Venclexta)

Sponsor: AbbVie Corporation

Therapeutic area: Acute myeloid leukemia

This multi-part report includes:

Clinical Review

Pharmacoeconomic Review

Clinical Review

Abbreviations

AML

acute myeloid leukemia

ANC

absolute neutrophil count

ATB-MPG

Alberta Tumour Board Myeloid Physicians Group

AZA

azacitidine

BSC

best supportive care

CBC

complete blood count

CI

confidence interval

CLL

chronic lymphocytic leukemia

CLSG

Canadian Leukemia Study Group

CMH

Cochran-Mantel-Haenszel

CNS

central nervous system

CR

complete remission

CRh

complete remission with incomplete hematological recovery

CRi

complete remission with incomplete bone marrow recovery

CrI

credible interval

DB

double-blind

ECOG PS

Eastern Cooperative Oncology Group Performance Status

EFS

event-free survival

EORTC QLQ-30C

European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire Core 30

EQ VAS

EuroQol Visual Analogue Scale

EQ-5D-5L

EuroQol 5-Dimensions 5-Levels questionnaire

FLT3

FMS-like tyrosine kinase 3

GHS/QoL

global health status quality of life scale (EORTC QLQ-30C)

HMA

hypomethylating agent

HRQoL

health-related quality of life

IA1

first interim analysis

IA2

second interim analysis

IDMC

independent data monitoring committee

ITC

indirect treatment comparison

LDAC

low-dose cytarabine

LLSC

Lymphoma Society of Canada

MDS

myelodysplastic syndrome

MID

minimal important difference

MLFS

morphological leukemia-free state

MRD

minimal/measurable disease

NMA

network meta-analysis

OR

odds ratio

OS

overall survival

PROMIS 7a

Patient-Reported Outcomes Measurement System Short Form v1.0–Fatigue 7a

QoL

quality of life

RCT

randomized controlled trial

SAE

serious adverse event

SD

standard deviation

WDAE

withdrawal due to adverse event

Executive Summary

An overview of the submission details for the drug under review is provided in Table 1.

Table 1: Submitted for Review

Item

Description

Drug product

Venetoclax (Venclexta), 10 mg, 50 mg, 100 mg, tablets, oral

Indication

Venclexta, in combination with azacitidine or low-dose cytarabine is indicated for the treatment of patients with newly diagnosed AML who are 75 years or older or who have comorbidities that preclude use of intensive induction chemotherapy

Reimbursement request

As per indication

Health Canada approval status

NOC

Health Canada review pathway

Standard

NOC date

December 4, 2020

Sponsor

AbbVie Corporation

AML = acute myeloid leukemia; NOC = Notice of Compliance.

Introduction

Acute myeloid leukemia (AML) is a hematological malignancy defined by WHO as a myeloid neoplasm with greater than 20% blasts in the peripheral blood or bone marrow. Proliferating myeloid precursor cells leads to disruption of normal hematopoiesis and a clinical presentation of symptoms and complications of pancytopenia or leukostasis. Diagnosis is by complete blood count (CBC) and bone marrow biopsy, with identification of characteristic mutations and chromosomal rearrangements for targeted treatment and cytogenetic risk stratification. AML predominately occurs in older adults, with a median age of diagnosis of 67 years in Canada, and increasing incidence with age. Standard treatment for patients who are medically fit is intensive induction therapy with cytarabine and an anthracycline, but a substantial portion of patients with AML are ineligible for induction therapy due to frailty associated with age or comorbidities. Patients ineligible for treatment with induction therapy may be treated with hypomethylating agents (HMAs), such as azacitidine or low-dose cytarabine (LDAC), but rates of complete remission (CR) are low and duration of remission tends to be short. Prognosis for AML in older patients is poor, with 1 study reporting 5-year overall survival (OS) as 6.3% in patients aged 65 years.

Venetoclax is an orally administered highly selective inhibitor of the anti-apoptotic protein B-cell lymphoma 2 (BCL2). Health Canada granted a Notice of Compliance on December 4, 2020 for the following indication: Venclexta, in combination with azacitidine or LDAC, is indicated for the treatment of patients with newly diagnosed AML who are 75 years or older or who have comorbidities that preclude use of intensive induction chemotherapy. The recommended dose of venetoclax in combination with azacitidine is 400 mg/day for each day of a 28-day cycle following a 3-day ramp-up; azacitidine should be administered at 75 mg/m2 for days 1 to 7 of the cycle. Dose adjustments are required in patients treated with strong and moderate inhibitors of CYP3A enzymes. Venetoclax has previously been reviewed by CADTH for its use in chronic lymphocytic leukemia (CLL) as monotherapy for patients with 17p deletion or without a 17p deletion who did not have other available treatment options in combination with obinutuzumab in previously untreated patients, and in combination with rituximab for patients who had received at least 1 prior therapy. A concurrent CADTH review of venetoclax with LDAC is ongoing.

The objective of the systematic review was to review the beneficial and harmful effects of venetoclax in combination with azacitidine for the treatment of patients with newly diagnosed AML who are 75 years or older, or who have comorbidities that preclude use of intensive induction therapy.

Stakeholder Perspectives

The information in this section is a summary of input provided by Canadian patient and clinician groups who responded to CADTH’s call for patient input and by clinical expert(s) consulted by CADTH for the purpose of this review.

Patient Input

One patient advocacy group, the Leukemia and Lymphoma Society of Canada (LLSC), provided input on venetoclax in combination with azacitidine for the treatment of AML. The LLSC used an online survey for its submission, which was conducted between December 7, 2020 and January 24, 2021. Twenty-nine patients responded, all from Canada, 5 of whom had experience with venetoclax in combination with azacitidine.

Many patients did not provide information on specific symptoms but described being diagnosed with AML as a life-changing event that affected not only themselves but their caregivers. Some patients needed to relocate to access treatment. Side effects of treatment, transfusion dependence and hospital admissions had a large impact on patients’ quality of life (QoL), as did isolation due to their vulnerability to infection. Patients reported the desired characteristics of treatment options as those that could maintain remission, were targeted with fewer side effects, covered by public plans, and accessible in their geographic region.

Clinician Input

Input From Clinical Experts Consulted by CADTH

The experts indicted that currently available lower-intensity treatments have low rates of CR, and the CRs that are produced are not durable. They indicated that venetoclax plus azacitidine (or other HMAs) would change the current treatment paradigm, becoming the new standard of care for patients with treatment-naive AML who were ineligible for standard induction therapy, and providing an option for patients aged 75 years or older who were eligible for intensive chemotherapy, following discussion about risks and benefits.

The experts indicated that, at this time, there is insufficient information to make treatment decisions based on disease characteristics, and that while certain subgroups had been excluded from clinical trials, such as patients with central nervous system (CNS) involvement, these groups might reasonably be expected to benefit. The experts indicated that current evidence does not fully support the use of venetoclax plus azacitidine in fit patients eligible for standard induction treatment or in patients aged 75 years and older with good cytogenetic risk (core binding factor) AML who are fit for intensive induction chemotherapy, and their opinions differed in its suitability for patients with relapsed or refractory disease.

The experts indicated that response to treatment would be determined by achievement of CR with or without complete hematological recovery, as measured by CBC and bone marrow biopsy and/or transfusion independence or stable disease. OS and hospital visits, transfusion needs, and QoL were the most important end points. Assessment of response could be carried out after the first or second cycle.

The experts indicated that discontinuation of treatment might be determined by disease progression or intolerable adverse events, but could not comment whether venetoclax could be continued after azacitidine discontinuation. One respondent indicated that a bone marrow biopsy should be performed after the first and second treatment cycles, as response would be expected after a maximum of 2 cycles. Another indicated that response should be assessed at minimum after 4 to 6 cycles, but that most practitioners assess after the first cycle, given cost and to guide dosing of venetoclax for subsequent cycles.

The experts indicated that treatment should be given in a hospital or outpatient setting by a physician with experience looking after acute leukemia patients. Pharmacist involvement would be needed for management of drug interactions (e.g., azoles). Hospitalization might be required for ramping up the dose of venetoclax, with prophylaxis for tumour lysis syndrome, and the need for admission to manage neutropenic fever and other complications during therapy should be anticipated.

Clinician Group Input

Four clinician groups provided input: the Canadian Leukemia Study Group (CLSG), the Ontario Health (Cancer Care Ontario) Hematology Disease Site Drug Advisory Committee (OH-CCO Hem-DAC), the Leukemia/Bone Marrow Transplant (L/BMT) Program of British Columbia, and the Alberta Tumour Board Myeloid Physicians Group (ATB-MPG).

There were no substantive differences in opinions between the clinical experts consulted by CADTH and the clinical groups. The groups noted that patients are aware of venetoclax and azacitidine, and some patients have been “self-funding” venetoclax by using CYP3A inhibitors to reduce the dose and, thus, the cost of venetoclax.

Drug Program Input

The drug programs indicated that current treatment options for patients with newly diagnosed AML who are ineligible for intensive chemotherapy include azacitidine, LDAC, and best supportive care (BSC). The reimbursement of venetoclax plus azacitidine would likely replace azacitidine in this treatment setting. Azacitidine is funded in most jurisdictions for patients with AML who are ineligible for intensive chemotherapy, and some jurisdictions fund alternate dosing schedules for azacitidine (i.e., 5 to 2-2 and 6 consecutive days) in addition to the schedule of 7 consecutive days. However, it was noted that some patients 75 years of age and older may be fit to tolerate intensive chemotherapy. The ramp-up dosing schedule for venetoclax with azacitidine differs significantly from the ramp-up dosing schedule already in use for chronic lymphocytic leukemia (CLL) indications and the current packaging for venetoclax is designed for the CLL ramp-up dosing schedule. Venetoclax plus azacitidine includes an oral and an IV and subcutaneous drug and therefore would be reimbursed through different programs in some jurisdictions. The drug programs identified the potential for indication creep for patients with a high risk of myelodysplastic syndrome (MDS), those who have progressed or have had an inadequate response on low-dose chemotherapy for AML, and patients who have relapsed after induction chemotherapy and are not eligible for stem cell transplant and who are then treated with azacitidine. It was noted that treatment combination increases the need for health care resources (i.e., hospital admission and additional pharmacy and nursing resources for the potential management of tumour lysis syndrome and monitoring for drug interactions). Affordability was also identified as an issue since the combination is expected to replace azacitidine monotherapy.

Clinical experts were consulted by CADTH for questions related to implementing venetoclax plus azacitidine into current provincial drug plans. Overall, most implementation questions related to the dosing schedule and administration and the eligible patient population.

Clinical Evidence

Pivotal Studies and Protocol Selected Studies

Description of Studies

One double-blind, placebo-controlled phase III randomized controlled trial (VIALE-A) contributed evidence to this review. The trial objective was to evaluate the efficacy and safety of venetoclax plus azacitidine compared with placebo plus azacitidine in adults with newly diagnosed AML who were 18 years or older and ineligible for standard induction therapy due to age or comorbidities. The trial was restricted to patients who had not previously been treated with an HMA and who had intermediate or poor risk cytogenetics. The primary outcomes were OS and composite complete remission rate (i.e., CR plus complete remission with incomplete marrow recovery [CR + CRi]). Secondary outcomes were CR, CR plus complete remission with incomplete hematological recovery [CR + CRh], rate of CR + CRi by the initiation of cycle 2, transfusion-independence rate, minimal/measurable disease (MRD) response rate, response rates and OS in molecular subgroups, fatigue, global health status and quality of life (GHS/QoL), and event-free survival (EFS).

A total of 431 patients were randomized in a 2:1 ratio: 286 to venetoclax plus azacitidine and 144 to placebo plus azacitidine. The most common reasons given for patients to be considered ineligible for standard induction therapy were age and Eastern Cooperative Oncology Group Performance Status (ECOG PS). Patients were elderly, with poor performance and markers of severe disease. The mean age was 75.4 years, with 60.6% aged 75 years or older. Almost all patients were White or Asian, and the majority of patients were male (60.1%). Most (75.2%) had de novo rather than secondary AML. Nearly 2-thirds had intermediate risk cytogenetics, 1-third had poor risk, and 1-half had bone marrow blasts of 50% or greater at baseline.

Efficacy Results

Table 2 shows a summary of the key efficacy and safety outcomes. Venetoclax plus azacitidine improved most outcome measures that were identified as being of interest to clinicians and patients. Statistically significant treatment differences were seen for OS, EFS, measures of disease response (CR + CRi, CR + CRh, CR), and post-baseline transfusion independence. Improvements were also seen for OS and CR + CRi in the subgroup of patients with isocitrate dehydrogenase 1 (IDH1) or IDH2 mutation and for CR + CRi among patients with FMS-like tyrosine kinase 3 (FLT3) mutations. No statistically significant difference was detected in OS for patients with FLT3 mutations; however, the subgroup was small, making it difficult to detect a difference. While clinically meaningful differences in patient-reported outcomes of GHS/QoL and fatigue were observed at individual end points, differences between treatment groups could not be interpreted because the sequential testing strategy failed before this level.

Harms Results

Table 2 shows a summary of the key efficacy and safety outcomes. All patients in both groups experienced at least 1 adverse event, and almost all experienced at least 1 grade 3 or greater adverse event. Compared with patients who received placebo plus azacitidine, a greater proportion of patients who received venetoclax plus azacitidine experienced 1 or more serious adverse events (SAEs), 1 or more adverse events leading to discontinuation or dose interruption of venetoclax or placebo or azacitidine, or 1 or more adverse events leading to death. Common harms in all categories are generally predictable from the known mechanism of action for venetoclax and/or azacitidine and the underlying disease. Cytopenias were common, with neutropenia, febrile neutropenia, thrombocytopenia, and anemia represented across all categories, as were gastrointestinal adverse effects. Febrile neutropenia and infections contributed substantially to most common SAEs and were the most frequent adverse events leading to death.

The notable harms identified for the protocol were neutropenia, febrile neutropenia, infections, tumour lysis syndrome, hemorrhage, and secondary malignancies. Neutropenia, febrile neutropenia, infections and infestations, and secondary primary malignancies all occurred in a greater proportion of patients who received venetoclax plus azacitidine than in patients who received placebo plus azacitidine. Hemorrhage and tumour lysis syndrome occurred in similar proportions, and the proportion of patients with tumour lysis syndrome was low (≤ 2.5%). The most common secondary malignancies were basal cell carcinoma and squamous cell carcinoma of the skin.

Table 2: Summary of Key Results From VIALE-A

Results

VEN + AZA

N = 286

PBO + AZA

N = 145

OS

Events (deaths), n (%)

161 (56.3)

109 (75.2)

Median OS, months (95% CI)

14.7 (11.9 to 18.7)

9.6 (7.4 to 12.7)

HR (Cox proportional hazards model)a (95% CI)

0.662 (0.518 to 0.845)

P value (stratified log-rank test)a

< 0.001b

Event-free survival

Number of patients with events, n (%)

191 (66.8)

122 (84.1)

Median duration of event-free survival (months; 95% CI)

9.8 (8.4 to 11.8)

7.0 (5.6 to 9.5)

HR (Cox proportional hazards model)a (95% CI)

0.632 (0.502 to 0.796)

P value (stratified log-rank test)a

< 0.001b

Best response (CR + CRi) by investigator assessment

CR + CRi rate at IA1, n (%; 95% CI)c

Number of patients at IA1

147

79

CR + CRi

96 (65.3; 57.0 to 73.0)

20 (25.3; 16.2 to 36.4)

P value (stratified CMH test)a

< 0.001

CR + CRi rate (as best response), n (%; 95% CI)c

CR

105 (36.7; 31.1 to 42.60)

26 (17.9; 12.1 to 25.2)

P value (stratified CMH test)a

< 0.001b

CR + CRi

190 (66.4; 60.6 to 71.9)

41 (28.3; 21.1 to 36.3)

CR + CRi rate (as best response) by initiation of cycle 2, n (%; 95% CI)a

CR + CRi

124 (43.4; 37.5 to 49.3)

11 (7.6; 3.8 to 13.2)

P value (stratified CMH test)a

< 0.001b

Time to response (CR + CRi) by investigator assessment

Time to first response, months, mean (SD) median (range)

CR + CRi

2.1 (1.82)
1.3 (0.6 to 9.9)

3.3 (2.61)
2.8 (0.8 to 13.2)

Time to best response, months, mean (SD) median (range)

CR + CRi

3.6 (3.66)
2.3 (0.6 to 24.5)

4.2 (2.89)
3.7 (0.8 to 13.2)

Duration of response (CR + CRi and CR) based on investigator assessment

CR + CRi

Number of patients with events, n/N (%)

84/190 (44.2)

23/41 (56.1)

DOR (months)a

Median (95% CI)

17.5 (13.6, NE)

13.4 (5.8 to 15.5)

CR

Number of patients with events, n/N (%)

39/105 (37.1)

13/26 (50.0)

DOR (months)a

Median (95% CI)

17.5 (15.3 to NE)

13.3 (8.5 to 17.6)

Post-baseline transfusion-independence rate

RBC and platelet, n (%; 95% CI)

166 (58.0; 52.1 to 63.8)

49 (33.8; 26.2 to 42.1)

Treatment difference, % (95% CI)

24.2 (14.7 to 33.8)

RBC

171 (59.8; 53.9 to 65.5)

51 (35.2; 27.4 to 43.5)

Treatment difference, % (95%)

24.6 (15.0 to 34.2)

P value (stratified CMH test)a

< 0.001b

Platelet

196 (68.5; 62.8 to 73.9)

72 (49.7; 41.3 to 58.1)

Treatment difference, % (95%)

18.9 (9.1 to 28.6)

P value (stratified CMH test)a

< 0.001b

Harms

Patients with any AE, n (%)

283 (100)

144 (100)

Patients with AE grade ≥ 3, n (%)

279 (98.6)

139 (96.5)

Patients with any SAE, n (%)

235 (83.0)

105 (72.9)

Patients with VEN- or PBO-related AE,a n (%)

241 (85.2)

96 (66.7)

Patients with AZA-related AE,a n (%)

246 (86.9)

108 (75.0)

Patients with any AE leading to discontinuation of VEN or PBO, n (%)

69 (24.4)

29 (20.1)

Patients with any AE leading to AZA discontinuation, n (%)

68 (24.0)

29 (20.1)

Patients with any AE leading to VEN or PBO dose interruption or reduction, n (%)

204 (72.1)

84 (58.3)

Patients with any AE leading to AZA dose interruption or reduction, n (%)

190 (67.1)

67 (46.5)

Patients with any AE leading to death, n (%)

64 (22.6)

29 (20.1)

Subgroups

OS

IDH1 and/or IDH2 mutation

N

61

28

Median OS, months (95% CI)

NE (12.1 to NE)

6.2 (2.3 to 12.7)

HR (unstratified Cox model) (95% CI)

0.345 (0.199 to 0.598)

P value (unstratified log-rank test)

< 0.0001b

FLT3 mutation

N

29

22

Median OS, months (95% CI)

12.7 (7.3 to 23.5)

8.6 (5.9 to 14.7)

HR (unstratified Cox model) (95% CI)

0.664 (0.351 to 1.257)

P value (unstratified log-rank test)

0.2054

CR + CRi

IDH1 and/or IDH2 mutation

N

61

28

CR, n (%; 95% CI)

26 (42.6; 30.0 to 55.9)

1 (3.6; 0.1 to 18.3)

CR + CRi, n (%; 95% CI)

46 (75.4; 62.7 to 85.5)

3 (10.7; 2.3 to 28.2)

Risk difference % (95% CI)

64.70 (48.9 to 80.4)

P value (Fisher's exact test)

< 0.001b

FLT3 mutation

N

29

22

CR, n (%; 95% CI)

10 (34.5; 17.9 to 54.3)

3 (13.6; 2.9 to 34.9)

CR + CRi, n (%; 95% CI)

21 (72.4; 52.8 to 87.3)

8 (36.4; 17.2 to 59.3)

Risk difference (%; 95% CI)

36.05 (10.2 to 61.9)

P value (Fisher’s exact test)

0.021b

AE = adverse event; AML = acute myeloid leukemia; AZA = azacitidine; CI = confidence interval; CMH = Cochran-Mantel-Haenszel; CR = complete remission; CRh = complete remission with incomplete hematological recovery; CRi = complete remission with incomplete blood count recovery; DOR = duration of response; FLT3 = FMS-like tyrosine kinase 3; HR = hazard ratio; IA1 = first interim analysis; OS = overall survival; PBO = placebo; RBC = red blood cell; SD = standard deviation; VEN = venetoclax.

Note: Data cut-off was January 4, 2020.

aStratified by age (18 to < 75 years, ≥ 75 years) and cytogenetic risk (intermediate risk, poor risk).

bStatistically significant under the preplanned testing strategy.

cCalculated from the exact binomial distribution.

Source: Clinical Study Report.1

Critical Appraisal

The study was well conducted, with no clinically meaningful imbalance in baseline characteristics, minimal loss to follow-up, and a collection of end points that were standardized and meaningful to patients. Multiplicity was controlled throughout testing of the primary and secondary efficacy end points, with pre-specified strategies for testing of end points. The overall rate of discontinuations from the study was low and assumptions surrounding missing data were conservative for most end points. Interpretation of patient-reported outcome data is limited due to attrition of numbers over cycles.

The generalizability concerns that were identified included the assumption that patients aged 75 years and older would not be eligible for standard induction therapy, and the need for venetoclax and azacitidine to be limited to settings that could provide monitoring and supportive care. In the Canadian setting, patients aged 75 years and older would be considered for treatment if they were medically fit, especially if they had good or intermediate risk cytogenetics. Patients from rural and remote Canadian settings would have to travel for care or would be limited to other treatment options.

Indirect Comparisons

Description of Studies

A systematic review was conducted of trials comparing venetoclax plus azacitidine, venetoclax plus LDAC, azacitidine alone, LDAC alone, and BSC in adults with AML who were not eligible for standard induction chemotherapy. Three indirect treatment comparison (ITC) analyses were conducted: 1 network meta-analysis (NMA) and 2 propensity score–weighting analyses that compared venetoclax plus azacitidine with LDAC and azacitidine with LDAC. For the NMA, HR data were available for OS for 4 trials in a connected network and for proportions of patients with CR + CRi for 3 trials. For the propensity score–weighting analysis, data were available for OS, EFS, and CR + CRi from VIALE-A and the LDAC group from VIALE-C.

Efficacy Results

In the NMA, the results favoured a lower hazard of death for patients assigned to venetoclax plus azacitidine compared with azacitidine (HR = 0.66; 95% credible interval [CrI], 0.52 to 0.85), LDAC (HR = 0.57; 95% CrI, 0.40 to 0.81), and BSC (HR = 0.37; 95% CrI, 0.24 to 0.58), with no treatment favoured between venetoclax plus azacitidine and venetoclax plus LDAC (HR = 0.81; 95% CrI, 0.50 to 1.31). For CR + CRi, venetoclax plus azacitidine was favoured over azacitidine (odds ratio [OR] = 5.05; 95% CrI, 3.30 to 7.87), LDAC (OR = 5.42; 95% CrI, 2.80 to 10.50), and BSC (OR = 61.55; 95% CrI, 8.23 to 1,881.53), with no treatment favoured between venetoclax plus azacitidine and venetoclax plus LDAC (OR = 0.86; 95% CrI, 0.30 to 2.35).

In the first propensity-score analysis, venetoclax plus azacitidine was favoured over LDAC for OS (HR = 0.50; 95% confidence interval [CI], 0.35 to 0.73), EFS (HR = 0.40; 95% CI, 0.28 to 0.58), and CR + CRi (OR = 10.17; 95% CI, 4.55 to 22.73). In the second propensity-score analysis for OS, venetoclax plus azacitidine was favoured over LDAC (HR = 0.52; 95% CrI, 0.36 to 0.77) and azacitidine (HR = 0.64; 95% CrI, 0.50 to 0.82), and no statistically significant difference was seen between azacitidine and LDAC (HR = 0.78; 95% CrI, 0.52 to 1.17). For EFS, venetoclax plus azacitidine was favoured over azacitidine (HR = 0.62; 95% CrI, 0.49 to 0.77) and LDAC (HR = 0.41; 95% CrI, 0.29 to 0.59), and azacitidine was favoured over LDAC (HR = 0.63; 95% CrI, 0.43 to 0.92). For CR + CRi, venetoclax plus azacitidine was favoured over azacitidine (OR = 5.02; 95% CrI, 3.24 to 7.77) and LDAC (OR = 9.69; 95% CrI, 4.30 to 21.85), and no statistically significant difference was seen between azacitidine and LDAC (OR = 1.93; 95% CrI, 0.82 to 4.54).

Harms Results

No analysis of harms was included in the indirect comparisons.

Critical Appraisal

A key limitation of the NMA was the clinical heterogeneity between studies in potential treatment-effect modifiers of blast count at baseline, prior treatment with an HMA, and cytogenetic risk. As the network was sparse, fixed-effects models had to be used, and there was no opportunity for baseline covariate adjustments. Due to these limitations, the comparative efficacy estimates may be biased, and it is not possible to quantify or identify the direction of the bias. Certain estimates, particularly for CR + CRi, were imprecise due to sparse data. In the propensity-score analyses, weighting was generally good, but the relatively small numbers of patients in the LDAC comparator group limited the number of covariates that could be included in the model. The comparisons were not randomized and the results were highly susceptible to bias due to imbalances in unmeasured confounders.

Conclusions

One double-blind, placebo-controlled phase III randomized clinical trial (RCT) (VIALE-A) and 1 ITC provided evidence supporting the efficacy and safety of venetoclax plus azacitidine in adult patients ineligible for standard induction chemotherapy due to age or comorbidities. Compared with azacitidine alone, patients treated with venetoclax (400 mg daily) and azacitidine (75 mg/m2 on days 1 through 7 of a 28-day cycle) showed benefits in important clinical end points of OS, overall and early composite complete remission (CR + CRi), EFS, CR, and transfusion-independence data (red blood cell or platelet). All study participants reported treatment-emergent adverse events. For most categories of adverse events, there was an overall higher proportion of patients reporting in venetoclax plus azacitidine. The most common adverse events were cytopenias and infections. No firm conclusions can be drawn for differences between groups in GHS/QoL and fatigue, and patient attrition reduced the number of observations over the cycles, which limits the interpretation for these end points. Overall, the study was well conducted.

The VIALE-A study did not include a comparison between venetoclax plus azacitidine and current standards of care of induction therapy (in patients aged ≥ 75 and fit), LDAC, or BSC, or the alternative combination of venetoclax plus LDAC. In an ITC, venetoclax plus azacitidine was favoured over monotherapies and basic supportive care, but no treatment was favoured for survival or for composite complete remission between venetoclax plus azacitidine and venetoclax plus LDAC. No data are available for the comparison of venetoclax plus azacitidine with induction therapy. Results for 2 propensity-score comparisons between venetoclax plus azacitidine and azacitidine and LDAC were consistent. Small study and patient numbers and the potential for bias limit the reliability of the ITC, and the propensity-score comparisons were not randomized and therefore highly susceptible to bias.

Introduction

Disease Background

AML is a hematological malignancy defined by WHO as a myeloid neoplasm with greater than 20% blasts in the peripheral blood or bone marrow.2 AML results from malignant transformation of myeloid precursor cells to produce 1 or more clonal populations that can proliferate but do not normally differentiate into their mature forms. This leads to an accumulation of leukemic blasts or immature cells in the bone marrow, peripheral blood, and extramedullary tissues, which disrupt normal hematopoiesis. WHO 2016 guidelines2 identify 6 distinct groups of AML:

AML is the most common form of acute leukemia in adults. According to the Canadian Cancer Society’s most recent data, in 2016, 1,090 people were diagnosed with AML, 610 men and 480 women.3 In 2017, 1,184 people died of AML, 678 men and 506 women.3 Projections for the Canadian population in 2020 only report figures for all forms of leukemia; according to these projections, 6,900 patients would be diagnosed with leukemia, and 3,000 would die. Assuming that around 24% of leukemia in Canada is AML (data from 1992 to 2008),4 the subgroup diagnosed with AML would comprise approximately 1,660 patients.

AML occurs predominately in older adults, with a median age at diagnosis of 67 years in Canada.4 Incidence increases with age. There is an increased incidence in men and non-Hispanic Whites compared with women and other racial or ethnic groups. Certain environmental exposures have been associated with an increased risk of AML (e.g., chemicals, radiation, tobacco, and retroviruses). AML can occur as a secondary malignancy following chemotherapy, or develop out of a pre-existing hematopoietic abnormality such as MDS, other myeloproliferative neoplasms, paroxysmal nocturnal hemoglobinuria, aplastic anemia, or clonal hematopoiesis of indeterminate prognosis. Rarely, it can be associated with an inherited genetic abnormality or familial predisposition to hematologic disorders.

Patients typically present with the symptoms or complications of disrupted hematopoiesis (bleeding or bruising, infection that can be life-threatening, fatigue or shortness of breath, headache or focal neurologic complaints) or symptoms of leukostasis resulting from an excess of immature white cells in the peripheral blood. A presumptive diagnosis of leukemia may be made from a CBC and smear, but confirmation is usually by bone marrow biopsy and aspirate. Characterization of specific genetic abnormalities enables stratification by genetic risk into favourable, intermediate, and poor risk categories, which is used to guide treatment decisions or use of therapies targeted against specific mutations.

The overall 5-year survival for AML in adults in Canada is 21%.5 Older patients have notably poorer survival. In US data, the 5-year survival is 44.8% in patients younger than 65 years and 6.3% in patients aged 65 years and older.6 In another study, patients aged 60 years and older had 1-year and 5-year survival of 20.1% and 8.4%, respectively.7 Survival is influenced by cytogenetic and genetic risk.

Standards of Therapy

Standard treatment for patients who are medically fit consists of cytotoxic remission induction therapy with cytarabine, administered by infusion over 7 days, combined with an anthracycline, usually daunorubicin or idarubicin, given daily for the first 3 days. Induction therapy is followed by high-intensity consolidation therapy. This may be accompanied by targeted therapy for specific clinical situations or genetic mutations: midostaurin in patients with FLT3, and gemtuzumab ozogamicin (monoclonal antibody against CD33) in patients with favourable and intermediate risk disease.8

Determination of eligibility for intensive chemotherapy is based on patient age, fitness, presence of comorbidities, and patient preferences. In general, intensive therapy is poorly tolerated by older patients. According to the 2017 Canadian Consensus Guidelines for treatment of older patients with AML8 induction therapy shows a survival benefit for patients up to age 80, with the exception of those with major comorbidities or those with adverse risk cytogenetics who were not candidates for hematopoietic stem cell transplant.8 Anthracycline and cytarabine are the recommended drugs for induction therapy, with the addition of midostaurin for patients with an FLT3 mutation, and the addition of gemtuzumab ozogamicin for patients with de novo AML and favourable or intermediate risk cytogenetics. For patients who are not eligible for induction therapy, azacitidine is recommended for those with adverse risk cytogenetics or transformed from MDS, while either HMA or LDAC could be used for others. Acute promyelocytic leukemia would be treated with arsenic trioxide plus all-trans retinoic acid (with an anthracycline for those with white blood cell count > 10 × 109/L).

Azacitidine has been approved by Health Canada for patients with low blast count AML (blast counts of 20% to 30%); however, in multiple jurisdictions, it is used and provides clinical benefit in patients with blast counts of 30% or greater. Some jurisdictions fund alternative dosing schedules for azacitidine besides the standard 7-day consecutive regimen. According to input from the clinicians consulted by CADTH for the purpose of this review, in real-world clinical practice in Canada, many eligible patients were unable to receive azacitidine-based therapy, as this drug has to be administered in an oncology clinic setting because of its instability after reconstitution. Many patients who live in rural areas and some in urban settings are unable to travel to these clinics regularly to receive treatment due to the distances involved, their overall frailty, and challenges in obtaining suitable transportation.9

Not all patients respond to first-line therapy and all become refractory to current treatment options, with limited life expectancy. There are few effective treatment options following relapse on front-line AML therapy8; some patients will receive off-label azacitidine with or without venetoclax or participate in a clinical trial. A minority of patients with FLT3 mutations receive gilteritinib, but many patients are not well enough to tolerate further therapy; hence, they receive BSC only.8

Drug

Venetoclax is an orally administered highly selective inhibitor of the anti-apoptotic protein BCL2. Increased expression of BCL2 has been measured in AML blasts, and a majority of AML stem cells express high levels of BCL2 and are dependent on BCL2 for survival. High levels of expression of BCL-2 have been associated with poorer response to chemotherapy and poorer survival in patients with AML. Azacitidine is also thought to affect the inhibition of the pro-survival proteins MCL1 and BCL-XL, so co-administration of azacitidine should increase the dependence of leukemia cells on the BCL2 pathway for survival, and potentiate the effect of venetoclax.

Venetoclax was granted a Health Canada Notice of Compliance on December 4, 2020. The approved indication was: Venclexta (venetoclax) in combination with azacitidine or LDAC, is indicated for the treatment of patients with newly diagnosed AML who are 75 years or older, or who have comorbidities that preclude use of intensive induction therapy. This is consistent with the reimbursement request. Venetoclax has also been approved for the treatment of CLL, as monotherapy for patients with or without a 17p deletion who do not have other available treatment options, in combination with obinutuzumab in previously untreated patients, and in combination with rituximab for patients who have received at least 1 prior therapy. A concurrent CADTH review of venetoclax with LDAC is ongoing, and previous CADTH reviews were conducted for the indications in CLL.

Table 3 summarizes the characteristics and indications for venetoclax, azacitidine, and cytarabine.

Table 3: Key Characteristics of Venetoclax, Azacitidine, and Cytarabine

Characteristic

Venetoclax

Azacitidine

Low-dose cytarabine

Mechanism of action

Selective inhibitor of the anti-apoptotic protein BCL2

Inhibits DNA methyltransferase, blocking methylation of new DNA. Hypomethylation of DNA can reverse hypermethylation leading to gene silencing

Kills cells undergoing DNA synthesis (S-phase). Under certain conditions, blocks progression of cells from G1 phase to S-phase. Acts through inhibition of DNA polymerase

Indicationa

In combination with azacitidine or low-dose cytarabine for the treatment of patients with newly diagnosed AML who are 75 years or older, or who have comorbidities that preclude use of intensive induction therapy

AML with 20% to 30% blasts and multi-lineage dysplasia, according to WHO classification

Route of administration

Oral, tablet

SC

SC

Recommended dose

In combination with azacitidine 400 mg/day following a 3-day ramp-up. In combination with LDAC 600 mg/day following a 4-day ramp-up

75 mg/m2 daily for 7 consecutive days in a 28-day treatment cycle for a recommended minimum of 6 cycles

20 mg SC twice daily, or 20 mg/m2 SC daily for 10 consecutive days in a 28-day treatment cycle for a recommended minimum of 4 cycles

Serious adverse effects or safety issues

Serious warnings and precautions10:

  • tumour lysis syndrome (prophylaxis required)

  • serious infections

Warnings and precautions:

  • secondary primary malignancies

  • hemorrhage

  • neutropenia

  • infections

Serious warnings and precautions11:

  • thrombocytopenia

  • renal failure

Warnings and precautions:

  • tumour lysis syndrome

  • anemia, neutropenia, thrombocytopenia

Serious warnings and precautions12:

  • cardiomyopathy with subsequent death

  • GI toxicity, at times fatal

  • acute pancreatitis

  • CNS toxicity, severe neurologic adverse reactions, paraplegia, necrotizing leukoencephalopathy, and spinal cord toxicity

  • infection

  • pulmonary toxicity, adult respiratory distress syndrome, pulmonary edema

  • myelosuppression

Other

Concomitant use of strong CYP3A inhibitors during initiation and ramp-up requires venetoclax dose reduction

Not beneficial in patients with poor risk cytogenetics

AML = acute myeloid leukemia; BCL2 = B-cell leukemia protein 2; CNS = central nervous system; GI = gastrointestinal; LDAC = low-dose cytarabine; SC = subcutaneous.

aHealth Canada–approved indication.

Source: Product monographs for venetoclax,10 azacitidine,11 and cytarabine.12

Stakeholder Perspectives

Patient Group Input

This section was prepared by CADTH staff based on the input provided by patient groups.

About the Patient Group(s) and Information Gathered

One patient advocacy group, the LLSC, provided input on venetoclax in combination with azacitidine for the treatment of AML.

The LLSC’s mission is to cure leukemia, lymphoma, Hodgkin’s disease, and myeloma, as well as to improve the QoL of all Canadians affected by blood cancers. The LLSC has received funding from AbbVie.

The LLSC used an online survey for its submission, which was conducted between December 7, 2020 and January 24, 2021.

A total of 29 patients responded. All respondents were from Canada: 13 from Ontario, 6 from Quebec, 6 from British Columbia, and 4 from Alberta. Patient ages ranged from 25 to 84 years old and 2 were 75 years old or older. There were 18 females and 10 males and 1 did not report gender. Comorbidities were not reported. All patients had been diagnosed with AML within the past 7 years. Five of the respondents had experience with venetoclax in combination with azacitidine.

Disease Experience

According to the patient respondents to the LLSC survey, the symptoms that patients with AML experience that impact QoL include fatigue, suddenness of symptom development, anxiety, fear of relapse (number of patients unspecified for preceding symptoms), and loss of eyesight (n = 1). One patient experienced a spleen rupture and was in a coma for 8 days. Fatigue was the symptom mentioned most often among patient respondents. Fatigue and other symptoms subsequently affected social and family life along with other symptoms. Patients reported that these symptoms compounded with the changes related to the coronavirus disease 2019 (COVID-19) pandemic, which led to further social isolation. Some patients reported they are unable to work due to their disease and associated symptoms. Many patients did not provide information on the specific symptoms they experienced but described being diagnosed with AML as a life-changing event. Below are comments from patients regarding their experiences with AML:

“Everything in my life stopped cold turkey-employment, social life, relationships, etc. I made a complete personal 360 degree pivot to focus on my healing and living.”

“Well COVID and my compromised immune system has caused me to be very socially isolated. I haven't seen some very important people in my life for almost 2 years at this point.”

When asked if there are any aspects or symptoms of AML that are easier to control, most patients (n = 7) indicated no, and 1 patient commented there was no control with AML. Three patients indicated exercise was helpful in alleviating some symptoms, reporting that exercise and keeping physically active helped, particularly with fatigue.

Two patients reported feeling no impact or back to normal at the time of survey.

AML affects not just those who are diagnosed, but also their caregivers, which may include a spouse, immediate family members, and friends. Patients reported needing assistance for physician visits and daily activities. According to the LLSC survey, patients reported that caregivers seemed to feel multiple emotions about the patient’s AML: stress, worry, sadness, insecurity, and fear of dying were all frequently mentioned. Their companion through the disease journey was important for patients.

Experiences With Currently Available Treatments

According to the LLSC survey, the front-line treatments that patients received after diagnosis included chemotherapy (n = 24), stem cell transplant or bone marrow transplant (n = 16), drug therapy (n = 6), radiation therapy (n = 5), and chimeric antigen receptor (CAR) T-cell therapy (n = 1). One patient reported receiving Vyxeos (daunorubicin and cytarabine). Patients reported a wide range of side effects with current treatments, and the ones they considered to have a large impact on their QoL included hair loss (n = 17); weakness (n = 15); extreme fatigue (n = 14); diarrhea (n = 10); infections (n = 8); anemia (n = 8); mouth sores (n = 8); nausea and vomiting (n = 7); fever (n = 6); low blood cell counts (n = 6); tingling sensations (n = 4); constipation (n = 2); graft-versus-host disease (n = 2); lung, heart, kidney, or nerve problems (n = 2); cough (n = 1); rashes (n = 1); shortness of breath (n = 1); and psychological distress (n = 1). The side effects due to chemotherapy and stem cell transplant had a large impact on patients’ QoL, summarized the LLSC survey administrator. These side effects from front-line treatments led to changes in physical activity (n = 15), anxiety (n = 11), problems in mental health and overall happiness (n = 11), eating challenges (n = 12), and social development (n = 6) and educational development (n = 6) challenges. Overall, the side effects from front-line treatments caused significant disturbance to daily living. Patients were isolated from visitors during stem cell transplant. Opportunistic infection could occur. The following are comments from patients regarding their experiences with front-line AML treatments:

“The main challenge was the nausea and vomiting. I didn’t seem to have much control over it and had my wonderful bucket always with me. I could be fast asleep and awake and vomit.”

“Your whole world changes when you are diagnosed with AML. Suddenly, you confront your mortality. You feel extremely weak, you have to go into hospital for months, and you don't realize you MUST go into remission to have a stem cell transplant.”

“Extremely tired and little desire to be active. Difficulty eating and keeping it down. A few days of low hemoglobin and fluid on the lung that caused shortness of breath.”

“The worst issue is that I have no more job and that the treatments made me lose a lot of concentration and I get exhausted easily.”

“Had to move to Vancouver for treatment for 9 months. 2 or 3 months total in hospitals. Daily outpatient care. Kinda turns your life upside-down.”

Patients who responded to the LLSC survey reported a mixture of both positive and negative experiences accessing treatments. Thirteen respondents reported generally positive experiences and some patients attributed their experience to the support from medical staff. Six patients reported negative experiences. Negative experiences were related to challenges with receiving care and treatment plans. Some patients needed to relocate to receive treatments.

Improved Outcomes

The majority of respondents to the LLSC survey indicated that the factors they considered about a new cancer treatment were physician recommendation (n = 19), possible impact on disease (n = 17), QoL (n = 12), closeness of home to the treatment centre (n = 9), and outpatient treatment (n = 8).

The LLSC survey patient respondents also reported the characteristics of new treatment options they hoped to have, particularly those that could maintain remission, have fewer side effects, be covered by public plans, and be accessible in their geographic region. The opportunity to have access to other supportive options, such as meditation, hypnosis, neuro-linguistic programming support, and awareness support (thoughts, emotions, and behaviours), was also mentioned.

Experience With Drug Under Review

Five of the LLSC respondents indicated they took venetoclax in combination with azacitidine. Three respondents received it through compassionate use from the pharmaceutical company and 2 respondents received it through physician prescription. Two respondents reported difficulties in accessing the drug because of the costs and 1 reported a long wait time. The financial costs reported by 3 patients ranged from $10,000 to $13,000. Three respondents took this treatment because of physicians’ recommendation and 2 patients considered side effects to be the main factor for adopting this treatment. One of them also considered the chance of survival the reason to take this treatment.

Patients’ responses to this treatment varied greatly, including an overall great experience (n = 1), experiences with side effects (tiredness and loss of appetite, n = 1), no side effects but relapse (n = 1), significant side effects (n = 1), and transition to transplant (n = 1). Serious and very serious side effects reported by patients were fatigue (n = 3), low platelets (n = 2), and anemia (n = 1). Other side effects were minor or manageable, such as diarrhea, nausea, constipation, cough, back pain, and headache. One patient thought multiple visits to the hospital, transfusion, and infection made managing the side effects more challenging.

Two patients strongly agreed that venetoclax in combination with azacitidine improved their QoL compared with other treatments they received. Two patients also agreed and 1 disagreed. Two patients thought this treatment led to remission and thus improved their QoL. Overall, patients’ experiences with this treatment varied greatly. Compared with other treatments they received, patients indicated this treatment was significantly more challenging (n = 2), more challenging (n = 1), neutral (n = 1), less challenging (n = 1), and significantly less challenging (n = 1). The 5 patients were willing to tolerate the side effects of this treatment. One patient tolerated the side effects to live and another thought there was no other choice.

Clinician Input

All CADTH review teams include at least 1 clinical specialist with expertise regarding the diagnosis and management of the condition for which the drug is indicated. Clinical experts are a critical part of the review team and are involved in all phases of the review process (e.g., providing guidance on the development of the review protocol, assisting in the critical appraisal of clinical evidence, interpreting the clinical relevance of the results, and providing guidance on the potential place in therapy). The following input was provided by 3 clinical specialists with expertise in the diagnosis and management of AML in adults.

Unmet Needs

The experts indicated that current lower-intensity treatments have low rates of CR, and CRs that are produced are not durable. Treatments producing higher rates of CR had increased toxicity and are not tolerable in the patient population under study. The Health Canada approval for azacitidine is only for treatment-naive patients with higher-risk MDS and AML (according to the International Prognostic Scoring System) and with up to 30% blasts by WHO classification, although certain local jurisdictions make azacitidine available for patients with 30% or greater blasts.

Place in Therapy

The experts indicated that venetoclax plus azacitidine (or other HMA) would change the current treatment paradigm. It would be the new standard of care for patients with treatment-naive AML aged 18 years or older who are not eligible for intensive chemotherapy, and would replace, for the most part, single-agent HMAs or LDAC. For patients with treatment-naive AML aged 75 years or older who were eligible for intensive chemotherapy, especially those with good or intermediate risk cytogenetics, there would have to be a discussion with the patient about the risks and benefits of the different treatment options. It should be noted there is no consistency as to the upper age limit at which an acute leukemia treatment centre would administer intensive chemotherapy. As venetoclax plus azacitidine is myelosuppressive, it may not be suitable for a small number of frail patients, or for those who would be unable to travel to the treating hospital for count checks. This, too, would need to be assessed by the treating physician in conjunction with the patient.

Patient Population

The experts indicated that patients must have a diagnosis of AML with greater than 20% blasts, and that making this diagnosis from blood and bone marrow is straightforward. Patients with isolated granulocytic sarcoma were not included in the trial.

The clinicians indicated that selection of patients for treatment would be based on clinician judgment and patient preferences. At this time, there is not enough information to make treatment decisions based on disease characteristics, and the data to predict response have not yet been validated in large studies.

Patients with good risk cytogenetics and patients with myeloproliferative neoplasm in blast crisis have been excluded from studies of venetoclax plus azacitidine or LDAC. Patients who had previously used an HMA were not eligible for the VIALE-A trial, but were eligible for the VIALE-C trial. One respondent indicated that studies suggest response to venetoclax plus HMA following HMA is similar to response to venetoclax plus LDAC; however, there are no direct comparisons of these 2 treatments post HMA. Patients with CNS involvement by AML have been excluded from all AML studies, but this does not mean this group of patients would not benefit from venetoclax plus azacitidine with concomitant intrathecal therapy, similar to the current practice of administering systemic intensive chemotherapy and intrathecal therapy to those patients who have CNS involvement by AML.

One expert suggested venetoclax with azacitidine was preferred over venetoclax plus LDAC in patients who had not received prior HMA. Venetoclax plus HMA was reasonable in patients with prior HMA use, and ivosidenib plus azacitidine would be reasonable in patients with IDH1 mutations, if ivosidenib were available.

The experts indicated that venetoclax plus azacitidine would not be suitable for fit patients (i.e., those eligible for standard induction treatment) with good cytogenetic risk AML, or for patients with acute promyelocytic leukemia. Emerging evidence may support the use of venetoclax plus azacitidine in fit patients as a bridge to allogeneic bone marrow transplant in higher-risk AML. Opinions differed in the use of venetoclax plus azacitidine in patients with relapsed or refractory disease, as venetoclax plus azacitidine has reduced activity in relapsed and refractory disease: 1 respondent did not recommend it, and another thought it was a reasonable option.

Assessing Response to Treatment

The experts indicated that, in clinical practice, response to treatment would be determined by achievement of CR, CRh, or CRi and/or transfusion independence or hematological improvement or stable disease. A clinically meaningful response to treatment would be represented by improved OS, improved EFS, achievement of durable CR, decreased hospitalizations, decreased transfusion requirements, slower progression, stabilization of disease (which would presumably improve or not worsen symptoms), and improved QoL. One clinician noted that the strict definitions of response did not necessarily identify responding patients. One clinician indicated that OS, hospital visits, transfusion dependence, and QoL were likely the most important end points. The clinicians indicated it is difficult to determine a minimum improvement over standard of care and what a meaningful response represents will vary by physician and patient.

Response would be measured by CBC and bone marrow blasts. One respondent indicated bone marrow biopsy should be performed after the first and second cycles of treatment, as response would be expected after a maximum of 2 cycles. Another indicated that response should be assessed at minimum after 4 to 6 cycles, but that most practitioners assess after the first cycle, given cost and to guide dosing of venetoclax for subsequent cycles. Once a response was obtained, then CBC could be followed for evidence of progression.

Discontinuing Treatment

The experts agreed that disease progression and intolerable adverse events were factors to be considered in the decision to discontinue treatment. Disease progression could be indicated by worsening CBC, increasing blasts in bone marrow, and/or loss of transfusion independence. The clinicians could not comment whether venetoclax could be continued if a patient stopped azacitidine.

Prescribing Conditions

The experts indicated that a hospital or outpatient clinic would be appropriate settings for treatment. As venetoclax plus azacitidine is myelosuppressive, physicians should have experience in looking after acute leukemia patients. Patients might require hospital admission for venetoclax dose ramping up. The proportion depends upon population: 1 respondent indicated the proportion would small, and another that it could be 25% to 50%. Patients would also require pre-treatment and monitoring for tumour lysis syndrome (occurring in 1% to 2% of patients). A not-insignificant proportion of patients will need to be hospitalized for neutropenic fever and other complications during their cycle of therapy. Pharmacists would be involved in reviewing medications, as a significant proportion of patients are on azoles, which interact with venetoclax and require dose modifications.

Clinician Group Input

This section was prepared by CADTH staff based on the input provided by clinician groups.

Four clinician groups provided input on the reimbursement review of venetoclax in combination with an HMA or in combination with LDAC, for the treatment of adult patients with newly diagnosed AML who are ineligible for intensive chemotherapy.

The ATB-MPG is a group of physicians who treat myeloid malignancies and acute leukemias (myelodysplastic syndromes, myeloproliferative neoplasms, AML, and acute lymphoblastic leukemia) within Alberta and function as a group within the Alberta Hematology Tumour Group. There are Edmonton and Calgary groups that meet regularly and provincially every 3 months as well as annually to update the treatment guidelines on these diseases for Alberta. Information was collected by ATB-MPG for this review through literature review and group discussions. The group reviews written guidelines in a group setting and modifies its opinions based on written and oral discussion among the members. The discussed information is then approved by the full group.

The CLSG is a cross-Canada collective of physicians who treat acute leukemia and who represent all major leukemia centres in all provinces. The purpose of CLSG is “to improve the diagnosis and treatment of leukemia in Canada by identifying diagnostic and management best practices, promoting Canada-wide standards of care, fostering clinical and basic leukemia research, and improving new drug access.” Information for this review was gathered through ongoing group discussions and polling of members, with input requested from other international experts as appropriate. The written opinions were further edited and approved by the full group.

Ontario Health’s (Cancer Care Ontario) Hematology Disease Site Drug Advisory Committee (OH-CCO Hem-DAC) provides evidence-based clinical and health system guidance on drug-related issues in support of OH-CCO’s mandate, including provincial drug reimbursement programs and the Systemic Treatment Program. The group gathered its information for this review through discussions at the DAC meeting.

The L/BMT Program of British Columbia is a joint program of BC Cancer and Vancouver Coastal Health with a primary mandate for the province to treat acute leukemia, perform stem cell transplantation, and deliver cellular therapies for patients with hematologic malignancies. Members from the acute working group within the program provided input for this review. The group primarily reviewed published data from the phase III trial (DiNardo)23 as well the data from the phase Ib trial and reviewed National Comprehensive Cancer Network (NCCN) guidelines.

Unmet Needs

All 4 clinician groups agreed that the current treatment paradigm for this disease includes azacitidine, LDAC, and BSC.

In addition, the L/BMT program of British Columbia noted that determination of eligibility for intensive chemotherapy is based on patient age, fitness, presence of comorbidities, and patient preferences. The group added that in British Columbia, azacitidine is given at many BC Cancer sites as well as in other hospital outpatient settings depending on the geographic location and treating physician. The treatment is given as a subcutaneous injection usually in either a 7-day or 5 days plus 2 days schedule. L/BMT members added that response to azacitidine is not evident before cycles 3 to 4 and is usually formally assessed around cycles 6 to 7 with a repeat bone marrow aspirate and biopsy. The group added that treatment with azacitidine continues indefinitely as long as the patient benefits and tolerates the treatment. L/BMT noted that LDAC is also given as a subcutaneous injection of 20 mg twice a day for 10 days every 4 to 6 weeks and can be given by the patient or a caregiver at home and also requires patient education by a chemotherapy-trained nurse before initiation. The group noted that LDAC is given less frequently than azacitidine and is beneficial for patients who live a long distance from a cancer centre that administers azacitidine or who would prefer to receive treatment at home. L/BMT also noted that in the province of British Columbia, LDAC is generally reserved for patients with intermediate risk karyotype. L/BMT added that older patients with AML also receive supportive treatments either with azacitidine or LDAC or alone, and these supportive treatments include transfusion support, hydroxyurea, antibiotic treatment, pain control, and palliative care. The group noted that currently there are no relevant special access programs available and that novel treatments for AML and non-intensive treatments with LDAC and azacitidine can improve symptoms and result in clinical responses, with a small proportion of patients achieving CR. L/BMT added that this treatment (venetoclax plus azacitidine) is associated with an improvement in OS, but it is not considered curative and responding patients will ultimately have disease progression.

The ATB-MPG added that, currently, for AML patients who are ineligible for induction chemotherapy, the common clinical practice for the majority of patients is to use azacitidine 75 mg/m2 per day for 7 days every 28 days, and some patients receive cytarabine 20 mg twice daily for 10 days. For elderly patients, the group noted there was no survival benefit noted with cytarabine, and azacitidine is preferred. They also added that azacitidine is approved in Canada for patients with low blast count AML (20% to 30%), but is commonly used and provides clinical benefit to patients unfit for induction chemotherapy who have blast counts higher than 30%. In addition, the group noted there is a temporary compassionate access to an oral decitabine and cedazuridine compound that is available to patients with low blast count AML (20% to 30%); however, ongoing access to this drug is not yet established. The ATB-MPG agreed that palliative basic supportive care options include hydroxyurea and blood transfusion support as well as antibiotics, and patients may be offered clinical trials when they are available. The group noted that patients are aware of venetoclax and azacitidine and some have been “self-funding” venetoclax by using CYP3A inhibitors to reduce the dose, and thereby the cost, of venetoclax.

CLSG agreed with the other clinician groups that approximately 40% to 50% of newly diagnosed AML patients are judged to be unfit for intensive induction chemotherapy and this includes patients aged 75 and older as well as younger patients with severe comorbidities. For these patients, CLSG noted that the treatment options include azacitidine, LDAC, or BSC alone. The group added that for patients with poor risk cytogenetics or AML transformed from MDS, azacitidine is the current treatment of choice, while for patients with AML arising de novo with standard risk cytogenetics, azacitidine or LDAC can be used. The group noted that in the real-world clinical setting, many patients in Canada are not able to receive azacitidine-based therapy, as this drug needs to be given in an oncology clinic setting due to its instability after reconstitution. As a result, many patients who live in rural areas and some in urban settings are unable to travel to these clinics regularly to receive treatment because of the distances, overall frailty, and challenges in obtaining suitable transportation.

All 4 clinician groups agreed with most important treatment goals. These included improvement in survival, improvement in QoL, improvement in hematopoiesis, and transfusion independence. The L/BMT program of British Columbia added that prevention of infection, time to remission, and minimization of toxicity and adverse events associated with treatment are also important treatment goals. In addition to these objectives, the ATB-MPG noted that the reduction of burden on caregivers is also an important treatment goal. All 4 clinician groups also agreed that the currently available treatments offer short survival advantage and short transfusion independence. The short remissions often require several monthly cycles of the therapy, up to 6 cycles, to achieve maximal effect, and translate to an extended period of transfusion dependence, as noted by CLSG. CLSG also added that once a maximally achieved response is lost, disease progression is quite rapid, typically followed quickly by death due to poor salvage therapies. The ATB-MPG added that time to remission for azacitidine is around 4 months and maximal response can take more than 6 months; during this time, patients are transfusion-dependent and have significant burden of disease. The treatment under review was noted by the ATB-MPG as having a median time to response of 1.2 months (faster time to response and clinical improvement). The L/BMT program of British Columbia added that the primary unmet goals for this population are low response rates and short OS.

All 4 clinician groups also agreed that patients with AML who are not eligible for standard 7 plus 3 induction therapy (older or with comorbidities) have the greatest unmet need.

Place in Therapy

All 4 clinician groups agreed that the combination of venetoclax and azacitidine would replace current front-line therapies, including azacitidine alone in a population that has an unmet medical need. The ATB-MPG noted that some patients who received induction chemotherapy and relapsed but are no longer eligible for transplantation, or who relapsed after transplantation and had never received an HMA such as azacitidine before, would commonly use azacitidine and the combination of azacitidine and venetoclax would be expected to be more effective; however, this population was not included in the current reimbursement request.

All 4 clinician groups also agreed it would not be appropriate to recommend that patients try other treatments before initiating treatment with venetoclax and azacitidine, as this therapy is for first-line use and there is no evidence to support sequencing this combination after other treatments. The groups added that the clinical trial was also for newly diagnosed patients.

With respect to sequencing, all 4 clinician groups agreed that the combination of venetoclax and azacitidine would replace current first-line treatment. CLSG also added that after failure of the combination of venetoclax and azacitidine, possible therapeutic options would include therapy targeted to a specific molecular lesion, if present and available, an early phase clinical trial, LDAC, BSC, or palliation. L/BMT agreed with CLSG on this approach and noted that, currently, there is no standard of care practice for patients who fail first-line treatment. The ATB-MPG noted that in second-line treatment, if patients have FLT3-positive AML, they can receive gilteritinib and, if they have FLT3-negative AML, they can receive cytarabine if they are being treated with azacitidine, or with azacitidine if they are being treated with cytarabine. In third-line treatment, the ATB-MPG noted that hydroxyurea and transfusions as well as basic supportive care can be used.

Patient Population

All 4 clinician groups agreed that patients with newly diagnosed AML who are unfit for intensive chemotherapy (due to age, comorbidities, or patient decision not to undergo intensive treatment that is potentially curative) are best suited for treatment with venetoclax and azacitidine, as these patients need more effective therapy options. The L/BMT program of British Columbia noted that the age cut-off of 75 years or greater, as per the Health Canada indication, is based on the clinical trial; however, they expect that, in most centres in Canada, only a small number of patients older than 70 years of age are treated with intensive chemotherapy. The group also added there is some evidence that AML patients with mutations in genes 1DH1 or 1DH2 have a particularly good response to azacitidine and venetoclax, but patients with other genetic subsets of AML as well as de novo and secondary AML also appear to benefit.

All 4 clinician groups also agreed there is a standard diagnosis of AML (i.e., the presence of greater than 20% myeloid blasts in the bone marrow or peripheral blood). Through clinical examination and judgment, as well as bone marrow results, patients can be objectively diagnosed. The groups added that testing is widely available and that patients should be treated at the time of diagnosis, as they would be expected to decline rapidly and develop serious infections or other complications that preclude effective treatment if they are not treated at diagnosis.

It was noted that patients who are least suitable for the venetoclax and azacitidine treatment are those who are younger and fit, without significant comorbidities, or very frail older adults. The clinician groups also noted that patients who are unable to travel to outpatient clinicians to receive azacitidine would be least suitable for treatment. Also, the L/BMT program of British Columbia noted that patients who are not able to have regular blood work monitoring for tumour lysis syndrome during the initial ramp-up and regular monitoring of blood work later on are also not good candidates.

With respect to identifying patients who are most likely to exhibit a response to treatment with venetoclax and azacitidine, the clinician groups noted that some patients with specific molecular mutations (IDH1 or IDH2) on next-generation sequencing may be expected to respond better; however, this is based on subgroup analysis and there is no patient group that would not be expected to benefit within the group of patients this reimbursement request is for. The groups also noted that at this point, there is no specific test or biomarker to indicate who will or will not respond or benefit from treatment.

Assessing Response to Treatment

All 4 clinician groups agreed that important outcomes in clinical practice are remission status following treatment, tolerance of treatment, QoL, and transfusion requirements. The groups added that bone marrow biopsy to assess disease response is an important outcome; however, it was done more frequently in the clinical trial than in practice.

All 4 groups agreed that a clinically meaningful response to treatment would be remission status on bone marrow biopsy, reduced or eliminated transfusion requirements for red blood cells and platelets, and improvement in symptoms (i.e., infections, bleeding, improved functional status due to improved hemoglobin and fewer hospital admissions or outpatient visits for transfusion support). Additionally, the groups added that in patients without CR or CRi (incomplete count recovery), a partial remission or improvement in blood counts may also be a meaningful improvement for some patients.

CLSG, L/BMT program of British Columbia, and the ATB-MPG noted that response should be assessed with a bone marrow biopsy as well as evaluation of blood counts following 1 to 2 cycles (4 to 8 weeks) of azacitidine and venetoclax. The L/BMT program of British Columbia noted that in patients achieving CR or CRi, they would suggest repeating the bone marrow aspirate biopsy as clinically indicated (e.g., repeated if there is concern that a patient is losing response due to worsening blood counts or the appearance of circulating blasts). The group added that in patients with less than CR or CRi after 1 to 2 cycles who continue on treatment, they would generally repeat bone marrow biopsies every 3 to 4 months to evaluate a response. The ATB-MPG added that once remission or maximal response is obtained, repeat bone marrow biopsy would be indicated if there is clinical deterioration or significant cytopenias requiring reassessment of disease status. The OH-CCO’s Hem-DAC noted that treatment should be assessed frequently with regular CBC and bone marrow assessments, as per clinician judgment.

Discontinuing Treatment

All 4 clinician groups agreed that adverse events such as severe nausea, neutropenic infections, and severe infections may lead to a decline in patients’ ability to safely administer the treatment. The groups also agreed that failure of response or disease progression (significant increase in bone marrow blasts), treatment-related toxicities, and patient preference would be the primary reasons for treatment discontinuation.

Prescribing Conditions

All 4 clinician groups agreed that the most appropriate setting for treatment administration can be in the community setting and outpatient clinic. Inpatient hospital treatment may also be required due to tumour lysis syndrome or AML complications while continuing on the treatment. CLSG noted there should be expertise in the outpatient clinic with chemotherapy preparation and administration. The L/BMT program of British Columbia noted that treatment with venetoclax and azacitidine requires monitoring of blood counts, renal function, and electrolytes more frequently early on during the first week of administration due to the risk of tumour lysis syndrome. CLSG was of the opinion that the treatment should be given in a setting where there is blood-bank support; physician, nursing, and pharmacy expertise in chemotherapy; an ability to deliver IV fluids and antibiotics; and an ability to admit patients to hospital for complications of treatment.

Additional Considerations

OH-CCO’s Hem-DAC noted that venetoclax dose adjustment with co-administration of azole is sometimes required. In addition, the group added that in patients presenting with hyperleukocytosis, a longer ramp-up phase should be considered when initiating venetoclax.

The group added that the additional toxicities of the combination are largely related to increased myelosuppression and increased rates of febrile neutropenia early on during treatment, but this is manageable and does not offset the benefit of the treatment combination. The group also strongly supported the reimbursement of this treatment for older and unfit patients with AML due to the large, anticipated benefit for this group of patents, where there currently exists an unmet need for more effective treatment options.

CLSG noted that patients are already treated in many jurisdictions with azacitidine alone and adding an oral medication, like venetoclax, which is well tolerated with a straightforward administration schedule, does not increase the complexity of the treatment regimen. The group also added that the benefits obtained with this combination are seen much quicker than with azacitidine alone.

Drug Program Input

The drug programs provide input on each drug being reviewed through CADTH’s reimbursement review processes by identifying issues that may impact their ability to implement a recommendation.

The drug programs indicated that current treatment options for patients with newly diagnosed AML who are ineligible for intensive chemotherapy include azacitidine, LDAC, and BSC. The reimbursement of venetoclax plus azacitidine would likely replace single-agent azacitidine in this treatment setting. Azacitidine is funded in most jurisdictions for patients with AML who are ineligible for intensive chemotherapy, and some jurisdictions fund alternate dosing schedules for azacitidine (i.e., 5 to 2-2, and 6 consecutive days) in addition to the schedule of 7 consecutive days. However, it was noted that some patients 75 years of age and older may be fit to tolerate intensive chemotherapy. The ramp-up dosing schedule for venetoclax with azacitidine differs significantly from the ramp-up dosing schedule already in use for CLL indications, and the current packaging for venetoclax is designed for the CLL ramp-up dosing schedule. Venetoclax plus azacitidine includes an oral and an IV and subcutaneous drug and, therefore, would be reimbursed through different programs in some jurisdictions. The drug programs identified the potential for indication creep for patients with a high risk of MDS, those who have progressed or have had an inadequate response on low-dose chemotherapy for AML, and patients who have relapsed after induction chemotherapy and are not eligible for stem cell transplant and who are then treated with azacitidine. It was noted that treatment combination may require the need for increased health care resources (i.e., hospital admission and additional pharmacy and nursing resources for the potential management of tumour lysis syndrome and monitoring for drug interactions). Affordability was also identified as an issue since the combination is expected to replace azacitidine monotherapy.

The implementation questions and corresponding responses from the clinical experts consulted by CADTH are summarized in Table 4.

Table 4: Summary of Drug Plan Input and Clinical Expert Response

Drug program implementation questions

Clinical expert response

Are all patients with newly diagnosed AML who are ineligible for treatment with intensive induction chemotherapy, regardless of cytogenetic risk, eligible for treatment with venetoclax plus azacitidine?

Although the VIALE-A trial excluded patients with favourable cytogenetic risk (defined according to NCCN guidelines for AML), all patients who are considered ineligible for treatment with intensive induction chemotherapy should be eligible for treatment with venetoclax plus azacitidine.

AML patients who previously received azacitidine for treatment of MDS were not eligible for treatment with venetoclax plus azacitidine in the VIALE-A trial but were included in the VIALE-C trial and eligible for treatment with venetoclax plus LDAC. Would these patients not be eligible for treatment with venetoclax plus azacitidine?

The VIALE-A trial excluded patients who had received previous treatment with an HMA for MDS; therefore, there is no evidence from the pivotal trial on the efficacy of venetoclax plus azacitidine in this group of patients. However, there is non-comparative clinical trial evidence13 demonstrating that patients previously treated with azacitidine for MDS benefit from venetoclax plus azacitidine; the response rate, although lower than what has been observed in patients without prior exposure to azacitidine, is comparable to the response rate observed in the VIALE-C trial among patients with prior exposure to an HMA treated with venetoclax plus LDAC.14 Based on these data, it would be reasonable to consider the use of venetoclax plus azacitidine in this subgroup of patients.

Can venetoclax be used with alternate azacitidine dosing schedules (e.g., 5-2-2 for 6 consecutive days)?

In clinical practice, azacitidine is usually administered on a 5-2-2 dosing schedule. There is evidence15 demonstrating there is no difference in clinical outcome based on the dosing schedule used (i.e., 5-2-2, 6 and 7 consecutive days); therefore, venetoclax can be used with alternative azacitidine dosing schedules.

The highest strength of venetoclax available is a 100 mg tablet. At full dose, patients will need to take 4 × 100 mg tablets to make up the dose, which is a high pill burden. Is there a plan to manufacture a higher-strength tablet?

Is any supportive care required during ramp-up (i.e., for TLS prophylaxis)?

During the ramp-up period of venetoclax, patients need to be treated in a setting where they can be monitored daily and would be treated with allopurinol as prophylaxis for TLS.

Hydroxyurea should be administered to patients with a high WBC count to lower the WBC to less than 25 × 109/L before administering venetoclax to reduce the risk of developing TLS (same as on study).

There are differences in the eligibility criteria of the trials, VIALE-A and VIALE-C. Should the eligibility criteria for venetoclax plus azacitidine be consistent with the criteria for venetoclax plus LDAC?

Should the following patients be considered for treatment with venetoclax plus azacitidine:

  • patients who have received prior HMA (azacitidine) or chemotherapy for the treatment of MDS (these patients were excluded from the VIALE-A trial)

  • patients with favourable cytogenic risk (these patients were excluded from the VIALE-A trial)

  • patients ≥ 75 years of age with an ECOG Performance Status greater than 2 (these patients were excluded from VIALE-A trial).

Although there were some differences in the patient eligibility requirements for each trial, criteria for reimbursement should be consistent for both venetoclax-based regimens.

Regarding the eligibility of the following patient groups:

  • As noted earlier, patients who have received prior HMA or chemotherapy for the treatment of MDS and patients with favourable cytogenetic risk should be eligible for venetoclax plus azacitidine.16

  • Patients ≥ 75 years of age with an ECOG Performance Status greater than 2 may be eligible for venetoclax plus azacitidine, depending on whether their performance status is judged to be related to their AML; therefore, eligibility for venetoclax plus azacitidine should be determined for these patients on an individual basis.

  • Only 4 patients (2%) in the VIALE-C trial had favourable risk cytogenetics (3 patients in the placebo + LDAC arm and 1 patient in the venetoclax + LDAC arm); this is not a significant difference between the 2 studies.

Can venetoclax plus azacitidine be given to improve response as a bridge to allogeneic SCT in patients with AML who have a contraindication to chemotherapy, but are otherwise candidates for an allogeneic SCT?

It is uncommon to have a patient with a contraindication to chemotherapy proceed to allogeneic SCT, but it may happen in some circumstances (e.g., for patients who have an ejection fraction of less than 50% and hence have a comorbidity that renders them ineligible for intensive chemotherapy). These patients may achieve a response to venetoclax plus azacitidine and a (reduced intensity conditioning) allogeneic SCT could be considered. Evidence is emerging on the use of venetoclax plus azacitidine as a bridge to allogeneic SCT, especially in patients with treatment-naive, poor-risk AML who are fit for intensive chemotherapy (due to high response rates, low early deaths or induction deaths, and/or infections) and, as such, the combination is being used for this indication in other countries.17-20 Hence, it would be reasonable to use venetoclax plus azacitidine as a bridge to allogeneic SCT in this small group of patients. However, long-term outcomes are limited.

There is a time-limited need to allow patients currently on azacitidine whose disease has not yet progressed to add venetoclax who otherwise meet the eligibility criteria. What is the appropriate time frame of treatment on azacitidine to consider the addition of venetoclax?

There is no evidence to inform the appropriate time frame to consider adding venetoclax for patients who are receiving azacitidine alone. In general, clinicians typically give up to 6 cycles (i.e., 6 months) of azacitidine alone to determine a patient’s response to therapy. Therefore, it would be reasonable to add venetoclax to azacitidine if patients were within the 6-month time frame of initiating azacitidine and had not progressed. The value of adding venetoclax for a patient who has achieved a response or remission on azacitidine alone is unknown.

Inpatient administration may be required during the ramp-up portion for venetoclax. Are there specific groups or an estimated percentage of patients who would require hospital admission for the ramp-up portion of venetoclax?

Hospital administration will be required for some patients and this is not necessarily limited to the ramp-up portion of venetoclax. This is an older patient population of whom some may be frail; and patients may develop febrile neutropenia or infection any time during the treatment window.

It is difficult to estimate, but up to 30% of patients may require hospitalization during the ramp-up portion of venetoclax, and this may vary depending on the treatment setting (i.e., treatment centre vs. community where they may not have the appropriate resources to monitor for TLS daily during the ramp-up period). However, this percentage is expected to decrease over time as clinicians become more experienced with administering venetoclax. Special groups of patients who may be an increased risk of hospitalization during the ramp-up period include those who have an elevated WBC count, high tumour burden, or underlying renal insufficiency.

Are patients with a good risk prognosis eligible for venetoclax plus azacitidine? Please confirm that all cytogenetic risks are eligible for treatment with venetoclax plus azacitidine.

As previously noted, all patients considered ineligible for intensive induction chemotherapy, regardless of prognostic or cytogenetic risk, should be eligible for venetoclax plus azacitidine.

If a patient stops treatment with the azacitidine component for reasons other than disease progression, can the venetoclax be continued until disease progression?

The VIALE-A clinical trial did not have a provision for patients to stop azacitidine and continue on venetoclax or placebo.

AML = acute myeloid leukemia; ECOG = Eastern Cooperative Oncology Group; HMA = hypomethylating agent; LDAC = low-dose cytarabine; MDS = myelodysplastic syndrome; NCCN = National Comprehensive Cancer Network; SCT = stem cell transplant; TLS = tumour lysis syndrome; WBC = white blood cell.

Clinical Evidence

The clinical evidence included in the review of venetoclax (Venclexta) in combination with azacitidine is presented in 3 sections. The first section, the systematic review, includes pivotal studies provided in the sponsor’s submission to CADTH and Health Canada, as well as those studies that were selected according to an a priori protocol. The second section includes indirect evidence from the sponsor and indirect evidence selected from the literature that met the selection criteria specified in the review. The third section includes sponsor-submitted long-term extension studies and additional relevant studies that were considered to address important gaps in the evidence included in the systematic review.

Systematic Review (Pivotal and Protocol Selected Studies)

Objectives

To perform a systematic review of the beneficial and harmful effects of venetoclax in combination with azacitidine for the treatment of patients with newly diagnosed AML who are 75 years or older, or who have comorbidities that preclude use of intensive induction therapy.

Methods

Studies selected for inclusion in the systematic review will include pivotal studies provided in the sponsor’s submission to CADTH and Health Canada, as well as those meeting the selection criteria presented in Table 5. Outcomes included in the CADTH review protocol reflect outcomes considered to be important to patients, clinicians, and drug plans.

Table 5: Inclusion Criteria for the Systematic Review

Criteria

Description

Patient population

Patients newly diagnosed with AML who are 75 years or older or who have comorbidities that preclude use of intensive induction therapy

Subgroups:

• age (75 years or older) or comorbidities

• Eastern Cooperative Oncology Group Performance Status

• prior myelodysplastic syndrome or myeloproliferative neoplasm

• prior exposure to chemotherapy or radiation due to other malignancies

• primary or secondary malignancy (secondary or therapy-related AML)

• cytogenetic risk

• blast count

• mutations (IDH1 and IDH2, FLT3, NPM1, TP53)

Intervention

Venetoclax oral 400 mg per day (every day) and azacitidine 75 mg/m2 per day, IV or SC (days 1 through 7 of 28-day cycle)

Comparators

Azacitidine monotherapy

Low-dose cytarabine monotherapy

Venetoclax + low-dose cytarabine

Induction chemotherapy (for patients aged 75 years or older)a

Best supportive care

Outcomes

Overall survivalb

Event-free survivalb

Complete remission rate with and without hematological recovery

Partial remission or hematological improvement

Time to remission

Duration of remission

Need for transfusion, transfusion independence

Hospital admission

Patient quality of life

Symptom severity

Harms outcomes:

• AEs, SAEs, TEAEs, WDAEs, mortality

Notable harms and harms of special interest:

• neutropenia

• febrile neutropenia

• infections

• tumour lysis syndrome

• hemorrhage

• secondary malignancies

Study design

Published and unpublished phase III and IV RCTs

AE = adverse event; AML = acute myeloid leukemia; CR = complete remission; CRi = complete remission with incomplete hematologic recovery; HRQoL = quality of life; LDAC = low-dose cytarabine; QoL = quality of life; RCT = randomized controlled trial; SAE = serious adverse event; TEAE = treatment-emergent adverse event; WDAE = withdrawal due to adverse event.

aInduction chemotherapy was added as a comparator based on feedback from the clinical experts consulted by CADTH on this review, as it is considered a potential option for approximately 10% of patients who are aged 75 years or older.

bThese outcomes were identified as being of particular importance to patients in the input received by CADTH from patient groups.

The literature search for clinical studies was performed by an information specialist using a peer-reviewed search strategy according to the PRESS Peer Review of Electronic Search Strategies checklist (https://www.cadth.ca/resources/finding-evidence/press).21

Published literature was identified by searching the following bibliographic databases: MEDLINE All (1946–) through Ovid and Embase (1974–) through Ovid. The search strategy comprised both controlled vocabulary, such as the National Library of Medicine’s MeSH (Medical Subject Headings), and keywords. The main search concepts were Venclexta (venetoclax) and AML. Clinical trials registries were searched: the US National Institutes of Health’s clinicaltrials.gov, WHO’s International Clinical Trials Registry Platform (ICTRP) search portal, Health Canada’s Clinical Trials Database, and the European Union Clinical Trials Register.

No filters were applied to limit the retrieval by study type. Retrieval was not limited by publication date or by language. Conference abstracts were excluded from the search results. See Appendix 1 for the detailed search strategies.

The initial search was completed on February 11, 2021. Regular alerts updated the search until the meeting of the CADTH pan-Canadian Oncology Drug Review Expert Committee (pERC) on June 10, 2021.

Grey literature (literature that is not commercially published) was identified by searching relevant websites from the Grey Matters: A Practical Tool For Searching Health-Related Grey Literature checklist (https://www.cadth.ca/grey-matters).22 Included in this search were the websites of regulatory agencies (US FDA and European Medicines Agency). Google was used to search for additional internet-based materials. See Appendix 1 for more information on the grey literature search strategy.

These searches were supplemented through contacts with appropriate experts. In addition, the sponsor of the drug was contacted for information regarding unpublished studies.

Two CADTH clinical reviewers independently selected studies for inclusion in the review based on titles and abstracts, according to the predetermined protocol. Full-text articles of all citations considered potentially relevant by at least 1 reviewer were acquired. Reviewers independently made the final selection of studies to be included in the review, and differences were resolved through discussion.

Findings From the Literature

One study23 was identified from the literature for inclusion in the systematic review (Figure 1). The included study is summarized in Table 6. A list of excluded studies is presented in Appendix 2.

Figure 1: Flow Diagram for Inclusion and Exclusion of Studies

Depicts the process of selection of studies, with number of “Citations identified in literature search” and “Potentially relevant reports from other sources” as inputs, and number of “Reports excluded” and “Reports included” as outputs.

Table 6: Details of Included Studies

Detail

VIALE-A

Designs and populations

Study design

Phase III, multi-centre, double-blind, placebo-controlled RCT

Locations

134 sites Canada, Europe, Russia, Asia, US, and South Africa

Patient enrolment dates

February 6, 2017 to May 31, 2019

Randomized (N)

433 (2 randomized under stratification criteria of original protocol)

Inclusion criteria

AML by WHO criteria, previously untreated and ineligible for treatment with standard cytarabine and anthracycline due to age or comorbidities

Ineligible for induction therapy defined by the following:

  • age ≥ 75 years, or

  • age 18 to 74 years with at least 1 of:

    • ECOG PS 2 or 3

    • history of CHF requiring treatment, EF ≤ 50%, or chronic stable angina

    • DLCO ≤ 65% or FEV1 ≤ 65%

    • creatinine clearance ≥ 30 mL/min to 45 mL/min

    • moderate hepatic impairment with total bilirubin > 1.5 to ≤ 3.0 ULN

    • other comorbidity considered incompatible with intensive chemotherapy, as reviewed and approved by sponsor

ECOG PS 0 to 2 (≥ 75 years), 0 to 3 (18 to 74 years)

Adequate renal function as demonstrated by creatinine clearance ≥ 30 mL/min calculated by Cockcroft Gault formula or measured by 24 hours of urine collection

Adequate liver function as demonstrated by:

  • aspartate aminotransferase (AST) ≤ 3.0 × ULNa

  • alanine aminotransferase (ALT) ≤ 3.0 × ULNa

  • bilirubin ≤ 1.5 × ULNa (patients < 75 years could have bilirubin ≤ 3.0 × ULN)

Exclusion criteria

Has received treatment with the following:

  • HMA and/or any chemotherapeutic drug for MDS

  • CAR T-cell therapy

  • experimental therapy for MDS or AML

  • current participation in another research or experimental study

History of myeloproliferative neoplasm, including myelofibrosis, essential thrombocythemia, polycythemia vera, chronic myeloid leukemia with or without BCR-ABL1 translocation, and AML with BCR-ABL1 translocation

Acute promyelocytic leukemia

Known active CNS involvement with AML

Known HIV infection

Known hepatitis B or C infection, with the exception of those with undetectable viral load within 3 months of screening

Received strong and/or moderate CYP3A inducers within 7 days before initiation of study treatment

Consumed grapefruit, grapefruit products, Seville oranges (including marmalade containing Seville oranges) or starfruit within 3 days before the initiation of study treatment

Cardiovascular disability status of NYHA class 2

Chronic respiratory disease requiring continuous oxygen; significant history of renal, neurologic, psychiatric, endocrinologic, metabolic, immunologic, or hepatic disease; cardiovascular disease; any other medication condition, or known hypersensitivity to any of the study medications, including excipients of azacitidine that in the opinion of the investigator would adversely affect the patient’s participation in the study

History of other malignancies within 2 years before study entry, with the exception of:

  • adequately treated in situ carcinoma of the cervix uteri or carcinoma in situ of breast

  • basal cell carcinoma of the skin or localized squamous cell carcinoma of the skin

  • previous malignancy confined and surgically resected (or treatment with other modalities) with curative intent (required discussion with sponsor)

White blood cell count > 25 × 109/L (hydroxyurea or leukapheresis were permitted to meet this criterion)

Drugs

Intervention

Venetoclax 400 mg per day after dose titration plus azacitidine 75 mg/m2 (days 1 to 7 of 28-day cycle), SC or IV. Venetoclax dose titration, cycle 1: day 1 = 100 mg, day 2 = 200 mg, day 3 and thereafter = 400 mg per day.

Comparator(s)

Azacitidine 75 mg/m2 (days 1 to 7, 28-day cycle) SC or IV

Duration

Phase

Double-blind

Until disease progression per investigator assessment, unacceptable toxicity, withdrawal of consent, or patient met other protocol criteria for discrimination

Follow-up

Survival information and post-treatment follow-up for approximately 2 years after enrolment of last patient. For patients discontinuing for a reason other than disease progression, hematological and disease assessment until approximately 1 year after last patient.

Outcomes

Primary end point

OS

CR + CRi

Secondary and exploratory end points

Secondary:

  • CR

  • CR + CRh

  • CR + CRi by initiation of cycle 2

  • EFS

  • transfusion independence

  • MRD response

  • fatigue

  • HRQoL

  • OS and CR + CRi in molecular subgroups

Exploratory:

  • biomarkers predictive of venetoclax activity and DOR

  • HRQoL (additional measures)

  • health utility

Notes

Publications

Di Nardo (2020)23

AML = acute myeloid leukemia; CAR T-cell therapy = chimeric antigen receptor T-cell therapy; CHF = congestive heart failure; CNS = central nervous system; CR = complete remission; CRh = complete remission with partial hematological recovery; CRi = complete remission with incomplete hematological recovery; DB = double-blind; DLCO = diffusing capacity of the lungs for carbon monoxide; DOR = duration of response; ECOG PS = Eastern Cooperative Oncology Group Performance Status; EF = ejection fraction; EFS = event-free survival; FEV1 = forced expiratory volume in 1 second; HMA = hypomethylating agent; HRQoL = health-related quality of life; MDS = myelodysplastic syndrome; MRD = minimal/measurable residual disease; NYHA = New York Heart Association; OS = overall survival; RCT = randomized controlled trial; SC = subcutaneous; ULN = upper limit of normal.

Note: Additional VIALE-A reports included a Clinical Study Report1 and patient-reported outcome report.1

aUnless considered due to leukemic organ involvement.

Source: Clinical Study Report.1

Description of Studies

VIALE-A

VIALE-A is a phase III, randomized, double-blind, placebo-controlled, multi-centre study comparing venetoclax plus azacitidine with placebo plus azacitidine in adults with newly diagnosed AML aged 18 years or older and ineligible for standard induction therapy due to age or comorbidities. The trial was conducted at 134 sites in Europe, Asia, South America, Canada, and the US. Trial characteristics are summarized in Table 6.

The primary objective was to evaluate whether venetoclax plus azacitidine would improve OS and composite complete remission rate (CR + CRi) compared with placebo plus azacitidine.

The secondary objectives were to evaluate whether, compared with placebo plus azacitidine, venetoclax plus azacitidine would improve the CR rate, CR + CRh rate, CR + CRi rate by the initiation of cycle 2, transfusion-independence rate, MRD response rate, response rates and OS in molecular subgroups, fatigue, GHS/QoL, and EFS. Improvement of fatigue and GHS/QoL were measured using patient-reported outcome assessments (Patient-Reported Outcomes Measurement Information System Short Form v1.0–Fatigue 7a [PROMIS 7a] and European Organization for Research and Treatment of Cancer Quality of Life Questionnaire Core 30 [EORTC QLQ-C30]). Definitions and further details of outcomes are given in Table 8.

Exploratory objectives included the study of biomarkers predictive of venetoclax plus azacitidine activity and the evaluation of additional subscales and items from the EORTC QLQ-C30 and EuroQol 5-Dimensions 5-Levels questionnaire (EQ-5D-5L) scales.

A total of 579 patients were screened and 433 patients were randomized in a 2:1 ratio to venetoclax plus azacitidine (287 patients), or placebo plus azacitidine (146 patients). Randomization was stratified for age (18 years to < 75 years, ≥ 75 years) and cytogenetic risk (intermediate, poor) for protocol amendment 1 and subsequent amendments. Prior to amendment 1, stratification was by age and region; the 2 patients (1 in each group) randomized under the original protocol were not included in the efficacy analysis. An additional open-label cohort of up to 12 patients was recruited to receive venetoclax plus azacitidine so as to provide pharmacokinetic and safety data required by the Chinese regulatory authorities before the recruitment of Chinese patients into the double-blind randomized portion of the study. These patients were not included in the efficacy or safety analyses for the main study and will not be described further in this report.

The first patient was randomized on February 6, 2017 and the last on May 31, 2019. Analysis of the co-primary efficacy end point of CR + CRi was planned for 6 months after the first 225 patients were randomized (first interim analysis [IA1]), at which time an interim analysis of survival was conducted. Interim data were reviewed by an independent data monitoring committee (IDMC) and the trial proceeded blinded. A second interim analysis (IA2) was planned when approximately 270 OS events accrued, before the final analysis at approximately 360 OS events. Review of the results against preplanned stopping criteria led to a recommendation by the IDMC to stop the trial on March 16, 2020. Data cut-off for IA1 was October 1, 2018 and January 4, 2020 for IA2.

Populations

Inclusion and Exclusion Criteria

Eligible patients had to be 18 years or older with confirmed AML according to WHO criteria and be considered ineligible for induction therapy on account of age (≥ 75 years) or significant cardiac, pulmonary, renal, hepatic, or other comorbidity. Patients aged 75 years and older had to have an ECOG PS of 0 to 2, and those aged 18 to 74 years could have an ECOG 0 to 3.

Patients were ineligible if they had acute promyelocytic leukemia, AML with favourable risk cytogenetics such as t(8;21), inv(16), t(16;16) or t(15;17) (as per NCCN Guidelines Version 2, 2016 for AML), AML with known active CNS involvement, a history of myeloproliferative neoplasm, or if they had previously received an HMA and/or any chemotherapeutic agent for MDS, or CAR T-cell therapy or any other experimental therapy.

Baseline Characteristics

Table 7 shows the baseline characteristics for the efficacy population. The mean age overall was 75.4 (SD = 5.95) years, and 60.6% of patients were aged 75 years or older. The majority were male (60.1%) and White (75.6%), followed by Asian (23.0%). Most had an ECOG PS of 1 (41.5%) or 2 (29.9%). The most common reasons patients were ineligible for standard induction therapy were because they were aged 75 years or older (56.8%) and had an ECOG PS of 2 or 3 (43.2%). Most patients (75.2%) had de novo AML; of those with secondary AML, 67.3% had prior MDS or chronic myelomonocytic leukemia. Cytogenetics were intermediate risk in 62.9% and poor in 37.1%. Of individual mutations, 23.9% of patients with known mutation status had mutations detected in IDH1 and/or IDH2, 16.2% had FLT3-ITD mutations and/or tyrosine kinase domain (TKD) mutations, 17.7% had nucleophosmin 1 (NPM1) mutations, and 20.9% had tumour protein p53 (TP53) mutations. Half (49.9%) had a bone marrow blast count of 50% or greater, 21.8% had a blast count between 30% and less than 50%, and 29.2% had a blast count of less than 30%. More than half (54.8%) had required a red blood cell or platelet transfusion within 8 weeks of first study treatment.

The demographic and baseline characteristics were well balanced. A greater proportion of patients receiving placebo plus azacitidine had mutations in FLT3 (20.4%) compared with those receiving venetoclax plus azacitidine (14.1%); conversely, a greater proportion of patients receiving venetoclax plus azacitidine had mutations in TP53 (23.3%) compared with placebo plus azacitidine (16.3%). Overall cytogenetic risk was a stratification variable and was well balanced.

Table 7: Demographic and Baseline Characteristics — Efficacy Population

Characteristic

VEN + AZA

(N = 286)

PBO + AZA

(N = 145)

Age (years)

Mean (SD)

75.6 (6.08)

75.1 (5.70)

Median

76

76

Minimum, maximum

49.0, 91.0

60.0, 90.0

Age category, n (%)

< 75 years

121 (42.3)

64(44.1)

≥ 75 years

165 (57.5)

81 (55.5)

Sex or gender, n (%)

Male

172 (60.1)

87 (60.0)

Female

114 (39.9)

59 (40.0)

Race, n (%)

White

217 (75.9)

109 (75.2)

Asian

66 (23.1)

33 (22.8)

Black or African American

3 (1.4)

2 (1.0)

American Indian or Alaska Native

0

1 (0.7)

Region, n (%)

US

50 (17.5)

24 (16.6)

EU

116 (40.6)

59 (40.7)

China

24 (8.4)

13 (9.0)

Japan

24 (8.4)

13 (9.0)

Rest of world

72 (25.5)

36 (24.8)

ECOG Performance Status, n (%)

0

37 (12.9)

23 (15.9)

1

120 (42.0)

58 (40.0)

2

113 (39.5)

59 (40.7)

3

16 (5.6)

5 (3.4)

Cytogenetic risk, n (%)

Intermediate

182 (63.6)

89 (61.4)

Poor

104 (36.4)

56 (38.6)

AML disease type, n (%)

Primary or de novo

214 (74.8)

110 (75.9)

Secondary (prior MDS and therapy-related AML), n (%)

72 (25.2)

35 (24.1)

Type of secondary AML, n (%, of those with secondary AML)

Therapy-related

26 (36.1)

9 (25.7)

Post MDS or CMML

46 (63.9)

26 (74.3)

Antecedent history of MDS, n (%)

Yes

49 (17.1)

27 (18.6)

No

237 (82.9)

118 (81.4)

RBC or platelet transfusion,b n (%)

Yes

155 (54.2)

81 (55.9)

No

131 (45.8)

64 (44.1)

RBC transfusion,b n (%)

Yes

144 (50.3)

76 (52.4)

No

142 (49.7)

69 (47.6)

Platelet transfusion,b n (%)

Yes

68 (23.8)

32 (22.1)

No

218 (76.2)

113 (77.9)

Bone marrow blast, n (%)

< 30%

85 (29.7)

41 (28.3)

≥ 30% to < 50%

61 (21.3)

33 (22.8)

≥ 50%

140 (49.0)

71 (49.0)

IDH1 or IDH2 mutation, nc,d (%)

IDH1

23 (9.4)

11 (8.7)

IDH2

40 (16.3)

18 (14.2)

IDH1 and/or IDH2

61 (24.9)

28 (22.0)

No mutation detected

184 (75.1)

99 (78.0)

Undetermined or missing

41

18

FLT3 mutation, nc,e (%)

ITD

23 (11.2)

13 (12.0)

TKD

7 (3.4)

10 (9.3)

ITD and/or TKD

29 (14.1)

22 (20.4)

Not detected

177 (85.9)

86 (79.6)

Undermined or missing

80

37

NPM1 mutation, nc (%)

Detected

27 (16.6)

17 (19.8)

Not detected

136 (83.4)

69 (80.2)

Undetermined or missing

123

59

TP53 mutation, nc (%)

Detected

38 (23.3)

14 (16.3)

Not detected

125 (76.7)

72 (83.7)

Undetermined or missing

123

59

Reasons for being ineligible for standard induction therapy,a n (%)

≥ 75 years of age

165 (57.7)

80 (55.2)

≥ 18 to 74 years of age

121 (42.3)

65 (44.8)

ECOG Performance Status of 2 or 3

95 (33.2)

50 (34.5)

History of congestive heart failure requiring treatment

2 (0.7)

3 (2.1)

Ejection fraction ≤ 50%

5 (1.7)

3 (2.1)

Chronic stable angina

5 (1.7)

1 (0.7)

DLCO ≤ 65%

11 (3.8)

12 (8.3)

FEV1 ≤ 65%

12 (4.2)

7 (4.8)

Creatinine clearance ≥ 30 mL/min to < 45 mL/min

11 (3.8)

5 (3.4)

Moderate hepatic impairment with total bilirubin > 1.5 to ≤ 3.0 × ULN

3 (1.0)

2 (1.4)

Other

12 (4.2)

6 (4.1)

CTC grade: Neutropenia

0

53 (18.5)

29 (20.0)

1

7 (2.4)

11 (7.6)

2

20 (7.0)

14 (9.7)

3

48 (16.7)

30 (20.8)

4

159 (55.4)

60 (41.4)

Missing

0

1

CTC grade: Anemia

0

2 (0.7)

2 (1.4)

1

39 (13.6)

17 (11.7)

2

157 (54.9)

74 (50.7)

3

88 (30.8)

52 (35.9)

CTC grade: Thrombocytopenia

0

36 (12.6)

19 (13.1)

1

61 (21.3)

28 (19.3)

2

44 (15.4)

25 (17.2)

3

78 (27.3)

42 (29.0)

4

67 (23.4)

31 (21.4)

AML = acute myeloid leukemia; AZA = azacitidine; CMML = chronic myelomonocytic leukemia; CTC = circulating tumour cell; DLCO = diffusing capacity of the lungs for carbon monoxide; ECOG = Eastern Cooperative Oncology Group; FEV1 = forced expiratory volume in 1 second; FLT3 = FMS-like tyrosine kinase 3; IDH = isocitrate dehydrogenase; ITD = internal tandem duplication; MDS = myelodysplastic syndrome; MRC = myelodysplasia-related changes; NPM1 = nucleophosmin 1; PBO = placebo; SD = standard deviation; TKD = tyrosine kinase domain; TP53 = tumour protein p53; ULN = upper limit of normal; VEN = venetoclax.

aPatients could have more than 1 reason for ineligibility.

bTransfusion within 8 weeks of start of PBO or VEN.

cPercentages exclude patients with undetermined or missing mutation status.

dA patient could have both the IDH1 and IDH2 mutations.

eA patient could have both FLT3-ITD and FLT3-TKD mutations.

Source: Clinical Study Report.1

Interventions

Venetoclax in the venetoclax plus azacitidine group was dosed at 400 mg oral per day after the initial dose titration of 100 mg at cycle 1 day 1, 200 mg at day 2, and 400 mg at day 3 and thereafter. Dosing was based on a phase Ib dose-escalation study of venetoclax in combination with HMAs (azacitidine and decitabine) in treatment-naive patients with AML who were aged 65 years or older and ineligible for the standard induction regime (Study M14 to 358).

Azacitidine in the venetoclax plus azacitidine and azacitidine groups was dosed at 75 mg/m2 subcutaneous or IV (depending on local practice) on days 1 to 7 of a 28-day cycle. The dose is as specified for the treatment of adult patients with AML in the EU Summary of Product Characteristics and for MDS in the US dose prescribing information.

Treatment was planned for a minimum of 6 cycles. Treatment could continue as long as the patient derived clinical benefit and did not have documented disease progression or develop unacceptable toxicity.

To allow for hematologic recovery from cytopenias, the dosing of venetoclax or placebo could be interrupted, the start of the next cycle delayed, or the duration reduced according to preplanned criteria. If the recovery did not reach a certain threshold, then the administered dose of azacitidine could be reduced by preplanned steps.

Protocol-specified dose adjustments were also made to adjust for drug interactions with systemic anti-fungal agents and other drugs that produced moderate or strong inhibition of CYP3A. Venetoclax or placebo was to be reduced at least twofold in patients receiving moderate CYP3A inhibitors and at least eightfold in those receiving strong CYP3A inhibitors. Moderate CYP3A inducers were excluded during the ramp-up phase and used with caution and after discussion with the sponsor.

All patients received prophylaxis against tumour lysis syndrome with oral and/or IV hydration and uric acid reducer and were admitted for monitoring during ramp-up of venetoclax plus placebo dosing. Patients with a white blood cell count greater than 25 × 109/L received cytoreduction before dosing.

Outcomes

Table 8 provides definitions of the efficacy end points that were assessed in the clinical trials included in this review and identified in the CADTH review protocol. Response measures (CR, CRi), partial remission, MLFS, resistant disease, and morphologic relapse were based on the revised guidelines for the International Working Group (IWG) for AML. Progressive disease was defined according to European LeukemiaNet recommendations. CRh is a derived end point based on bone marrow and hematological measurements. These end points are further summarized in Table 8. A detailed discussion and critical appraisal of the outcome measures is provided in Appendix 4.

Table 8: Summary of Outcomes of Interest Identified in the CADTH Review Protocol

Outcome

VIALE-A definition

OS

Number of days from date of randomization to the date of death.

EFS

Number of days from randomization to the date of progressive disease, relapse from CR or CRi, treatment failure (failure to achieve CR, CRi, or MLFS after at least 6 cycles of study treatment), or death from any cause.

CR

Absolute neutrophil counts > 109/L, platelets > 100 × 109/L, RBC transfusion independence, and bone marrow with < 5% blasts. Absence of circulating blasts and blasts with Auer rods; absence of extramedullary disease.

CRi

All criteria as CR except for residual neutropenia ≤ 109/L (1,000/μL), thrombocytopenia ≤ 100 × 109/L (100,000/μL), or RBC dependence.

CRh

Peripheral blood neutrophil count > 0.5 × 109/L, peripheral blood platelet count > 50 × 109/L, bone marrow < 5% blasts.

PR

All hematologic values for a CR but with a decrease of at least 50% in the percentage of blasts to 5% to 25% in the bone marrow aspirate.

MLFS

Less than 5% blasts in aspirate sample with marrow spicules and with a count of at least 200 nucleated cells. Absence of circulating blasts and extramedullary disease without peripheral blood cell recovery that meets thresholds for either CR or CRi.

RD

Failure to achieve CR, CRi, PR, or MLFS; only for patients surviving at least 7 days following completion of cycle 1 treatment with evidence of persistent leukemia by blood or bone marrow examination.

MR

Reappearance of ≥ 5% blasts after CR/CRi in peripheral blood or bone marrow or development of extramedullary disease.

PDa

50% increase in marrow blasts over baseline (minimum 15% increase required in cases with < 30% blasts at baseline); or persistent marrow blast percentage of > 70% over at least 3 months without at least a 100% improvement of ANC to an absolute level (> 0.5 × 109/L [500/μL], and/or platelet count to > 50 × 109/L [50,000/μL]); or

50% increase in peripheral blasts (WBC × % blasts) to > 25 × 109/L [25,000/μL]); or

New extramedullary disease.

DOR

The number of days from the date of first response (CR, CRi, or CRh) to the earliest evidence of confirmed MR, PD, or death due to disease progression.

Transfusion independence

≥ 56 days with no transfusion between the first dose of the study drug and the last dose of the study drug + 30 days. Applies to both RBC and platelets.

EORTC QLQ-C30

Consists of 30 items assessing quality of life in cancer patients. Includes 15 questions to assess HRQoL domains, including 5 multi-item functional scales (physical, emotional, cognitive, social, and role functioning), 3 multi-item symptom scales (fatigue, nausea and vomiting, pain), 6 single-item symptom scales (dyspnea, insomnia, appetite loss, constipation, diarrhea, and financial difficulties) and a global health status and quality of life scale (MCT = 10).

PROMIS 7a

Consists of 7 items assessing impact of fatigue over past 7 days in patients with cancer. Each response is on a 5-item scale ranging from 1 = never to 5 = always (MCT = 5).

EQ VAS

Visual analogue scale ranging from 100 (best imaginable health) to 0 (worst imaginable health) (MCT = 7).

ANC = absolute neutrophil count; CR = complete remission; CRh = complete remission with partial hematological recovery; CRi = complete remission with incomplete blood count recovery; EORTC QLQ-C30 = European Organization for Research and Treatment of Cancer Quality of Life Questionnaire; EQ-5D-5L VAS = EuroQol 5-Dimensions 5-Levels questionnaire Visual Analogue Scale; EFS = event-free survival; MCT = meaningful change threshold; MLFS = morphological leukemia-free state; MR = morphologic relapse; OS = overall survival; PD = progressive disease; PR = partial remission; PROMIS 7a = Patient-Reported Outcomes Measurement Information System Short Form v1.0–Fatigue 7a; RBC = red blood cell; RD = resistant disease; WBC = white blood cell.

aPR defined by European LeukemiaNet criteria.

Source: Clinical Study Report.1

EORTC QLQ-C30 (Version 3.0) is a cancer-specific measure of health-related quality of life (HRQoL).24 It comprises 30 individual questions organized into 5 functional scales (physical, role, cognitive, emotional, and social function), 3 symptom scales (fatigue, pain, and nausea and vomiting), 6 single-item symptom scales (dyspnea, insomnia, appetite loss, constipation, diarrhea, financial impact), and a global QoL scale (GHS/QoL). Function and symptoms are assessed over a 1-week recall period. Most questions have 4 response options (“not at all,” “a little,” “quite a bit,” “very much”), with scores on these items ranging from 1 to 4.25 For the 2 items that form the GHS/QoL scale, the response format is a 7-point Likert-type scale, with anchors between 1 (very poor) and 7 (excellent).25 Each raw scale score is converted to a standardized score ranging from 0 to 100, with a higher score reflecting better function on the function scales, higher symptoms on the symptom scales, and better QoL on the QoL scales.

Construct validity and internal consistency reliability were assessed by 2 separate studies using convenience samples of cancer patients in Singapore26 and Kenya,27 most of whom had breast or colon cancer. No relevant studies of responsiveness were found. Two studies estimated the minimal important difference (MID), 1 in patients with breast and small-cell lung cancer, using an anchor-based approach using change as measured by the subjective significance questionnaire,28 and the other in Canadian patients newly diagnosed with breast and colorectal cancer.29 Both estimated an MID of 10 points. A third study in Canadian patients30 used the EORTC (the GHS/QoL questions) to estimate MIDs for the overall EORTC QLQ C30 scales, but the method did not allow estimation of the MID for the GHS/QoL subscale itself.

The PROMIS 7a is a 7-item, patient-reported, tool that measures both the experience of fatigue and the interference of fatigue on daily activities over the past week. Responses are measured on a 5-point Likert scales from 1 = never to 5 = always, with total scores ranging from 7 to 35, with higher scores indicating greater fatigue.

There were no published validation studies of PROMIS 7a in cancer. Concurrent and discriminant validity were assessed in a mixed group of non-cancer patients and healthy controls. Known-groups validity was assessed by comparing PROMIS 7a scores in the clinical samples with healthy controls.31

The researchers who conducted the VIALE-A trial assessed the MID using anchor- and distribution-based approaches in a group of AML patients from the VIALE-C study.1 A 3-point difference that fell within the range of 3 to 5 proposed in the literature was considered an appropriate MID for patients with AML.

The EQ-5D-5L is a generic HRQoL instrument applicable to a wide range of health conditions.32,33 The first of 2 parts of the EuroQol 5-Dimensions questionnaire (EQ-5D) is a descriptive system that classifies respondents (aged ≥ 12 years) in 5 dimensions: mobility, self-care, usual activities, pain/discomfort and anxiety/depression. The EQ-5D-5L has 5 possible domains, representing “no problems,” “slight problems,” “moderate problems,” “severe problems” and “extreme problems.” Respondents are asked to choose the level that reflects their health state for each of the 5 dimensions, corresponding with 3,125 different health states. The second part is a visual analogue scale in which patients are asked to appraise their overall QoL on a scale of 0 to 100. There are 3 outputs: a 5-digit profile indicating the health states on the 5 dimensions, a population preference-weighted health index score based on the descriptive system, and a self-reported assessment of health status based on the EuroQol Visual Analogue Scale (EQ VAS).

Validity was assessed in 184 Canadian cancer patients with breast, colorectal, or lung cancer.34 The EQ-5D was able to discriminate between groups based on self-reported health status (excellent/good versus fair/very poor), somewhat based on ECOG PS (0 versus 1 to 3), but not for stage of cancer.34 Internal consistency reliability was calculated for the same study, and all 5 functioning scales along with GHS showed acceptable consistency (alpha > 0.70). Responsiveness was not reported. No relevant studies reported the MID.

Statistical Analysis

The analysis of the co-primary efficacy end point of CR + CRi was planned for 6 months after the first 225 patients were randomized. Three analyses were planned for the co-primary efficacy end point of OS:

There was a formal interim analysis for safety after approximately 20 patients received study medication and had been followed for at least 3 months. Subsequently, safety reviews were conducted every 3 months.

Interim data were reviewed by an IDMC that could make recommendations for the ongoing conduct of the trial. The trial design allowed for early regulatory submission in the EU following IA1, if desired. Following review of the results of IA, the IDMC recommended no modification and the trial continued without unblinding. Review of the results of IA2 led to an IDMC recommendation to stop the trial on March 16, 2020.

Table 9: Statistical Analysis of Efficacy End Points

End point

Statistical model

Stratification and adjustment factors

Sensitivity analyses

VIALE-A

OS

HR estimated from stratified Kaplan–Meier model

Stratified log-rank test

For IDH1/IHD2 and FLT3 subgroups, unstratified log-rank test

Stratified by age (18 to < 75 years, ≥ 75 years), cytogenetics (intermediate risk, poor risk)

All data in extracted database

Censoring of patients who received post-study treatment before experiencing event at start of post-study treatment

EFS

HR estimated from stratified Kaplan–Meier model

Stratified log-rank test

Stratified by age (18 to < 75 years, ≥ 75 years), cytogenetics (intermediate risk, poor risk)

Censoring of patients who received post-study treatment before experiencing event at start of post-study treatment

CR + CRi (investigator assessment)

Proportions with CR + CRi

Comparisons by OR

Comparisons by CMH stratified by age, cytogenetics

For IDH1/IHD2 and FLT3 subgroups, Fisher’s Exact Test

Stratified by age (18 to < 75 years, ≥ 75 years), cytogenetics (intermediate risk, poor risk)

CR + CRh (investigator assessment)

Proportions with CR + CRh

Comparisons by OR

Comparisons by CMH stratified by age, cytogenetics

For IDH1/IHD2 and FLT3 subgroups, Fisher’s Exact Test

Stratified by age (18 to < 75 years, ≥ 75 years), cytogenetics (intermediate risk, poor risk)

Duration of CR + CRi

HR estimated from stratified Kaplan–Meier model

Stratified by age (18 to < 75 years, ≥ 75 years), cytogenetics (intermediate risk, poor risk)

Censoring of patients who received post-study treatment before experiencing event at start of post-study treatment

Transfusion independence

Proportions with transfusion independence

Comparisons by stratified CMH

Stratified by age (18 to < 75 years, ≥ 75 years), cytogenetics (intermediate risk, poor risk)

Fatigue (PROMIS 7a)

Comparisons by linear mixed effects regression model fitted to longitudinal data with covariance structure

Exploratory TTD HR estimated from stratified Kaplan–Meier model (MCT = 5)

Models includes baseline score, stratification factors (age and cytogenetics), treatment arm, visit, and treatment arm by visit interaction

QoL (EORTC QLQ-C30, GHS/QoL)

Comparisons by linear mixed effects regression model fitted to longitudinal data with covariance structure

Exploratory TTD HR estimated from stratified Kaplan–Meier model (MCT = 10)

Models includes baseline score, stratification factors (age and cytogenetics), treatment arm, visit, and treatment arm by visit interaction

Cochrane regression models including treatment arm and prognostic variables (age, baseline ECOG store, AML type, cytogenetic risk, baseline PRO)

AML = acute myeloid leukemia; CMH = Cochran-Mantel-Haenszel; CR = complete remission; CRh = complete remission with incomplete hematological recovery; CRi = complete remission with incomplete blood count recovery; EFS = event-free survival; EORTC QLQ C30 = European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire Core 30; FLT3 = FMS-like tyrosine kinase 3; GHS/QoL = global health status quality of life scale; HR = hazard ratio; IDH = isocitrate dehydrogenase; MCT = meaningful change threshold; PROMIS 7a = Patient-Reported Outcomes Measurement Information System Short Form v1.0–Fatigue 7a; OS = overall survival; PRO = patient-reported outcome; TTD = time to deterioration.

Source: Clinical Study Report.1

Table 9 summarizes the methods used for statistical analysis of the efficacy end points. Time-to-event end points were analyzed with stratified Kaplan–Meier estimates and stratified log-rank comparisons. Proportions were compared by calculation of stratified ORs with statistical comparison by stratified Cochrane-Mantel-Haenszel tests. Stratification was by age and cytogenetic risk categories. Patient-reported outcomes were compared by linear mixed-effects regression models fitted to longitudinal data with covariance structure.

The sample size calculation assumed that patients would be randomized 2:1 to venetoclax plus azacitidine to placebo plus azacitidine. Other assumptions were:

Based on these assumptions, 225 patients (150 venetoclax plus azacitidine, 75 placebo plus azacitidine) would give 88% power to detect difference at a 2-sided alpha of 0.01, and 360 death events would have 86.7% power to detect a difference in OS with a 2-sided alpha of 0.04. Approximately 400 patients would be randomized: 267 to venetoclax plus azacitidine, and 133 to placebo plus azacitidine.

A hierarchical testing strategy was used for the control of multiplicity. The co-primary analysis of CR + CRi was performed on the first 225 patients (IA1). Two interim analyses of OS were planned, the first at IA1, with an administrative significance level of 0.0001. IA2 was performed after approximately 270 deaths (75% of predetermined deaths), using a Lan-DeMets alpha-spending function with O’Brien-Fleming boundary to control the 1-sided false-positive rate. Based on this analysis and pre-specified stopping rules, the IDMC made a recommendation to stop for success at IA2 rather than proceed to the final analysis. For the CR + CRi rate, CR + CRh rate, CR + CRi rate by the initiation of cycle 2, CR + CRh rate by the initiation of cycle 2, CR rate, and transfusion independence rate outcomes, the information fraction to be used at IA2 was calculated based on the proportion of the planned number of patients who had reached the desired amount of follow-up (e.g., for CR + CRi, at least 6 months of follow-up).

An overall 2-sided significance level of 0.05 was initially allocated between the co-primary end points: 0.01 to the analysis of CR + CRi and 0.04 to the analysis of OS. If the results of statistical testing for CR + CRi at IA1 were significant, then the 0.01 allocated to CR + CRi would be recycled to the OS analysis. If the results of statistical testing for OS were significant, then the fixed testing procedure would be performed at a 2-sided significance level of 0.05 for each of the selected secondary efficacy end points in sequence. If the results of statistical testing for OS were not significant, then statistical significance would not be declared for any of the end points.

Table 10 shows the actual alpha-spending boundary and information fraction for the end points tested under the hierarchical testing strategy, in order of testing.

Table 10: Actual Alpha-Spending Boundary and Information Fraction for End Points in Hierarchical Testing Strategy at IA2 (EU and EU Reference Countries)

End point

Information fraction

Interim boundary P value (2-sided)

Included in CADTH review

Primary

1. CR + CRi

First 226 patients

0.01

Yes

2. OS

75% (270 events)

0.02

Yes

Secondary

3. CR + CRi by cycle 2

100%

0.05

Yes

4. Post-baseline RBC transfusion independence

98%

0.047

Yes

5. CR + CRi rate IDH1/IDH2 subgroup

100%

0.05

Yes

6. CR rate

98%

0.047

Yes

7. CR + CRi rate FLT3 subgroup

100%

0.05

Yes

8. Post-baseline platelet transfusion independence

98%

0.047

Yes

9. EFS

87% (313 of 360 events)

0.032

Yes

10. CR + CRi MRD response rate

100%

0.05

No

11. OS in IDH1/2 subgroup

NA

0.0002

Yes

12. OS in FLT3 subgroup

NA

0.0002

Yes

13. EORTC QLQ-C30 GHS/QoL

NA

0.0002

Yes

14. PROMIS Short Form v1.0–Fatigue 7a

NA

0.0002

Yes

CR = complete remission; CRi = complete remission with incomplete blood count recovery; EFS = event-free survival; EORTC QLQ C30 = European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire Core 30; FLT3 = FMS-like tyrosine kinase 3; IA2 = second interim analysis; IDH = isocitrate dehydrogenase; GHS/QoL = global health status quality of life scale; MRD = minimal/measurable residual disease; NA = not applicable; OS = overall survival; PROMIS = Patient-Reported Outcomes Measurement Information System; RBC = red blood cell.

Source: Clinical Study Report.1

Drop-outs or missing data were handled using the following rules:

Analysis Populations

Two analysis populations were identified:

The longitudinal analysis population in the supplementary analysis of patient-reported outcomes included all patients in the efficacy population who survived up to a given time point and had available data for at least 1 patient-reported outcome measurement at baseline and at that time point.

Results

Patient Disposition

Table 11 shows the patient disposition. Of the 579 patients screened, 2 patients were randomized under the original protocol (group 1) and 431 patients were randomized under protocol amendment 1 and subsequent amendments (group 2). As the stratification variables changed under amendment 1, only patients in group 2 were included in the efficacy population. All eligible patients were included in the safety population.

A total of 579 patients was screened, with 146 excluded on screening, primarily because they did not meet the inclusion or exclusion criteria. Of the 431 patients randomized under amendment 1 and subsequent amendments, 286 were randomized to venetoclax plus azacitidine and 144 to placebo plus azacitidine. The primary reason for study discontinuation was death, in 56.7% of patients randomized to venetoclax plus azacitidine, and 75.2% of patients randomized to placebo plus azacitidine. Both loss to follow-up and withdrawal of consent were very low: less than 2% were lost to follow-up in both arms, and less than 2.5% withdrew consent. Three patients randomized to venetoclax plus azacitidine and 1 randomized to placebo plus azacitidine did not receive their assigned treatment and were excluded from the safety population.

Table 11: Patient Disposition for VIALE-A

Patient disposition

VIALE-A

VEN + AZA

PBO + AZA

Screened, N

579

Excluded on screening

146

Did not meet inclusion or exclusion criteria

98

Withdrew consent

21

Other

27

Randomized to both groups 1a and 2b; N (%)

287

146

Randomized and in group 2b; N (%)

286

145

Randomized, in group 2b; and received treatment, N (%)

283

144

Primary reason for study discontinuation, group 1 N (%)

Death

1 (100)

1 (100)

Primary reason for study discontinuation, group 2, N (%)

Death

161 (56.4)

109 (75.2)

Lost to follow-up

5 (1.7)

2 (1.4)

Withdrew consent

7 (2.4)

1 (0.7)

Efficacy population, N (group 2 only)

286

145

Safety population, N (all patients enrolled and treated)

283

144

PBO + AZA = placebo plus azacitidine; VEN + AZA = venetoclax plus azacitidine.

Note: Data cut-off was January 4, 2020.

aGroup 1 comprised the patients randomized under the original protocol.

bGroup 2 comprised the patients randomized under protocol amendment 1 and subsequent amendments.

Source: Clinical Study Report.1

Exposure to Study Treatments

Table 12 shows a summary of exposure for the safety population. Patients randomized to venetoclax plus azacitidine had longer exposure on average than those randomized to placebo plus azacitidine. The median duration of exposure for patients randomized to venetoclax plus azacitidine was 7.6 months (range, 0.0 to 30.7) and the median number of cycles was 7 (range, 1 to 30), compared with 4.3 months (range, 0.1 to 24.0) and 4.5 cycles (range, 1 to 26), respectively, for placebo plus azacitidine. A total of 106 patients (37.4%) who received venetoclax plus azacitidine and 42 (29.1%) patients who received placebo plus azacitidine received 10 or more cycles.

Table 12: Summary of Exposure to Treatment, Safety Population

Characteristic

VEN + AZA

N = 283

PBO + AZA

N = 144

Duration of exposure, months

Mean (SD)

9.9 (8.25)

6.7 (6.55)

Median

7.6

4.3

Minimum to maximum

0.0 to 30.7

0.1 –to 24.0

Duration interval, months

0 to 1

45 (15.9)

31 (21.5)

> 1 to 2

22 (7.8)

18 (12.5)

> 2 to 4

29 (10.2)

18 (12.5)

> 4 to 6

20 (7.1)

23 (16.0)

> 6 to 8

27 (9.5)

5 (3.5)

> 8 to 10

24 (8.5)

11 (7.6)

> 10 to 12 months

10 (3.5)

8 (5.6)

> 12

106 (37.5)

30 (20.8)

Number of cycles

Mean (SD)

8.8 (7.32)

6.9 (6.53)

Median

7

4.5

Minimum to maximum

1.0 to 30.0

1.0 to 26.0

Number of cycles, n

1

45 (15.9)

33 (22.9)

2

32 (11.3)

16 (11.1)

3

16 (5.7)

12 (8.3)

4

15 (5.3)

11 (7.6)

5

13 (4.6)

11 (7.6)

6

19 (6.7)

8 (5.6)

7

20 (7.1)

3 (2.1)

8

9 (3.2)

4 (2.8)

9

8 (2.8)

4 (2.8)

≥ 10

106 (37.4)

42 (29.1)

Dose reduction, n (%)

No reduction

202 (71.4)

112 (77.8)

1 reduction

61 (21.6)

26 (18.1)

2 reductions

13 (4.6)

3 (2.1)

≥ 2 reductions

7 (2.5)

3 (2.1)

Dose interruption, n (%)

No interruption

16 (5.7)

32 (22.2)

All reasons

267 (94.3)

112 (77.8)

1 interruption

50 (17.7)

34 (23.6)

2 interruptions

33 (11.7)

29 (20.1)

> 2 interruptions

184 (65.0)

49 (34.0)

Due to count recovery

187 (66.1)

29 (20.1)

1 interruption

28 (9.9)

15 (10.4)

2 interruptions

22 (7.8)

4 (2.8)

> 2 interruptions

137 (48.4)

10 (6.9)

Venetoclax or placebo dose interruption, n (%)

Due to count recovery

114 (52.8)

13 (27.7)

1 interruption

49 (22.7)

7 (14.9)

2 interruptions

32 (14.8)

5 (10.6)

> 2 interruptions

33 (15.3)

1 (2.1)

AZA dose interruption, n (%)

Due to count recovery

141 (65.3)

12 (25.5)

1 interruption

34 (15.7)

6 (12.8)

2 interruptions

21 (9.7)

3 (6.4)

> 2 interruptions

86 (39.8)

3 (6.4)

AZA = azacitidine; PBO = placebo; SD = standard deviation; VEN = venetoclax.

Note: Data cut-off was January 4, 2020.

Source: Clinical Study Report.1

More patients who received venetoclax plus azacitidine had a dose reduction and/or dose interruption than those who received placebo plus azacitidine, either drug in each combination, venetoclax or placebo alone, and azacitidine alone. Of the patients who received venetoclax plus azacitidine, 21.6% had 1 dose reduction, 4.6% had 2 dose reductions, and 2.5% had more than 2 dose reductions, compared with 18.1%, 2.1%, and 2.1%, respectively, for patients who received placebo plus azacitidine. Of the patients who received venetoclax plus azacitidine, 94.3% had at least 1 dose interruption for any reason and 66.1% had at least 1 dose interruption for count recovery, compared with 77.8% and 20.1%, respectively, for patients who received placebo plus azacitidine. Of the patients who received venetoclax plus azacitidine, 52.8% had a dose interruption of venetoclax or placebo and 65.3% had a dose interruption of azacitidine, compared with 27.7% and 25.5%, respectively, for patients who received placebo plus azacitidine.

Efficacy

Table 13 shows an overall summary of the efficacy outcomes and the subgroup analyses for the efficacy population at the time of the second interim analysis. Only those efficacy outcomes and analyses of subgroups identified in the review protocol are reported subsequently. Between-treatment differences that were statistically significant with controlled multiplicity (co-primary end points and sequential testing protocol) are indicated with a footnote.

At the time of the second interim analysis, the median duration of follow-up for patients randomized to venetoclax plus azacitidine was 20.7 months (95% CI, 20.1 to 22.0), and for those randomized to placebo plus azacitidine, it was 20.2 months (95% CI, 19.6 to 22.4).

Table 13: Summary of Efficacy Outcomes and Subgroup Analyses, Efficacy Population, IA2

Outcomes and analyses

VEN + AZA

N = 286

PBO + AZA

N = 145

OS

Events (deaths), n (%)

161 (56.3)

109 (75.2)

Median OS, months (95% CI)

14.7 (11.9 to 18.7)

9.6 (7.4 to 12.7)

HR (Cox proportional hazards model)a (95% CI)

0.662 (0.518 to 0.845)

P value (stratified log-rank test)a

< 0.001b

6-month OS estimate, % (95% CI)

71.9 (66.3 to 76.8)

63.9 (55.5 to 71.2)

12-month OS estimate, % (95% CI)

55.8 (49.7 to 61.5)

43.8 (35.5 to 51.8)

24-month OS estimate, % (95% CI)

36.5 (29.7 to 43.3)

18.3 (11.1 to 27.0)

Event-free survival

Number of patients with events, n (%)

191 (66.8)

122 (84.1)

Confirmed morphologic relapse or disease progression

83 (43.5)

35 (28.7)

Treatment failure

4 (2.1)

12 (9.8)

Death

104 (54.5)

75 (61.5)

Number of patients without an event

95 (33.2)

23 (15.9)

Treatment comparison

HR (Cox proportional hazards model)a (95% CI)

0.632 (0.502 to 0.796)

P value (stratified log-rank test)a

< 0.001b

Median duration of event-free survival (months; 95% CI)

9.8 (8.4 to 11.8)

7.0 (5.6 to 9.5)

No event rate at month 6, % (95% CI)

67.7 (61.8 to 72.8)

56.2 (47.6 to 63.9)

No event rate at month 12, % (95% CI)

43.5 (37.4, 49.3)

31.3 (23.6, 39.2)

No event rate at month 24, % (95% CI)

23.8 (17.9 to 30.2)

NA

Best response (CR + CRi) by investigator assessment

CR + CRi rate at IA1, n (%; 95% CI)c

Number of patients at IA1

147

79

CR

44 (29.9; 22.7 to 36.0)

12 (15.2; 8.1 to 25.0)

CRi

52 (35.4; 27.7 to 43.7)

8 (10.1; 4.5 to 19.0)

CR + CRi

96 (65.3; 57.0 to 73.0)

20 (25.3; 16.2 to 36.4)

P value (stratified CMH test)a

< 0.001

CR + CRi rate (as best response), n (%; 95% CI)c

CR

105 (36.7; 31.1 to 42.60)

26 (17.9; 12.1 to 25.2)

P value (stratified CMH test)a

< 0.001b

CRi

85 (29.7; 24.5 to 35.4)

15 (10.3; 5.9 to 16.5)

CR + CRi

190 (66.4; 60.6 to 71.9)

41 (28.3; 21.1 to 36.3)

Best IWG response, n (%)

CR

105 (36.7)

26 (17.9)

CRi

85 (29.7)

15 (10.3)

PR

3 (1.0)

3 (2.1)

MLFS

24 (8.4)

6 (4.1)

RD

36 (12.6)

69 (47.6)

MR

0

0

PD

3 (1.0)

6 (4.1)

Discontinued with no response data

30 (10.5)

20 (13.8)

No response data but still active

0

0

CR + CRi rate (as best response) by initiation of cycle 2, n (%; 95% CI)a

CR

37 (12.9; 9.3 to 17.4)

3 (2.1; 0.4 to 5.9)

CRi

87 (30.4; 25.1 to 36.1)

8 (5.5; 2.4 to 10.6)

CR + CRi

124 (43.4; 37.5 to 49.3)

11 (7.6; 3.8 to 13.2)

P value (stratified CMH test)a

< 0.001b

Time to response (CR + CRi) by investigator assessment

Time to first response, months, mean (SD) median (range)

CR + CRi

2.1 (1.82)
1.3 (0.6 to 9.9)

3.3 (2.61)
2.8 (0.8 to 13.2)

Time to best response, months, mean (SD) median (range)

CR

4.5 (4.38)
3.2 (0.9 to 24.5)

4.5 (2.95)
4.0 (1.0 to 13.2)

CRi

2.4 (2.03)
1.3 (0.6 to 8.8)

3.5 (2.77)
3.4 (0.8 to 11.2)

CR + CRi

3.6 (3.66)
2.3 (0.6 to 24.5)

4.2 (2.89)
3.7 (0.8 to 13.2)

Best response (CR + CRh) by investigator assessment

CR + CRh rate (as best response), n (%; 95% CI)a

CR

105 (36.7; 31.1 to 42.6)

26 (17.9; 12.1 to 25.2)

CRh

80 (28.0; 22.8 to 33.6)

7 (4.8; 2.0 to 9.7)

CR + CRh

185 (64.7; 58.8 to 70.2)

33 (22.8; 16.2 to 30.5)

Patients with best response to CR + CRh

CR + CRh rate (as best response) by initiation of cycle 2, n (%; 95% CI)a

CR

37 (12.9; 9.3 to 17.4)

3 (2.1; 0.4 to 5.9)

CRh

77 (26.9; 21.9 to 32.5)

5 (3.4; 1.1 to 7.9)

CR + CRh

114 (39.9; 34.1 to 45.8)

8 (5.5; 2.4 to 10.6)

Time to response (CR + CRh) by investigator assessment

Time to first response (months) mean (SD) median (range)

CR + CRh

2.2 (2.23)
1.0 (0.6 to 14.3)

3.0 (2.35)
2.6 (0.8 to 13.2)

Time to best response (months) mean (SD) median (range)

CR

4.5 (4.38)
3.2 (0.9 to 24.5)

4.5 (2.95)
4.0 (1.0 to 13.2)

CRh

2.6 (2.66)
1.0 (0.6 to 14.3)

2.7 (1.52)
2.8 (1.1 to 5.5)

CR + CRh

3.6 (3.84)
2.3 (0.6 to 24.5)

4.1 (2.79)
3.6 (1.0 to 13.2)

Duration of response (CR + CRi and CR) based on investigator assessment

CR + CRi

Number of patients with events, n/N (%)

84/190 (44.2)

23/41 (56.1)

DOR (months)a

Median (95% CI)

17.5 (13.6 to NE)

13.4 (5.8 to 15.5)

No event rate, month 6, % (95% CI)

80.6 (73.8 to 85.8)

65.6 (47.2 to 78.9)

No event rate, month 12, % (95% CI)

60.6 (52.6 to 67.7)

51.0 (32.3 to 66.9)

No event rate, month 18, % (95% CI)

48.0 (39.4 to 56.0)

20.4 (6.8 to 39.1)

CR

Number of patients with events, n/N (%)

39/105 (37.1)

13/26 (50.0)

DOR (months)a

Median (95% CI)

17.5 (15.3 to NE)

13.3 (8.5 to 17.6)

No event rate, month 6, % (95% CI)

83.3 (74.1 to 89.4)

84.4 (63.7 to 93.9)

No event rate, month 12, % (95% CI)

72.6 (62.1 to 80.6)

59.4 (33.2 to 78.2)

No event rate, month 18, % (95% CI)

47.5 (34.2 to 59.7)

13.0 (1.0 to 40.6)

Post-baseline transfusion independence

Post-baseline transfusion-independence rate

RBC and platelet, n (%; 95% CI)

166 (58.0; 52.1 to 63.8)

49 (33.8; 26.2 to 42.1)

Treatment difference, % (95% CI)

24.2 (14.7 to 33.8)

RBC

171 (59.8; 53.9 to 65.5)

51 (35.2; 27.4 to 43.5)

Treatment difference, % (95% CI)

24.6 (15.0 to 34.2)

P value (stratified CMH test)a

< 0.001b

Platelet

196 (68.5; 62.8 to 73.9)

72 (49.7; 41.3 to 58.1)

Treatment difference, % (95%)

18.9 (9.1 to 28.6)

P value (stratified CMH test)a

< 0.001b

Duration of post-baseline transfusion independence (days)

RBC and platelet

N

166

49

Mean (SD)

256.7 (204.49)

245.1 (182.48)

Median (minimum to maximum)

179.5 (57 to 33)

188 (56 to 727)

RBC

N

171

51

Mean (SD)

262.5 (202.35)

241.6 (181.94)

Median (minimum to maximum)

199 (57 to 933)

193 (56 to 727)

Platelet

N

196

72

Mean (SD)

282.8 (223.24)

276.7 (191.52)

Median (minimum to maximum)

210 (56 to 933)

227.5 (58 to 730)

Post-baseline transfusion-independence rate by baseline transfusion status, n/N (%; 95% CI)

RBC or platelet transfusion within 8 weeks before first dose of study drug

76/155 (49.0; 40.9 to 57.2)

22/81 (27.2; 17.9 to 38.2)

No transfusion within 8 weeks before first dose of study drug

90/131 (68.7; 60.0 to 76.5)

27/64 (42.2; 29.9 to 55.2)

Post-baseline RBC transfusion-independence rate by baseline transfusion status, n/N (%; 95% CI)

RBC transfusion within 8 weeks before first dose of study drug

71/144 (49.3; 40.9 to 57.8)

21/76 (27.6; 18.0 to 39.1)

No RBC transfusion within 8 weeks before first dose of study drug

100/142 (70.4; 62.2 to 77.8)

30/69 (43.5; 31.6 to 56.0)

Post-baseline platelet transfusion-independence rate by baseline transfusion status, n/N (%; 95% CI)

Platelet transfusion within 8 weeks before first dose of study drug

34/68 (50.0; 37.6 to 62.4)

12/32 (37.5; 21.1 to 56.3)

No platelet transfusion within 8 weeks before first dose of study drug

162/218 (74.3; 68.0 to 80.0)

60/113 (53.1; 43.5 to 62.5)

AML = acute myeloid leukemia; AZA = azacitidine; CI = confidence interval; CMH = Cochran-Mantel-Haenszel; CR = complete remission; CRh = complete remission with incomplete hematological recovery; CRi = complete remission with incomplete blood count recovery; DOR = duration of response; HR = hazard ratio; IA2 = second interim analysis; IWG = International Working Group; MLFS = morphologic leukemia-free state; MR = morphologic relapse; NA = not applicable; NE = not estimable; NPM1 = nucleophosmin 1; OS = overall survival; PBO = placebo; PD = progressive disease; PR = partial remission; RBC = red blood cell; RD = resistant disease; SD = standard deviation; VEN = venetoclax.

Note: Data cut-off was January 4, 2020.

aStratified by age (18 to < 75 years; ≥ 75 years) and cytogenetic risk (intermediate, poor risk).

bStatistically significant under preplanned testing strategy.

cCalculated from the exact binomial distribution.

Source: Clinical Study Report.1

Overall Survival

Table 13 summarizes the OS results for the efficacy population at IA2 (data cut-off: January 4, 2020) and Figure 2 shows the Kaplan–Meier survival curves.

Venetoclax plus azacitidine improved survival over placebo plus azacitidine (P value [stratified log-rank test] < 0.001; P value boundary = 0.02), with median survival in patients randomized to venetoclax plus azacitidine of 14.7 months (95% CI, 11.9 to 18.7) compared with 9.6 months (59% CI, 7.4 to 12.7) in patients randomized to placebo plus azacitidine (HR for mortality = 0.662; 95% CI, 0.518 to 0.845). The estimated survival at 12 months for venetoclax plus azacitidine was 55.8% (95% CI, 49.7% to 61.5%); for placebo plus azacitidine it was 43.8% (95% CI, 35.5% to 51.8%).

A sensitivity analysis that included all of the data in the extracted database showed similar results, with an HR for mortality of 0.653 (95% CI, 0.513 to 0.832), and an estimated survival rate at 12 months of 56% for venetoclax plus azacitidine (95% CI, 49.9% to 61.6%) and 43.9% for placebo plus azacitidine (95% CI, 35.6% to 51.9%).

Figure 2: Overall Survival, Efficacy Population, IA2

Depicts survival curves for venetoclax plus azacitidine and placebo plus azacitidine from 0 to 33 months of follow-up. Curves are smooth and diverge from around 1 month onward, with venetoclax plus azacitidine above and placebo plus azacitidine below. Probability of no event increases along the y-axis and time in months along the x-axis. Patients at risk in the 2 groups and survival estimates are also shown for the duration.

CI = confidence interval; COX PH = Cox proportional hazards model; IA2 = second interim analysis; PBO + AZA = placebo plus azacitidine; VEN + AZA = venetoclax plus azacitidine.

Note: Data cut-off was January 4, 2020.

Source: Clinical Study Report.1

Event-Free Survival and Duration of Event-Free Survival

Table 13 summarizes the results for EFS for the efficacy population at IA2 (data cut-off: January 4, 2020) and Figure 3 shows the Kaplan–Meier survival curves.

Venetoclax plus azacitidine improved EFS over placebo plus azacitidine (P value [stratified log-rank test] < 0.001; P value boundary of 0.032), with a median EFS duration of 9.8 months (95% CI, 8.4 to 11.8) for patients randomized to venetoclax plus azacitidine compared with 7.0 months (95% CI, 5.6 to 9.5) for patients randomized to placebo plus azacitidine (HR for EFS = 0.632; 95% CI, 0.502 to 0.796). At 12 months, 43.5% of the patients randomized to venetoclax plus azacitidine were free of confirmed morphologic relapse or disease progression, treatment failure, or death, compared with 31.3% of patients randomized to placebo plus azacitidine. A higher proportion of patients in the venetoclax plus azacitidine group (43.5%) had a confirmed morphologic relapse or disease progression event than in the placebo plus azacitidine group (28.7%); conversely, a higher proportion of patients had death as an event in the placebo plus azacitidine group (61.5%) than in the venetoclax plus azacitidine group (54.5%).

Figure 3: Event-Free Survival, Efficacy Population, IA2

Figure depicts survival curves for venetoclax plus azacitidine and placebo plus azacitidine from 0 to 33 months of follow-up. Curves are smooth and diverge immediately after start, with venetoclax plus azacitidine above and placebo plus azacitidine below. Probability of no event increases along the y-axis and time in months along the x-axis. Patients at risk in the 2 groups and survival estimates are also shown for the duration.

CI = confidence interval, COX PH = Cox proportional hazards model, IA2 = second interim analysis; PBO + AZA = placebo plus azacitidine, VEN + AZA = venetoclax plus azacitidine.

Note: Data cut-off was January 4, 2020.

Source: Clinical Study Report.1

Response: CR + CRi, CR, CR + CRh

Table 13 summarizes the results for composite complete remission (CR + CRi) for the efficacy population at the second interim assessment. The table also summarizes the results for CR + CRh, which has required minimum values for neutrophils and platelet counts.

CR + CRi at IA1 was a co-primary efficacy end point, as assessed for the first 226 patients (data cut-off: October 1, 2018). Venetoclax plus azacitidine improved CR + CRi (P value for stratified Cochran-Mantel-Haenszel [CMH] test < 0.001; P value boundary = 0.01) with 65.3% (95% CI, 57.0 to 73.0%) of patients meeting the end point compared with 25.3% (95% CI, 16.2% to 36.4%) of those who received placebo plus azacitidine.

Similar results were seen for conventional care regimen at IA2, with 66.4% (95% CI, 60.6 to 71.9%) of patients randomized to venetoclax plus azacitidine reaching the end point of CR + CRi compared with 28.3% (95% CI, 21.1% to 36.3%) of patients randomized to placebo plus azacitidine. This end point was not tested. Venetoclax plus azacitidine improved CR (P value [stratified CMH test] < 0.001; P value boundary = 0.047), with 36.7% (95% CI, 31.1% to 42.6%) of patients meeting the criteria for CR compared with 17.9% (95% CI, 12.1% to 25.2) of patients randomized to placebo plus azacitidine.

Among patients who had a partial response, 1.0% of those randomized to venetoclax plus azacitidine and 2.1% of those randomized to placebo plus azacitidine met the criteria for partial remission, while 8.4% of those randomized to venetoclax plus azacitidine and 4.1% of those randomized to placebo plus azacitidine met the criteria for MLFS.

Venetoclax plus azacitidine also improved early composite complete remission (P value [stratified CMH test] < 0.001; P value boundary = 0.05), with 43.4% (95% CI, 37.5% to 49.3%) of patients reaching the end point of CR + CRi by the beginning of cycle 2, compared with 7.6% (95% CI, 3.8 to 13.2) of patients randomized to placebo plus azacitidine. Results for time to first response (next section) were consistent.

Results for CR + CRh were consistent with those for composite complete remission, with 64.7% (95% CI, 58.8% to 70.2%) of patients randomized to venetoclax plus azacitidine reaching the end point of CR + CRh compared with 22.8% (95% CI, 16.2% to 30.5%) of patients randomized to placebo plus azacitidine, and similar results for time to first and time to best response.

Time to Response

Table 13 shows time to first and time to best response for CR + CRi and CR + CRh. Time to first response was 1.3 months (95% CI, 0.6 to 9.9) for patients randomized to venetoclax plus azacitidine and 2.8 months (95% CI, 0.8 to 13.2) for those randomized to placebo plus azacitidine. Median times to best CR or CRi were 2.3 (95% CI, 0.6 to 24.5) months and 3.7 (0.8 to 13.2) months for patients randomized to venetoclax plus azacitidine and placebo plus azacitidine, respectively. Very similar results were seen for CR + CRh.

Duration of Response

Table 13 shows the duration of response for CR + CRi and CR for the efficacy population at IA2 (data cut-off: January 4, 2020). The duration of response for patients who reached CR or CRi was 17.5 months (95% CI, 13.6 to not estimable) for patients randomized to venetoclax plus azacitidine and 13.4 (95% CI, 8.5 to 17.6) months for patients randomized to placebo plus azacitidine. At 12 months, 60.6% of responding patients randomized to venetoclax plus azacitidine still met the response criteria compared with 51.0% of responding patients randomized to placebo plus azacitidine.

Post-Baseline Transfusion Independence

Table 13 summarizes independence and duration of independence from red blood cell and platelet transfusions and from both combined for the efficacy population at IA2 (January 4, 2020). The table also shows results for patients who were and were not transfusion-independent at baseline.

Venetoclax plus azacitidine improved post-baseline transfusion independence for red blood cells (P value [stratified CMH test] < 0.001; P value boundary = 0.047). For red blood cells, 59.8% (95% CI, 53.9% to 65.5%) of patients randomized to venetoclax plus azacitidine were transfusion-independent compared with 35.2% (95% CI, 27.4% to 43.5%) of patients randomized to placebo plus azacitidine, a treatment difference of 24.6% (95% CI, 15.0 to 34.2). The median duration of red blood cells transfusion independence was 199 days (range of 57 to 933 days) for patients randomized to venetoclax plus azacitidine and 193 days (range of 56 to 727 days) for patients randomized to placebo plus azacitidine. For both groups, a greater proportion of patients who were transfusion-independent at baseline were transfusion-independent post baseline compared with patients who were not transfusion-dependent at baseline.

Venetoclax plus azacitidine improved post-baseline transfusion independence for platelets (P value [stratified CMH test] < 0.001; P value boundary = 0.047). For platelets, 68.5% (95% CI, 62.8% to 73.9%) of patients randomized to venetoclax plus azacitidine were transfusion-independent compared with 49.7% (95% CI, 41.3% to 58.1%), a treatment difference of 18.9% (95% CI, 9.1% to 28.6%). The median duration of platelet transfusion independence was 210 days (range of 56 to 933 days) for patients randomized to venetoclax plus azacitidine, and 227.5 days (range of 58 to 730 days) for patients randomized to placebo plus azacitidine. For both groups, a greater proportion of patients who were transfusion-independent at baseline were still transfusion-independent post baseline compared with patients who were not transfusion-independent at baseline.

Hospitalization

Overall rates of hospitalization were not reported. A higher percentage of patients who received venetoclax plus azacitidine experienced adverse events leading to hospitalization than those who received placebo plus azacitidine (80.6% versus 66.7%). The most common adverse events leading to hospitalization involved cytopenias or were infectious: febrile neutropenia (29.3% for venetoclax plus azacitidine versus 10.4% for placebo plus azacitidine), pneumonia (14.8% versus 20.1%), anemia (4.9% versus 4.2%), neutropenia (4.6% versus 2.1%), sepsis (4.6% versus 6.9%), and thrombocytopenia (3.9% versus 0.7%).

Overall Survival and CR + CRi in Subgroups

Table 38 (Appendix 3) shows the results for the comparison of the molecular subgroups in the efficacy population, as of IA2, for OS and CR + CRi. Data were available for the subgroups of age (age < 75 years, age ≥ 75 years), ECOG PS (ECOG < 2, ECOG ≥ 2), cytogenetic risk (intermediate versus poor), de novo versus secondary AML, AML with and without myelodysplastic syndrome, blast count at baseline (< 30%, 30% to < 50%, and 50%), and mutations (IDH1 and/or IDH2, FLT3, NPM1, and TP53).

OS in patients with IDH1 and/or IDH2 and in patients with FLT3, and CR + CRi with IDH1 and/or IDH2 and in patients with FLT3 were included as end points in the sequential testing strategy.

Venetoclax plus azacitidine improved OS in patients with an IDH1 and/or IDH2 mutation compared with placebo plus azacitidine (P value unstratified log-rank test < 0.0001; P value boundary = 0.0002). Median survival for patients randomized to venetoclax plus azacitidine was not estimable, compared with 6.2 months (95% CI, 2.3 to 12.7) for patients randomized to placebo plus azacitidine (HR = 0.345; 95% CI, 0.199 to 0.598).

No statistically significant difference was identified for OS in patients with FLT3 mutation between patients randomized to venetoclax plus azacitidine and those randomized to placebo plus azacitidine (P value [unstratified log-rank test] < 0.2054; P value boundary = 0.0002). Median survival for patients randomized to venetoclax plus azacitidine was 12.7 months (95% CI, 7.3 to 23.5) compared with 8.9 months (95% CI, 5.9 to 14.7) for patients randomized to placebo plus azacitidine (HR = 0.664; 95% CI, 0.351 to 1.257).

Venetoclax plus azacitidine improved CR + CRi in patients with IDH1 and/or IDH2 mutation compared with placebo plus azacitidine (P value [Fisher’s exact test] < 0.001; P value boundary = 0.05). The composite remission rate for patients randomized to venetoclax plus azacitidine was 75.4% (95% CI, 52.9% to 87.3%), compared with 10.7% (95% CI, 2.3% to 28.2%) for patients randomized to placebo plus azacitidine.

Venetoclax plus azacitidine improved CR + CRi in patients with FLT3 mutation, compared with placebo plus azacitidine (P value [Fisher’s exact test] < 0.021; P value = 0.05). Median survival for patients randomized to venetoclax plus azacitidine was 72.4% (95% CI, 52.8% and 87.3%) compared with 36.4% (95% CI, 17.2% to 59.3%) for patients randomized to placebo plus azacitidine.

For the other subgroups, the greatest difference in point estimates for OS and CR + CRi was observed for age and cytogenetic risk. In patients aged less than 75 years, the HR for OS for the comparison of venetoclax plus azacitidine to placebo plus azacitidine was 0.888 (95% CI, 0.591 to 1.33), compared with an HR of 0.535 (95% CI, 0.394 to 0.727) in patients aged 75 years and older. In patients aged less than 75 years, the risk difference in CR + CRi for the comparison of venetoclax plus azacitidine to placebo plus azacitidine was 21.12% (95% CI, 5.6% to 36.6%), compared with 49.43% (95% CI, 38.6% to 60.2%) in patients aged 75 years and older.

In patients with intermediate cytogenetic risk, HR for OS for the comparison of venetoclax plus azacitidine versus placebo plus azacitidine was 0.566 (95% CI, 0.407 to 0.786), compared with an HR of 0.775 (95% CI, 0.538 to 1.117) in patients with poor cytogenetic risk. In patients with intermediate risk, the risk difference in CR + CRi for the comparison of venetoclax plus azacitidine versus placebo plus azacitidine was 42.72% (95% CI, 31.2% to 54.3%), compared with 29.67% (95% CI, 15.0% to 44.3%) in patients with poor cytogenetic risk.

Differences in point estimates were minimal for the other identified subgroups.

Patient-Reported Outcomes

Overall QoL was captured by the EORTC QLQ-C30 GHS/QoL and health utility was captured by the EQ-5D. Fatigue was reported in the PROMIS 7a scale and the fatigue subscale in the EORTC QLQ-C30. Nausea and vomiting, pain, dyspnea, insomnia, appetite loss, constipation, and diarrhea were all reported as part of the EORTC.

The EORTC QLQ-30 GHS/QoL and PROMIS 7a were secondary end points and included in the statistical testing strategy. EQ-5D was reported as an exploratory end point. Exploratory analyses were conducted of the time to deterioration of individual scores and subgroups.

EORTC QLQ-C30 GHS/QoL

Table 38 shows the mean scores at baseline and day 1 of subsequent treatment cycles, the least squares (LS) mean change from baseline, and the LS mean difference between venetoclax plus azacitidine and placebo plus azacitidine, for EORTC QLQ-C30 GHS/QoL. Data are shown up to cycle 13, after which the attrition of patients due to death and discontinuation due to disease progression had reduced the study cohorts to a small number.

Figure 4 shows the time to deterioration on the EORTC QLQ-C30 GHS/QoL. At baseline, compliance in responding to the questionnaire was 92.9% and 90.9% in the venetoclax plus azacitidine and placebo plus azacitidine groups, respectively. In subsequent cycles (up to cycle 13), compliance in individual treatment groups ranged from 72.4% to 83.9%.

Figure 4: Time to Deterioration on the EORTC QLQ-C30 GHS/QoL, Longitudinal Analysis Population

The figure depicts survival curves for venetoclax plus azacitidine and placebo plus azacitidine from 0 to 30 months of follow-up. Curves diverge from start, touch around month 2, and continue to diverge thereafter, with venetoclax plus azacitidine above and placebo plus azacitidine below. There is a discontinuity in slope at 2 months, where both curves steepen. Proportion stable/improved increases along the y-axis and time in months along the x-axis, up to 36 months. Patients at risk in the 2 groups and survival estimates are also shown for the duration.

AZA = azacitidine; CI = confidence interval; GHS/QoL = global health status quality of life scale; HRQoL = health-related quality of life; LAP = longitudinal analysis population; EORTC QLQ-30C = European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire Core 30; PBO = placebo; PRO = patient-reported outcome; VEN = venetoclax.

Source: Patient-reported outcome report.1

The mean EORTC QLQ-C30 GHS/QoL at baseline was similar in patients in the venetoclax plus azacitidine group and placebo plus azacitidine group: 52.61 (n = 262) and 55.96 (n = 130), respectively. There was a greater change from baseline in the venetoclax plus azacitidine group than in the placebo plus azacitidine group at all points except cycle 19. The difference met or exceeded the MID of 5 points at cycles 5 and 21. There were no clinically meaningful differences in mean change from baseline between treatment groups. This end point was part of the statistical testing hierarchy, but testing failed for an end point before this. Thus, conclusions cannot be drawn based on the results of this outcome without risk of increased type I error.

The median time to deterioration for patients randomized to venetoclax plus azacitidine was 16.5 months, compared with 9.3 months for patients randomized to placebo plus azacitidine. The adjusted HR was 0.81 (95% CI, 0.553 to 1.183). The meaningful change threshold was 10 points.

PROMIS 7a

Fatigue was assessed using the patient-reported PROMIS 7a scale, which assessed the experience and impact of fatigue over the previous 7 days. Table 38 shows the mean scores at baseline and on day 1 of subsequent treatment cycles, the LS mean change from baseline, and the LS mean difference between venetoclax plus azacitidine and placebo plus azacitidine, for PROMIS 7a. Figure 5 shows the time to deterioration on the EORTC QLQ-C30 GHS/QoL. Values run from 0 to 100, with higher values indicating greater fatigue. At baseline, compliance in completing the PROMIS 7a was 93.6% and 92.3% in the venetoclax plus azacitidine and the placebo plus azacitidine groups, respectively. In subsequent cycles (up to cycle 13), compliance in individual treatment groups ranged from 72.4% to 84.9%.

Figure 5: Time to Deterioration on the PROMIS Fatigue 7a, Longitudinal Analysis Population

The figure depicts survival curves for venetoclax plus azacitidine and placebo plus azacitidine from 0 to 30 months of follow-up. Curves diverge at start, touch around 2 months, and diverge slightly thereafter, with venetoclax plus azacitidine above and placebo plus azacitidine below. There is a discontinuity in slope at 2 months, where both curves steepen. Proportion stable/improved increases along the y-axis and time in months along the x-axis, up to 36 months. Patients at risk in the 2 groups and survival estimates are also shown for the duration.

AZA = azacitidine; CI = confidence interval; GHS/QoL = global health status quality of life scale; HRQoL = health-related quality of life; LAP = longitudinal analysis population; EORTC QLQ-30C = European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire Core 30; PBO = placebo; PRO = patient-reported outcome; PROMIS Fatigue 7a = Patient-Reported Outcomes Measurement Information System Short Form v1.0–Fatigue 7a; VEN = venetoclax.

Source: Patient-reported outcome report.1

The mean PROMIS 7a score at baseline was similar among patients in the venetoclax plus azacitidine group and the placebo plus azacitidine group: 53.86 (n = 264) and 54.97 (n = 132), respectively. There was a greater change from baseline in PROMIS 7a scores among patients in the venetoclax plus azacitidine group versus the placebo plus azacitidine group on day 1 of cycles 5, 7, 9, 11, and 13, but no clinically meaningful differences in mean change from baseline between treatment groups. This end point was part of the statistical testing hierarchy, but testing failed for an end point before this. Thus, conclusions cannot be drawn based on the results of this outcome without risk of increased type I error.

The median time to deterioration for patients randomized to venetoclax plus azacitidine was 9.3 months compared with 8.6 months for patients randomized to placebo plus azacitidine. The adjusted HR was 0.72 (95% CI, 0.509 to 1.011). The meaningful change threshold was 5 points.

EORTC QLQ-C30 Symptom Scales

The mean baseline EORTC QLQ-C30 fatigue subscale score for venetoclax plus azacitidine was 47.67 (n = 262) and for placebo plus azacitidine it was 49.83 (n = 130). Change from baseline showed improvement in subsequent cycles for both treatments, but the observed differences in LS means between the treatments were small at each time point. At baseline, compliance in responding to the questionnaire was 92.2% and 90.9% in the venetoclax plus azacitidine and the placebo plus azacitidine groups, respectively. In subsequent cycles (up to cycle 13), compliance in individual treatment groups ranged from 72.4% to 82.9%.

The mean baseline EORTC QLQ-30 nausea and vomiting subscale score for venetoclax plus azacitidine was 7.95 (n = 262) and for placebo plus azacitidine it was 7.63 (n = 130). Change from baseline showed improvement in subsequent cycles for both treatments, but the observed differences in LS means between the treatments were small at each time point.

The mean baseline EORTC QLQ-30 pain subscale score for venetoclax plus azacitidine was 21.95 (n = 262) and for placebo plus azacitidine it was 25.90 (n = 130). There was a numerically greater improvement for venetoclax plus azacitidine at most time points, but the observed differences in LS means between the treatments were small at each time point.

The mean baseline EORTC QLQ-30 appetite loss subscale score for venetoclax plus azacitidine was 31.42 (n = 262) and for placebo plus azacitidine it was 30.77 (n = 130). There was a greater improvement for venetoclax plus azacitidine from cycle 5 to cycle 13, but the observed differences in LS means between the treatments were small at each time point.

The mean baseline EORTC QLQ-30 constipation subscale score for venetoclax plus azacitidine was 20.23 (n = 262) and for placebo plus azacitidine it was 15.8 (n = 130). There was a greater improvement in subsequent cycles for venetoclax plus azacitidine, but the observed differences in LS means between the treatments were small at each time point.

The mean baseline EORTC QLQ-30 diarrhea subscale score for venetoclax plus azacitidine was 10.56 (n = 262) and for placebo plus azacitidine it was 9.74 (n = 130). There was improvement in subsequent cycles for venetoclax plus azacitidine for both treatments, but the observed differences in LS means between the treatments were small at each time point.

EuroQol Visual Analogue Scale

Table 38 shows the mean scores at baseline and day 1 of subsequent treatment cycles, the LS mean change from baseline, and the LS mean difference between venetoclax plus azacitidine and placebo plus azacitidine, for the EQ VAS. Figure 6 shows the time to deterioration on the EQ VAS.

The mean baseline EQ-5D-5L index score for venetoclax plus azacitidine was 0.76 (n = 260) and for placebo plus azacitidine it was 0.74 (n = 130). There was improvement (increase) in subsequent cycles for venetoclax plus azacitidine for both treatments, but the observed differences in LS means between the treatments were small at each time point. The mean baseline EQ VAS for venetoclax plus azacitidine was 60.29 (n = 260) and for placebo plus azacitidine it was 64.27 (n = 130). There was improvement (increase) in subsequent cycles for venetoclax plus azacitidine for both treatments, but the observed differences in LS means between the treatments were small at each time point.

The median time to deterioration for patients randomized to venetoclax plus azacitidine was 10.7 months, compared with 3.9 months for patients randomized to placebo plus azacitidine. The adjusted HR was 0.55 (95% CI, 0.394 to 0.768). The meaningful change threshold was 7 points.

Figure 6: Time to Deterioration on the EQ VAS, Longitudinal Analysis Population

The figure depicts survival curves for venetoclax plus azacitidine and placebo plus azacitidine from 0 to 30 months of follow-up. Curves diverge at start, touch around 2 months, and diverge slightly thereafter, with venetoclax plus azacitidine above and placebo plus azacitidine below. There is a discontinuity in slope at 2 months, where both curves steepen. Proportion stable or improved increases along the y-axis and time in months along the x-axis, up to 36 months. Patients at risk in the 2 groups and survival estimates are also shown for the duration.

AZA = azacitidine; LAP = longitudinal analysis population; EQ-5D = EuroQol 5-Dimensions questionnaire; EQ VAS = EuroQol Visual Analogue Scale; PBO = placebo; PRO = patient-reported outcome; VEN = venetoclax.

Source: PRO report.1

Harms

Only those harms identified in the review protocol are reported subsequently. Table 14 shows an overall summary of the treatment-emergent adverse events, most common treatment-emergent adverse events, grade 3 or greater treatment-emergent adverse events, SAEs, adverse events leading to death, adverse events leading to discontinuation of venetoclax or placebo and azacitidine, and notable harms (January 4, 2020). Individual adverse events are in descending order of frequency for patients receiving venetoclax plus azacitidine.

All patients in both groups experienced at least 1 adverse event, and almost all experienced at least 1 grade 3 or greater adverse event. Compared with patients who received placebo plus azacitidine, a greater proportion of patients who received venetoclax plus azacitidine experienced 1 or more SAEs, 1 or more adverse events leading discontinuation or dose interruption for venetoclax or placebo or azacitidine, or 1 or more adverse events leading to death. Of the patients who received venetoclax plus azacitidine, 279 (98.6%) had grade 3 or greater adverse events, 235 (83.0%) had SAEs, 69 (24.4%) had adverse events leading to discontinuation of venetoclax or placebo, 68 (24.0%) had adverse events leading to discontinuation of azacitidine, and 64 (22.6%) had adverse events leading to death. Of the patients who received placebo plus azacitidine, 139 (96.5%) had grade 3 or greater adverse events, 105 (72.9%) had SAEs, 29 (20.1%) had adverse events leading to discontinuation of venetoclax or placebo, 29 (20.1%) had adverse events leading to discontinuation of azacitidine, and 29 (20.1%) had adverse events leading to death.

Table 14: Treatment-Emergent Adverse Events, Serious Adverse Events, and Adverse Events Leading to Discontinuation, Safety Population, IA2

Adverse events

VEN + AZA (N = 283)

n (%)

PBO + AZA (N = 144)

n (%)

Patients with any AE

283 (100)

144 (100)

Patients with AE grade ≥ 3

279 (98.6)

139 (96.5)

Patients with any SAE

235 (83.0)

105 (72.9)

Patients with VEN- or PBO-related AEa

241 (85.2)

96 (66.7)

Patients with AZA-related AEa

246 (86.9)

108 (75.0)

Patients with any AE leading to VEN or PBO discontinuation

69 (24.4)

29 (20.1)

Patients with any AE leading to AZA discontinuation

68 (24.0)

29 (20.1)

Patients with any AE leading to VEN or PBO dose interruption or reduction

204 (72.1)

84 (58.3)

Patients with any AE leading to AZA dose interruption or reduction

190 (67.1)

67 (46.5)

Patients with any AE leading to death

64 (22.6)

29 (20.1)

Treatment-emergent adverse events reported in ≥ 10% of patients in either arm

Thrombocytopenia

130 (45.9)

58 (40.3)

Nausea

124 (43.8)

50 (34.7)

Constipation

121 (42.8)

56 (38.9)

Neutropenia

119 (42.0)

42 (29.2)

Febrile neutropenia

118 (41.7)

27 (18.8)

Diarrhea

117 (41.3)

48 (33.3)

Vomiting

84 (29.7)

33 (22.9)

Hypokalemia

81 (28.6)

41 (28.5)

Anemia

78 (27.6)

30 (20.8)

Decreased appetite

72 (25.4)

25 (17.4)

Edema peripheral

69 (24.4)

26 (18.1)

Pyrexia

66 (23.3)

32 (22.2)

Pneumonia

65 (23.0)

39 (27.1)

Fatigue

59 (20.8)

24 (16.7)

Leukopenia

58 (20.5)

20 (13.9)

Asthenia

44 (15.5)

12 (8.3)

Dizziness

37 (13.1)

10 (6.9)

Dyspnea

37 (13.1)

11 (7.6)

Weight decreased

37 (13.1)

14 (9.7)

Cough

35 (12.5)

20 (13.9)

Hypophosphatemia

35 (12.4)

17 (11.8)

Insomnia

35 (12.4)

15 (10.4)

Stomatitis

33 (11.7)

8 (5.6)

Arthralgia

33 (11.7)

7 (4.9)

Abdominal pain

31 (11.0)

12 (8.3)

Headache

30 (10.6)

10 (6.9)

Treatment-emergent adverse events of grade ≥ 3 reported for ≥ 5% of the patients in either arm

Thrombocytopenia

126 (44.5)

55 (38.2)

Neutropenia

119 (42.0)

41 (28.5)

Febrile neutropenia

118 (41.7)

27 (18.8)

Anemia

74 (26.1)

29 (20.1)

Leukopenia

58 (20.5)

17 (11.8)

Pneumonia

56 (19.8)

36 (25.0)

Hypokalemia

30 (10.6)

15 (10.4)

Hypophosphatemia

21 (7.4)

11 (7.6)

Atrial fibrillation

17 (6.0)

3 (2.1)

Sepsis

17 (6.0)

13 (9.0)

Hypertension

17 (6.0)

6 (4.2)

Urinary tract infection

11 (3.9)

8 (5.6)

Serious adverse events reported in ≥ 2% of the patients in either arm

Febrile neutropenia

84 (29.7)

15 (10.4)

Pneumonia

47 (16.6)

32 (22.2)

Sepsis

16 (5.7)

12 (8.3)

Anemia

14 (4.9)

6 (4.2)

Neutropenia

13 (4.9)

3 (2.1)

Thrombocytopenia

12 (4.2)

2 (1.4)

Atrial fibrillation

13 (4.6)

2 (1.4)

Escherichia sepsis

8 (2.8)

2 (1.4)

Influenza

8 (2.8)

2 (1.4)

Lung infection

8 (2.8)

3 (2.1)

Pyrexia

7 (2.5)

3 (2.1)

Septic shock

7 (2.5)

1 (0.7)

Urinary tract infection

7 (2.5)

3 (2.1)

Diarrhea

6 (2.1)

2 (1.4)

Acute kidney injury

5 (1.8)

5 (3.5)

Respiratory failure

5 (1.8)

1 (0.7)

General physical health deterioration

3 (1.1)

4 (2.8)

Malignant neoplasm progression

2 (0.7)

5 (3.5)

Acute myocardial infarction

2 (0.7)

3 (2.1)

Pleural effusion

2 (0.7)

3 (2.1)

Fall

1 (0.4)

3 (2.1)

AEs leading to death for > 1 patient

Pneumonia

11 (3.9)

3 (2.1)

Sepsis

6 (2.1)

5 (3.5)

Death

4 (1.4)

2 (1.4)

Cardiac arrest

3 (1.1)

2 (1.4)

Hemorrhage intracranial

3 (1.1)

0

Respiratory failure

3 (1.1)

1 (0.7)

Septic shock

3 (1.1)

1 (0.7)

Atrial fibrillation

2 (0.7)

0

Multiple organ dysfunction syndrome

2 (0.7)

1 (0.7)

Systemic inflammatory response syndrome

2 (0.7)

1 (0.7)

General physical health deterioration

1 (0.4)

1 (0.7)

Klebsiella infection

1 (0.4)

1 (0.7)

Cerebral hemorrhage

1 (0.4)

1 (0.7)

AEs leading to PBO/VEN discontinuation for ≥ 2 patients in either group

Acute kidney injury

4 (1.4)

2 (1.4)

Atrial fibrillation

4 (1.4)

0

Febrile neutropenia

4 (1.4)

1 (0.7)

Neutropenia

4 (1.4)

2 (1.4)

Pneumonia

4 (1.4)

4 (2.8)

Sepsis

4 (1.4)

5 (3.5)

Thrombocytopenia

3 (1.1)

3 (2.1)

Malignant neoplasm progression

3 (1.1)

3 (2.1)

Respiratory failure

3 (1.1)

1 (0.7)

Cardiac failure

2 (0.7)

0

Death

2 (0.7)

0

Systemic inflammatory response syndrome

2 (0.7)

1 (0.7)

Fatigue

2 (0.7)

1 (0.7)

Klebsiella infection

2 (0.7)

0

Septic shock

2 (0.7)

0

AEs leading to AZA discontinuation for ≥ 2 patients in either group

Acute kidney injury

4 (1.4)

2 (1.4)

Atrial fibrillation

4 (1.4)

0

Neutropenia

4 (1.4)

1 (0.7)

Pneumonia

4 (1.4)

4 (2.8)

Sepsis

4 (1.4)

5 (3.5)

Febrile neutropenia

3 (1.1)

1 (0.7)

Malignant neoplasm progression

3 (1.1)

3 (2.1)

Thrombocytopenia

3 (1.1)

3 (2.1)

Cardiac failure

2 (0.7)

0

Death

2 (0.7)

0

Systemic inflammatory response syndrome

2 (0.7)

1 (0.7)

Fatigue

2 (0.7)

1 (0.7)

Klebsiella infection

2 (0.7)

0

Septic shock

2 (0.7)

0

Summary of notable harms for VEN + AZA and PBO + AZA

Neutropenia searchb

201 (71.0)

64 (44.4)

Febrile neutropenia

118 (41.7)

27 (18.8)

Infections and infestations, all

239 (84.5)

97 (67.4)

Most commonc

Pneumonia

65 (23.0)

39 (27.1)

Upper respiratory tract infection

26 (9.2)

13 (9.0)

Urinary tract infection

26 (9.2)

11 (7.6)

Lung infection

19 (6.7)

4 (2.8)

Sepsis

18 (6.4)

13 (9.0)

Oral herpes

17 (6.0)

6 (4.2)

Cellulitis

16 (5.7)

8 (5.6)

Oral candidiasis

16 (5.7)

5 (3.5)

Bronchitis

15 (5.3)

4 (2.8)

Hemorrhage

107 (37.8)

53 (36.8)

Tumour lysis syndromed

Met Howard criteria

7 (2.5)

3 (2.1)

Reported AE of tumour lysis syndrome

3 (1.1)

0

Secondary primary malignancy

11 (3.9)

2 (0.7)

AE = adverse event; AZA = azacitidine; IA2 = second interim analysis; PBO = placebo; VEN = venetoclax.

aReported as “any reasonable possibility” of being a treatment-related AE, as assessed by investigator.

bIncludes terms: neutropenia, neutrophil count decreased, febrile neutropenia, agranulocytosis, neutropenic infection, and neutropenic sepsis.

cIndividual AEs reported for ≥ 5% of patients in either group.

dEvaluated between the first dose of the study drug and 7 days after the first dose of the study drug.

Note: Data cut-off was January 4, 2020.

Source: Clinical Study Report.1

Treatment-Emergent Adverse Events

Table 14 shows treatment-emergent adverse events that were reported in 10% or more of patients and treatment-emergent adverse events reported in 5% or more of patients in the safety population at the time of IA2 (January 4, 2020). The most common adverse events were thrombocytopenia, nausea, constipation, neutropenia, febrile neutropenia, diarrhea, vomiting, hypokalemia, and anemia. The most common grade 3 or greater treatment-emergent adverse events were thrombocytopenia, neutropenia, febrile neutropenia, anemia, leukopenia, pneumonia, and hypokalemia.

The treatment-emergent adverse events reported with a frequency of 5% or greater in patients receiving venetoclax plus azacitidine compared with those receiving placebo plus azacitidine were thrombocytopenia, neutropenia, febrile neutropenia, anemia, leukopenia, nausea, diarrhea, vomiting, stomatitis, hemorrhoids, peripheral edema, asthenia; decreased appetite, arthralgia; dizziness, syncope, presyncope, vertigo; dyspnea; pruritus; and rash maculopapular.

Serious Adverse Events

Table 14 shows SAEs reported in 2% or more patients in the safety population at IA2 (January 4, 2020). The most common SAEs were febrile neutropenia, pneumonia, sepsis, anemia, and neutropenia. Febrile neutropenia, anemia, neutropenia, and thrombocytopenia were more frequent in patients who received venetoclax plus azacitidine, and pneumonia and sepsis were more frequent in patients who received placebo plus azacitidine.

Mortality

Table 14 shows adverse events leading to death for the safety population. A total of 64 (22.6%) patients who received venetoclax plus azacitidine and 29 (20.1%) who received placebo plus azacitidine had adverse events leading to death. The most frequent adverse events leading to death were pneumonia, sepsis, cardiac arrest, and an event recorded as death. Pneumonia, death, and cardiac arrest occurred more frequently in patients who received venetoclax plus azacitidine.

Treatment Discontinuations Due to Adverse Events

Table 14 shows adverse events leading to discontinuation of placebo or venetoclax for 2 or more patients in either group in the safety population by adverse event and system organ class.

The most frequent adverse events leading to venetoclax or placebo treatment discontinuation in the venetoclax plus azacitidine group were acute kidney injury, atrial fibrillation, febrile neutropenia, neutropenia, pneumonia, sepsis, thrombocytopenia, malignant neoplasm progression, and respiratory failure. In patients who received venetoclax plus azacitidine, atrial fibrillation, febrile neutropenia, cardiac failure, Klebsiella infection, and septic shock were more frequent, and in patients who received placebo plus azacitidine, pneumonia, sepsis, thrombocytopenia, and malignant neoplasm progression were more frequent.

The most frequent adverse events leading to azacitidine treatment discontinuation in the venetoclax plus azacitidine group were acute kidney injury, atrial fibrillation, neutropenia, pneumonia, sepsis, febrile neutropenia, and malignant neoplasm progression. In patients who received venetoclax plus azacitidine, atrial fibrillation, neutropenia, febrile neutropenia, cardiac failure, Klebsiella infection, and septic shock were more frequent, and in patients who received placebo plus azacitidine, pneumonia, sepsis, malignant neoplasm progression, and thrombocytopenia were more frequent.

Notable Harms

The notable harms identified for the protocol were neutropenia, febrile neutropenia, infections, tumour lysis syndrome, hemorrhage, and secondary malignancies. The search for adverse events of neutropenia included terms for neutropenia, neutrophil count decreased, febrile neutropenia, agranulocytosis, neutropenic infection, and neutropenic sepsis. Tumour lysis syndrome was a known risk and all patients received prophylaxis with oral and/or IV hydration and uric acid reducer, and were admitted for monitoring during ramp-up of venetoclax. Patients with a white blood cell count greater than 25 × 109/L required cytoreduction before treatment.

Neutropenia, febrile neutropenia, infections and infestations, and secondary primary malignancies all occurred in a greater proportion of patients who received venetoclax plus azacitidine than in patients who received placebo plus azacitidine. Hemorrhage and tumour lysis syndrome occurred in similar proportions, and the proportion of patients with tumour lysis syndrome was low (≤ 2.5%).

The most common secondary malignancies were basal cell carcinoma and squamous cell carcinoma of the skin: 3 (1.1%) and 2 (0.7%) instances, respectively, in patients receiving venetoclax plus azacitidine, and none in patients receiving placebo plus azacitidine. Other malignancies reported for patients who received venetoclax plus azacitidine were gastric adenocarcinoma (recurrence), adenocarcinoma of colon, chloroma (manifestation of AML), erythroleukemia (AML), neuroendocrine carcinoma of the skin, and plasma cell myeloma. Other malignancies reported for patients receiving placebo plus azacitidine were malignant melanoma and renal cancer.

Critical Appraisal

Internal Validity

Randomization was conducted by an independent statistician, allocation was through an interactive voice recognition system (IVRS), and the study was double blinded, with identical-appearing oral venetoclax and placebo. Venetoclax does not appear to have any adverse events that are so specific as to unblind patients or physicians; therefore, the blinding was likely to remain intact. Blinding was not assessed.

There was no clinically meaningful imbalance in the baseline characteristics that might favour 1 group or the other, and there was minimal loss to follow-up (1.7% for venetoclax plus azacitidine and 1.4% for placebo plus azacitidine). The number of patients who withdrew consent after randomization was small (approximately 2%).

The duration of exposure and number of treatment cycles received was longer in the venetoclax plus azacitidine group, which is probably reflective of the difference in survival: patients receiving venetoclax plus azacitidine survived longer and received more cycles. A similar proportion of patients discontinued treatment due to adverse events. Procedures for assessing compliance with treatment (i.e., counting of returned doses) were described in the protocol but the results were not reported.

OS is a standard outcome in oncology drug investigation, with robust methods for ascertainment. Collection was likely to be complete and the timing of events was likely to be accurately determined. Standard methods for survival analysis were used, with surviving patients censored at the date they were known to be alive on or before the cut-off date. There was minimal loss to follow-up or withdrawal and good balance at baseline, so censoring is unlikely to be related to prognosis. The prognosis of the patients recruited is unlikely to have changed with time, as there were no changes to the inclusion and exclusion criteria that would be likely to affect prognosis, and recruitment took place over a relatively short time period.

EFS is a composite end point consisting of death from any cause, confirmed morphologic relapse from CR + CRi, confirmed disease progression, and treatment failure. Treatment failure was defined as failure to reach CR, CRi, or MLFS after at least 6 cycles. No protocol-specific support for the validity of the end point was offered in the protocol or statistical analysis plan. EFS is an accepted end point in the development of treatments for leukemia,25 although empirical data show inconsistent correlation between EFS and OS.35 However, it provides a more direct measurement of the ability of the treatment to achieve a response and the durability of the response achieved than OS, since EFS is affected by trial treatment alone, while OS is affected by trial treatment, post-trial treatment, and supportive or palliative care.35 A time-to-event analysis of all individual end points making up the composite was not reported, making it difficult to fully assess for violations of the assumptions underlying the composite end points (i.e., the events were of equal importance to patients, occur with similar frequency, and have a similar sensitivity to the treatment). Death was reported for the greatest proportion of patients (54.5% and 61.5% for venetoclax plus azacitidine and placebo plus azacitidine, respectively), followed by confirmed morphologic relapse or confirmed disease progression (43.5% and 28.7% for venetoclax plus azacitidine and placebo plus azacitidine, respectively), and then by treatment failure (2.1% and 9.8% for venetoclax plus azacitidine and placebo plus azacitidine, respectively). The proportion of patients with each end point was reported, and the distribution for each treatment was consistent with the observed results for survival and CR + CRi, which were higher in the venetoclax plus azacitidine group than in the placebo plus azacitidine group. Results for individual analyses of OS, duration of response, and CR + CRi show similar directions of effect, but this does not adjust for competing events. Standard methods for survival analysis were used, with surviving patients censored at the date they were known to be alive on or before the cut-off date. There was minimal loss to follow-up or to withdrawal and a good balance at baseline, so censoring is unlikely to be related to prognosis. The prognosis of the patients recruited is unlikely to have changed with time, as there were no changes to the inclusion and exclusion criteria that would be likely to affect prognosis, and recruitment took place over a relatively short time period.

Composite complete remission (CR + CRi) and CR were investigator-assessed based on laboratory and clinical findings, with independent review. No protocol-specific support for the validity of the end point was offered in the protocol or statistical analysis plan. CR + CRi is an accepted end point in the development of treatments for leukemia,25 although empirical data suggest the strength of the correlation between CR + CRi and OS may be population- and treatment-dependent.35 The results were cross tabulated, and the differences minimal between investigator and independent review results. Treatment effect was not calculated for the end point and its 2 components, and statistical testing for CR + CRi and CR was performed at different interim analyses. CR and CRi both reflect bone marrow and peripheral blood improvement, with different thresholds, and the direction of effect was the same for both. Randomized patients without a post-baseline disease assessment were considered nonresponders. This is a conservative assumption, biasing the individual estimates of response downward, but accounts for a competing risk of death in an aged population.

Transfusion-independence rate was a pre-specified end point that was included in the sequential testing strategy. It is not clear how data on transfusion were collected, or whether these data might be susceptible to survivor bias, i.e., whether patients had to survive until the next transfusion visit for the previous visit to be captured. This risks undercounting transfusions in seriously ill patients. Patients who did not receive the study drug were considered transfusion-dependent, a conservative assumption affecting only a small number of patients.

Overall QoL was measured using the EOTRC QLQ-C30 GHS/QoL scale, cancer-related fatigue was measured using the PROMIS 7a, fatigue and other symptoms of interest were measured using EORTC QLQ-30C subscales, and health utility was measured using the EQ-5D-5L. The EOTRC QLQ-C30 GHS/QoL scale had previously been validated in mixed groups of cancer patients and an MID established. The PROMIS 7a has been validated in published studies of chronic illness, and the sponsor reported steps to validate the measure using the data from the VIALE-C study of venetoclax plus LDAC. Mean change from baseline was calculated using available data without imputation, and the level of compliance with the tool was not reported. Compliance post baseline was around 80%, meaning about 20% of available patients were not represented, and attrition due to death and disease progression was pronounced, meaning that later time points in particular represent a small survivor subgroup.

There were 2 preplanned interim analyses with preplanned stopping boundaries, with recommendations to stop or proceed made by an independent monitoring committee. Multiplicity due to interim analyses and the testing of multiple end points was controlled by a preplanned alpha-spending strategy and pre-specified hierarchy of testing with gatekeeping. Testing boundaries for selected end points representing proportions were adjusted by an information fraction to accommodate incomplete accrual at the interim analysis. Planned methods were reported on and adhered to.

Protocol violations included violations of inclusion and exclusion criteria, where patients were included although their bone marrow blast count did not meet the threshold at the time of testing for the study or the data were missing. A review provided evidence that they had previously met the criteria. Protocol violations in dosing included the use of strong and moderate CYP3A inhibitors without modifying the dose of venetoclax or placebo. These cases were reviewed for safety concerns. No violations were reported that substantially affected the internal validity of the study.

Subgroups of interest were pre-specified. Age (18 to < 75 years and ≥ 75 years) and cytogenetic risk (intermediate, poor) were used as stratification variables; other subgroups were not stratified. The aseline balance of all covariates was not reported for subgroups. Some subgroups were small, which was reflected in the high uncertainty of treatment estimates. This particularly affected estimates of CR + CRi, where both the number of specific events and total number of events could be small. Subgroup analyses of patients with IHD1, IHD2, and FLT3 mutations were included in the sequential testing strategy. Other subgroup analyses, including those on patients negative for mutations, were not adjusted for multiplicity.

There were 7 protocol amendments with corresponding changes in the statistical analysis plan. An early change to stratification factors meant that 2 patients randomized under the original protocol were not included in the efficacy analysis. Minor changes were made to inclusion criteria for safety reasons, there were clarifications on the dosing of concomitant medications, and the sample size was adjusted to allow for longer follow-up. Changes were made to the definition of EFS to align it with VIALE-C and to CR and CRi. All changes were made before the stopping and unblinding of the study following IA2 and are unlikely to have affected the internal validity of the study.

External Validity

The inclusion criteria for VIALE-A assumed that patients aged 75 years and older would not be eligible for standard induction chemotherapy, and age was the most common reason given for randomized patients being considered ineligible for standard induction chemotherapy. In Canadian practice, there is no consistency in defining an upper age limit for intensive chemotherapy. Chemotherapy may be considered for patients with treatment-naive AML aged 75 years and older, especially those with good or intermediate risk cytogenetics. The myelosuppressive nature of venetoclax plus azacitidine means it may not be suitable for frail patients or those who cannot travel for frequent lab visits, regardless of age.

Patients with CNS involvement were excluded, but clinical experts indicated that patients with CNS involvement might benefit from venetoclax plus azacitidine with concomitant intrathecal therapy. Patients with secondary AML (arising from prior myeloproliferative neoplasm including myelofibrosis, essential thrombocythemia, and polycythemia vera) were excluded from enrolling onto the study. However, there is data that shows activity of venetoclax plus azacitidine in this group of patients.36,37 Patients with isolated granulocytic sarcoma were not included in the study.

Venetoclax dosing in the trial was aligned with the Health Canada–approved dosing. Up-titration and monitoring would be expected to be the same in clinical practice. Dosing of azacitidine was also aligned with Health Canada–indicated dosing; however, the current Health Canada approval for azacitidine is for patients with AML with less than 30% blasts. In practice, experts and clinician groups noted that jurisdictions are funding azacitidine and centres are already using it in patients with a blast count of 30% or greater. There is no maximum blast restriction in the Health Canada–approved indication for venetoclax plus azacitidine for patients with AML who are aged 75 years and older or ineligible for standard induction therapy, and the indication aligns with the submission. In the study, azacitidine was dosed for 7 consecutive days while, in practice, alternative dosing regimens are used to reduce the hematological toxicity, e.g., 5 to 2-2 and 6 consecutive days.

The outcome measures were relevant to patients and clinicians. They captured clinically important end points such as OS, clinically important surrogate end points of response and remission, overall QoL, and factors identified as influencing QoL (e.g., transfusion dependence). Disease remission, avoidance of relapse, symptoms, QoL, independence from transfusion and avoidance of hospitalization, and OS were all identified as important to patients and physicians. In clinical practice, strict responder definitions may not capture responding patients; patients may not reach strict response categories but may still derive clinical benefit.

The settings for the study were predominately urban hospitals and clinics. It therefore does not necessarily address the rural or remote Canadian context, where patients would not have access to frequent laboratory testing to monitor the ramp-up of venetoclax and cytopenias, nor access to outpatient or inpatient treatment for side effects and complications. Patients would be required to travel for treatment or to receive an alternative.

Duration of follow-up was around 20 months, with a median OS for venetoclax plus azacitidine of 14.7 months. Five-year follow-up is standard in oncology trials.

Indirect Evidence

Objectives and Methods for the Summary of Indirect Evidence

An ITC was required because of a lack of studies directly comparing venetoclax plus azacitidine and venetoclax plus LDAC with other treatments currently in use in the Canadian setting.

Search Methods

A focused literature search for NMAs dealing with Venclexta (venetoclax) and AML was run in MEDLINE All (1946–) on February 11, 2021. No limits were applied.

Description of Indirect Treatment Comparison

One report that included ITCs was supplied by the sponsor. It included a systematic review with an NMA comparing venetoclax plus azacitidine and venetoclax plus LDAC with azacitidine, LDAC and BSC, and 2 propensity-score analyses comparing venetoclax plus azacitidine with LDAC (2-way comparison), and venetoclax plus azacitidine with azacitidine with LDAC (3-way comparison).

Table 15 shows the study selection criteria and key aspects of the methods for the systematic review. The patient population of interest included treatment-naive adult patients with AML who were ineligible for intensive chemotherapy, but the search allowed flexible wording to ensure retrieval of studies. Treatment naive was considered interchangeable with “previously untreated” or “newly diagnosed,” and “ineligible for chemotherapy” included patients described as old or elderly, unfit for intensive chemotherapy, unfit for standard chemotherapy, or unfit for high-dose chemotherapy. The initial search for articles included a broader set of comparators and included controlled clinical trials as a study design. More restricted selection criteria that were developed for a planned EUnetHTA submission were applied at the full-text review stage; the table reflects these criteria. The reasons for selection of comparators were not given, but the overall declared intention was to select high-quality studies that might enable ITCs.

Table 15: Study Selection Criteria and Methods for the Systematic Review

Criteria

ITC

Population

Treatment-naive adult patients (age ≥ 18 years) with AML who were ineligible for intensive chemotherapy:

  • Patients who had not received any prior treatment for AML with the exception of hydroxyurea (allowed through the first cycle of treatment). Prior treatment for MDS was allowed, except for cytarabine.

  • Patients with secondary AML with or without prior treatment with an HMA for MDS were included.

Studies were excluded if they were not on humans; not on adults; not on treatment-naive AML; specifically recruited patients with HIV, HBV, or HCV infection; or included patients with APL.

Intervention or comparator

Studies with at least 1 of the following regimens:

  • venetoclax + azacitidine

  • venetoclax + low-dose cytarabine

  • venetoclax + decitabine

  • azacitidine

  • low-dose cytarabine

  • decitabine

  • glasdegib + low-dose cytarabine

  • best supportive care, including blood transfusion, etoposide, mercaptopurine, or hydroxyurea

Outcome

Studies reporting at least 1 of the following outcomes:

  • overall survival

  • event-free survival

  • progression-free survival

  • relapse-free survival

  • complete remission (CR)

  • CR with incomplete blood count recovery (CRi)

  • composite complete remission (CR + CRi)

  • CR with partial hematologic recovery (CRh)

  • objective response

  • partial remission

  • duration of remission

  • minimal/measurable residual disease

  • grade 3 or 4 adverse events

  • discontinuation due to adverse events

Study design

Included designs:

  • RCTs

Other selection criteria

• Inclusion restricted to English-language studies

• Inclusion limited to studies with ≥ 20 patients per arm

• Exclusion of studies with mixed MDS and AML populations, unless outcomes were reported for the AML subgroup

• Bibliographies of systematic reviews and meta-analyses identified in the search were screened for studies before exclusion

Databases searched

Searched through Ovid:

  • MEDLINE and Epub Ahead-of-Print, In-Process and Other Non-Indexed Citations, Daily and Versions

  • EMBASE

  • Cochrane Central Register of Controlled Trials (CENTRAL)

  • Cochrane Database of Systematic Reviews

  • Database of Abstracts of Reviews of Effects

Abstract search (2017 onward) through Ovid Northern Light Life Sciences Conference Abstracts (http://www.ovid.com/site/catalog/databases/13207.jsp) or through the conference website if the latest conference abstracts were not indexed in Northern Light database:

Also searched:

• Validated filters (Scottish Intercollegiate Guidelines Network) were used to retrieve RCTs

Selection process

Level 1 screening was by title and abstract. Potentially relevant studies were passed on to level 2, where the full text was screened. Each level of screening was conducted by 2 independent reviewers. Discrepancies were reconciled by a third reviewer.

Data extraction process

Data were extracted independently by 2 reviewers into a predefined extraction table. Discrepancies were reconciled by a third reviewer.

Quality assessment

The quality assessment was done according to the Centre for Reviews and Dissemination Risk of Bias Assessment checklist for RCTs:

  • Was the method used to generate random allocations adequate?

  • Was the concealment of treatment allocation adequate?

  • Were the groups similar at the outset of the study in terms of prognostic factors, for example, severity of disease?

  • Were the care providers, participants, and outcome assessors blind to treatment allocation? If any of these people were not blinded, what might be the likely impact on the risk of bias (for each outcome)?

  • Were there any unexpected imbalances in drop-outs between groups? If so, were they explained or adjusted for?

  • Were there any evidence to suggest that the authors measured more outcomes than they reported?

  • Did the analysis include an intention-to-treat analysis? If so, was this appropriate and were appropriate methods used to account for missing data?

AML = acute myeloid leukemia; APL = acute promyelocytic leukemia; CR = complete remission; CRh = complete remission with incomplete hematological recovery; CRi = complete remission with incomplete bone marrow recovery; HBV = hepatitis B virus; HCV = hepatitis C virus; HMA = hypomethylating agent; ITC = indirect treatment comparison; MDS = myelodysplastic syndrome; RCT = randomized controlled trial.

Source: Systematic review report.38

Methods of the ITC

Objectives

The objective of this study was to compare the efficacy of venetoclax combination therapies with alternative treatments in treatment-naive patients with AML who were ineligible for intensive chemotherapy, including:

Study Selection Methods

To be included in the NMAs, trials retrieved by the systematic review had to meet the following criteria:

The decision to restrict selection to phase III RCTs for reasons of quality led to the exclusion of trials containing glasdegib, as there was no phase III trial connected to the network containing venetoclax plus azacitidine and venetoclax plus LDAC.

ITC Analysis Methods

Three analyses were conducted: 1 NMA and 2 propensity score–weighted comparisons.

The NMA compared venetoclax plus azacitidine and venetoclax plus LDAC with comparators for the available end points of OS and CR + CRi. The feasibility of pooling to create a network for analysis was pre-assessed on the basis of study and patient characteristics. The main analysis excluded patients from the VIALE-C LDAC group who would not have been eligible to enter VIALE-A because they had previously been treated with an HMA or had good cytogenetic risk. For OS, the proportional hazards assumption was assessed using log-log cumulative hazard plots, which led to the decision to model OS using proportional hazards.

The model was a Bayesian mixed-treatment comparison using a generalized linear model framework, with OS modelled using the identity link and dichotomous outcomes modelled using the logit link. Due to limited data, only fixed-effects models were estimated. Prior distributions were non-informative and selected according to a process that was not detailed. Posterior probabilities were modelled using Markov chain Monte Carlo methods, with 50,000 iterations on 3 chains and a burn-in period of 50,000 iterations. Convergence was assessed using trace and density plots and Gelman-Rubin plots and diagnostics. Selection between models was made by the difference information criterion (DIC). The chosen definition of a meaningful difference in DIC was not given.

Two propensity score–weighting analyses were conducted. No specific rationale was provided for these additional analyses. The comparisons were:

For both analyses, the propensity score for treatment was calculated for each patient using a logistic regression model with treatment with venetoclax plus azacitidine versus LDAC as the outcome and the baseline demographic and clinical characteristics as covariates. Analyses of OS, EFS, and CR + CRi were then conducted using data weighted by the inverse of the probability score. The covariates were:

For comparability, the propensity score–weighting analyses excluded patients from the VIALE-C LDAC group who would not have been eligible to enter VIALE-A: those who had been previously treated with an HMA and those who had good cytogenetic risk. For the 2-way propensity-score analysis, a subgroup analysis was conducted that was restricted to those patients in the main analysis who had greater than 30% bone marrow blasts at the baseline assessment, with a corresponding sensitivity analysis of all patients who had 30% bone marrow blasts. For the 3-way propensity-score analysis, a planned subgroup analysis of patients with 20% to 30% bone marrow blasts was not conducted, as the number of patients meeting these criteria in the LDAC arm was small.

Standard mean differences, t-tests (continuous variables), and chi-square tests (categorical variables) were used to assess balance before weighting, and weighted standard mean differences, weighted t-tests, and weighted chi-square tests were used to assess balance after weighting. An effective sample size was calculated and distributions of weights inspected to identify potential sensitivity to extreme weights. Time-to-event comparisons (OS and EFS) were made using weighted Cox proportional hazards models. Standard errors, 95% CIs, and P values were based on robust estimates of variances accounting for variability in propensity-score weights.

Results of ITC

Summary of Included Studies

Following removal of duplicates, 7,319 records were screened by title and abstract; of these, 225 were screened in full text. With the addition of the VIALE-A and VIALE-C study reports, the final selection was 7 RCTs with at least 2 arms of interest.

With the additional restriction of the comparators for the NMA inclusion criteria, removing decitabine from the comparators, 4 trials were included in the NMA: VIALE-A, VIALE-C, AZA-001, and AZA-AML-001. Table 16 shows a summary of the study characteristics for these 4 trials.

Table 16: Study Characteristics of Trials Included in the Systematic Review

Study

Design

N

Intervention vs. comparator

Key inclusion criteria

Key exclusion criteria

VIALE-A (M15-656)

Phase III, double-blind, RCT

Randomized 2:1, VEN + AZA:PBO + AZA

VEN + AZA: 286

PBO + AZA: 145

VEN + AZA vs. PBO + AZA

VEN 400 mg orally once a day (1 to 28 days)

AZA 75 mg/m2 SC or IV daily (1 to 7 days)

Aged ≥ 18 years, with AML, ineligible for standard induction due to age or comorbidities

Treatment-naive

ECOG:

  • aged 75 years: 0 to 2

  • aged 18 to 74 years: 0 to 3

Prior treatment for AML, except hydroxyurea; prior HMA or VEN chemotherapy for MDS; prior CAR T-cell therapy; received strong or moderate CYP3A inducers within 7 days

Prior myeloproliferative neoplasm, acute promyelocytic leukemia, active CNS involvement

Cytogenetic risk: Good

VAILE-C (M16-043)

Phase III, double-blind

Randomized 2:1, VEN + AZA:PBO + AZA

VEN + LDAC: 143

LDAC: 68

VEN + LDAC vs. LDAC

VEN 600 mg orally once a day (1 to 28 days)

LDAC 20 mg/m2 SC (1 to 10 days)

≥ 18 years, with AML, ineligible for intensive induction therapy, (aged ≥ 75 years, or ≥ 18 to 74 years and met at least 1 of criteria for lack of fitness for intensive induction therapy)

Treated for MDS (except cytarabine)

ECOG 75 years: 0 to 2

ECOG 18 to 74 years: 0 to 3

Prior treatment for AML, except hydroxyurea

Prior myeloproliferative neoplasm, acute PML, active CNS involvement

AZA-001

Phase III, open-label

AZA: 55

LDAC: 20

BSC: 27

AZA vs. LDAC AZA vs. BSC

AZA 75 mg/m2 SC daily (1 to 7 days)

BSC (blood product infusion, antibiotics, GSF)

LDAC 20 mg/m2 SC (1 to 14 days)

AML patients ≥ 20% bone marrow or peripheral blasts

Therapy-related disease

AZA-AML-001

Phase III, open-label

AZA: 241

LDAC: 158

BSC: 45

AZA vs. BSC

AZA vs. LDAC

AZA 75 mg/m2 SC daily (1 to 7 days)

BSC (blood product infusion, antibiotics, GSF)

LDAC 20 mg/m2 SC (1 to 10 days)

Aged ≥ 65 years, newly diagnosed AML, > 30% blasts

Intermediate or poor risk cytogenetics

Acute AML with t(15;17)(q22;q12) and AML with inv(16)(p13.1q22) or t(16;16)(p13.1;q22), t(8;21)(q22;q22), or t(9;22)(q34;q11.2). Not FAB M3 AML

AML = acute myeloid leukemia; AZA = azacitidine; BSC = best supportive care; CAR T-cell therapy = chimeric antigen receptor T-cell therapy; CNS = central nervous system; ECOG = Eastern Cooperative Oncology Group; GSF = granulocyte stimulating factor; HMA = hypomethylating agent; LDAC = low-dose cytarabine; MDS = myelodysplastic syndrome; PBO = placebo; PML = promyelocytic leukemia; RCT = randomized controlled trial; SC = subcutaneous; VEN = venetoclax.

Source: Indirect treatment comparison report.39

Table 17 shows a summary of patient baseline characteristics for the 4 studies included in the NMA. Only the arms used in the NMA are included. The table is in 2 panels, the first showing demographic and clinical characteristics and the second showing the cytogenetic and mutation data. Table 19 shows an assessment of heterogeneity based on the study and patient characteristics. The most important source of heterogeneity was in indicators of disease severity, bone marrow blast counts, proportion of patients with poor cytogenetic risk, and baseline ECOG PS.

Table 17: Summary of Patient Baseline Characteristics

Study

Treatment

N

Age (years),

median (range)

Gender (male)

n (%)

ECOG/ WHO PS

0 or 1, n (%)

ECOG/ WHO PS

2, n (%)

Primary/de novo

AML, n (%)

Secondary AML,

n (%)

VIALE-A

VEN + AZA

286

76.0
(49 to 91)

172 (60.1)

157 (54.9)

113 (39.5)

214 (74.8)

72 (25.2)

PBO + AZA

145

76.0
(60 to 90)

87 (60.0)

81 (55.9)

59 (40.7)

110 (75.9)

35 (24.1)

VIALE-C

VEN + LDAC

143

76.0
(36 to 93)

78 (54.5)

74 (51.7)

63 (44.1)

85 (59.4)

58 (40.6)

Placebo + LDAC

68

76.0
(41 to 88)

39 (57.4)

34 (50.0)

25 (36.8)

45 (66.2)

23 (33.8)

CCR + preselected LDAC

20

71.0
(56 to 83)

15 (75.0)

19 (95.0)

0 (0.0)

NR

NR

CCR + preselected BSC

45

78.0
(67 to 89)

29 (64.4)

30 (66.7)

15 (33.3)

NR

NR

CCR + preselected LDAC

158

75.0
(65 to 88)

94 (59.5)

123 (77.8)

35 (22.2)

NR

NR

AML = acute myeloid leukemia; AZA = azacitidine; BSC = best supportive care; CCR = conventional care regimen; ECOG = Eastern Cooperative Oncology Group; LDAC = low-dose cytarabine; NR = not reported; PS = Performance Status; VEN = venetoclax.

Source: Systematic review report.38

Table 18: Summary of Patient Baseline Characteristics

Trial

Treatment

Cytogenetic risk

WBC (95% CI)

n (%)

Platelets

n (%)

Bone marrow blasts

Intermediate, n (%)

Poor, n (%)

% (95% CI)

< 30%
n (%)

30 to < 50%
n (%)

≥ 50%

n (%)

VIALE-A

VEN + AZA

182 (63.6%)

104 (36.4%)

NR

NR

47.0 (4.4 to 100.0)

85 (29.7%)

61 (21.3%)

140 (49.0%)

PBO + AZA

89 (61.4%)

56 (38.6%)

NR

NR

47.0 (11.0 to 99.0)

41 (28.3%)

33 (22.8%)

71 (49.0%)

VIALE-C

VEN + LDAC

91 (63.6%)

47 (32.9%)

NR

NR

NR

42 (29.4%)

36 (25.2%)

65 (45.5%)

PBO + LDAC

46 (67.6%)

20 (29.4%)

NR

NR

NR

18 (26.5%)

22 (32.4%)

28 (41.2%)

CCR + preselected LDAC

18 (90.0%)

1 (5.0%)

NR

NR

22.0 (20.0 to 28.0)

NR

NR

NR

CCR + preselected LDAC

104 (65.8%)

54 (34.2%)

2.3 (0.0 to 73.0)

54 (6, 327)

74.0 (4.0 to 100.0)

NR

NR

128 (81.0%)

AML = acute myeloid leukemia; AZA = azacitidine; BSC = best supportive care; CCR = conventional care regimen; LDAC = low-dose cytarabine; NR = not reported; PBO = placebo; VEN = venetoclax.

Source: Systematic review report.38

Table 19: Assessment of Heterogeneity for NMA

Detail

Description and handling of potential effect modifiers

Disease severity

Patient groups varied in bone marrow blast counts. Where available, median bone marrow blasts ranged from 23.0% to 76%, and 42.1% to 81% of patients had ≥ 50% blasts.

Where available, the proportion of patients with poor cytogenetic risk ranged from 29.6% to 38.6%, with the exception of 1 arm with a single patient (0.5%).

The proportion of patients with poorer ECOG PS ( = 2) varied from 0% to 44.1% across trial arms.

Treatment history

All studies included treatment-naive or newly diagnosed patients with AML.

Clinical trial eligibility criteria

Three studies selected older adults and/or treatment-ineligible patients. One did not specify.

Two studies did not specify threshold for bone marrow blasts, 1 specified ≥ 20% blasts, and 1 specified > 30% blasts.

Three studies prohibited prior treatment with HMAs; 1 study (VIALE-C) permitted it.

Comparators

Dosing was largely consistent across studies:

  • AZA was administered at a dose of 75 mg/m2 SC per day for 7 consecutive days of a 28-day cycle, whether given alone or in combination.

  • LDAC when given alone was administered at a dose of 20 mg/m2 twice a day, and when given in combination with VEN was administered at a dose of 20 mg/m2 once a day. Dosing was for 10 days of a 28-day cycle, except for AZA-001, where it was 14 days.

  • VEN in combination with AZA was administered at a dose of 400 mg once a day for a continuous 28-day cycle, following ramp-up over 3 days (100 mg, 200 mg, 400 mg).

  • VEN in combination with LDAC was administered at a dose of 600 mg once a day for a continuous 28-day cycle, following ramp-up over 4 days (100 mg, 200 mg, 400 mg, 600 mg).

Definitions of end points

Details of end points were not extracted in the report. Variability in end point definitions or assessments was not identified as a source of heterogeneity.

Timing of end point evaluation or trial duration

Median length of study follow-up ranged from 17.5 (VIALE-C) to 24 months (AZA-AML-001).

Withdrawal frequency

Not reported in data extraction. Quality appraisal rated risk of bias due to unexpected imbalances in drop-outs between groups as high for AZA-001 and low for other studies.

Clinical trial setting

Details of setting were not extracted in the report.

Study design

All were parallel group randomized controlled trials. VIALE-A and VIALE-C were double-blind, and AZA-001 and AZA-AML-001 were open-label. AZA-001 and AZA-AML-001 included stratified randomization according to investigator's pre-selection of comparator to LDAC, AZA, and intensive chemotherapy. (Data from the intensive chemotherapy was not used in the ITC.)

AML = acute myeloid leukemia; AZA = azacitidine; ECOG PS = Eastern Cooperative Oncology Group Performance Status; HMA = hypomethylating agent; LDAC = low-dose cytarabine; NMA = network meta-analysis; SC = subcutaneous; VEN = venetoclax.

Source: Systematic review report,38 indirect treatment comparison report.39

Table 20 shows the results of the risk-of-bias assessment for the 4 trials included in the ITC. The quality assessment questions appear in Table 15. Risk of bias was low for all trials for treatment randomization, allocation concealment, and baseline balance. Trials AZA-001 and AZA-AML-001 were open-label, so the risk of bias was high, whereas VIALE-A and VIALE-C were double-blind, with a low risk of bias. AZA-001 was at high risk of bias for imbalance in drop-outs, as more patients appear to have dropped out of the conventional care arm, and there was selective reporting, as overall adverse events were not available. All were at unclear risk of bias for the inclusion of intention-to-treat analyses. Where a reason was given, the concern was with lack of detail on methods for handling missing data.

Table 20: Summary of Risk-of-Bias Assessment

Trial

Randomization

Allocation concealment

Baseline balance

Blinding

Imbalance in drop-outs

Selective reporting

Inclusion of ITT analysis

Risk of bias

(high/low/ unclear)

Risk of bias

(high/low/ unclear)

Risk of bias

(high/low/ unclear)

Risk of bias

(high/low/ unclear)

Risk of bias

(high/low/ unclear)

Risk of bias

(high/low/ unclear)

Risk of bias

(high/low/ unclear)

VIALE-A

Low

Low

Low

Low

Low

Unclear

Unclear

VIALE-C

Low

Low

Low

Low

Low

Unclear

Unclear

AZA-001

Low

Low

Low

High

High

High

Unclear

AZA-AML-001

Low

Low

Low

High

Low

Unclear

Unclear

ITT = intention to treat.

Source: Systematic review report.38

Trial Networks

Figure 7 shows the network for the NMA for OS. Four trials reported this end point and were included in the NMA. The network was linear with a single branch and included 5 treatments. There were no closed loops. Azacitidine was the best-represented treatments with 3 trials contributing data, followed by LDAC with 2.

Figure 7: Network Diagram for the NMA for OS

Schematic of network of nodes depicting treatments with links annotated with the names and dates of individual trials. Nodes are arranged with venetoclax plus low-dose cytarabine and low-dose cytarabine alone on the upper row and venetoclax plus azacitidine, azacitidine alone, and best supportive care on the lower row. Each node on the individual rows is linked with the next, and there is a link between rows between low-dose cytarabine and azacitidine.

BSC = best supportive care; LDAC = low-dose cytarabine; NMA = network meta-analysis; OS = overall survival.

Source: Indirect treatment comparison report.39

Figure 8 shows the network for the NMA for CR + CRi. Three trials reported this end point and were included in the NMA; the fourth trial, AZA-001, did not report data on CRi. The network was linear with a single branch and included 5 treatments. There were no closed loops. Azacitidine and LDAC were the best-represented treatments, with 2 contributing trials each.

Figure 8: Network Diagram for the NMA for CR + CRi

Schematic of network of nodes depicting treatments with links annotated with the names and dates of individual trials. Nodes are arranged with venetoclax plus low-dose cytarabine and low-dose cytarabine alone on the upper row and venetoclax plus azacitidine, azacitidine alone, and best supportive care on the lower row. Each node on the individual rows is linked with the next, and there is a link between rows between low-dose cytarabine and azacitidine.

BSC = best supportive care; CR = complete remission; CRi = complete remission with incomplete hematological recovery; LDAC = low-dose cytarabine; NMA = network meta-analysis.

Source: Indirect treatment comparison report.39

Table 21 shows the data included in the NMAs for the 2 end points of OS and CR + CRi. In the 2 trials comparing azacitidine with BSC, the HRs for OS were 0.60 (95% CI, 0.38 to 0.95) and 0.48 (95% CI, 0.24 to 0.94) for AZA-001 and AZA-AML-001, respectively. In the 2 trials comparing azacitidine with LDAC, the HRs for OS were 0.37 (95% CI, 0.12 to 1.13) and 0.90 (95% CI, 0.70 to 1.16) for AZA-001 and AZA-AML-001, respectively.

Table 21: Data Included in NMAs of OS and CR + CRi, Whole Population

Trial

Treatment arm

OS

CR + CRi

N

HR (95% CI)

N

n

%

VIALE-A

VEN + AZA

286

0.66 (0.52 to 0.85)

286

190

66.43

AZAd

145

145

41

28.28

VIALE-C

VEN-LDAC

143

0.70 (0.50 to 0.99)

143

69

48.25

LDACd

68

68

9

13.24

AZA-001b

AZA

36

0.48 (0.24 to 0.94)

NR

NR

NR

BSCd

27

NR

NR

NR

AZA-001b

AZA

14

0.37 (0.12 to 1.13)

NR

NR

NR

LDACd

20

NR

NR

NR

AZA-AML-001a,c

AZA

44

0.60 (0.38 to 0.95)

44

7

15.91

BSCd

45

47

1

2.13

AZA-AML-001a

AZA

154

0.90 (0.70 to 1.16)

154

42

27.27

LDACd

158

158

41

25.95

AZA = azacitidine; BSC = best supportive care; CR = complete remission; CRi = complete remission with incomplete hematological recovery; LDAC = low-dose cytarabine; NR = not reported; NMA = network meta-analysis; OS = overall survival; VEN = venetoclax.

aAZA-AML-001 (Dombret, 2015) included patients > 30% bone marrow blasts. Patients were randomly assigned on the basis of local pathology assessment of baseline bone marrow blast count; which was subsequently reviewed by the central pathologist; in a small number of cases; baseline blast count was < 30% upon central review.

bAZA-001 (Fenaux, 2009) included patients with 20% to 30% bone marrow blasts. One patient in the BSC group had a bone marrow blast count of 13% but was included based on a peripheral blast count of 20%. In addition; 1 patient in the LDAC arm had blast count of 34%.

cA CR + CRi rate of 0 was reported. In accordance with National Institute for Health and Care Excellence guidance; the numerator and denominator were increased by 1 and 2 respectively to allow estimation of treatment effect.

dComparator treatment.

Source: Indirect treatment comparison report.39

Results
Results of the NMA

OS: Table 22 shows the results for the NMA for OS. Venetoclax plus azacitidine was favoured over comparators azacitidine (HR = 0.66; 95% CrI, 0.52 to 0.85), LDAC (HR = 0.57; 95% CrI, 0.40 to 0.81), and BSC (HR = 0.37; 95% CrI, 0.24 to 0.58), with no treatment favoured between venetoclax plus azacitidine, and venetoclax plus LDAC (HR = 0.81; 95% CrI, 0.50 to 1.31).

Table 22: Pairwise Treatment Comparisons for OS

Treatment,

HR (95% CrI)

LDAC

VEN + AZA

AZA

BSC

VEN + LDAC

LDAC

0.57a

(0.40 to 0.81)

0.86

(0.67 to 1.10)

1.54

(0.98 to 2.43)

0.70a

(0.50 to 0.99)

VEN + AZA

1.75 a

(1.24 to 2.49)

1.51a

(1.18 to 1.94)

2.70a

(1.72 to 4.25)

1.23

(0.76 to 2.01)

AZA

1.16

(0.91 to 1.49)

0.66a

(0.52 to 0.85)

1.78a

(1.22 to 2.62)

0.82

(0.54 to 1.24)

BSC

0.65

(0.41 to 1.03)

0.37a

(0.24 to 0.58)

0.56a

(0.38 to 0.82)

0.46a

(0.26 to 0.81)

VEN + LDAC

1.42a

(1.01 to 1.99)

0.81

(0.50 to 1.31)

1.23

(0.80 to 1.86)

2.19a

(1.23 to 3.85)

AZA = azacitidine; BSC = best supportive care; HR = hazard ratio; LDAC = low-dose cytarabine; OS = overall survival; VEN = venetoclax.

Comparisons should be read as HR for the treatment specified in the column vs. that specified in the row. An HR < 1 favours the treatment specified in the column.

aThe 95% credible interval does not contain 1 (indicating what what be interprested as representing a treatment difference).

Source: Indirect treatment comparison report.39

CR + CRi: Table 23 shows the results for the NMA for OS. Venetoclax plus azacitidine was favoured over comparators azacitidine (OR 5.05; 95% CrI, 3.30 to 7.87), LDAC (OR = 5.42; 95% CrI, 2.80 to 10.50), and BSC (OR = 61.55; 95% CrI, 8.23 to 1,881.53), with no treatment favoured between venetoclax plus azacitidine and venetoclax plus LDAC (OR = 0.86; 95% CrI, 0.30 to 0.35).

Table 23: Pairwise Treatment Comparisons for CR + CRi

Treatment,

OR (95% CrI)

LDAC

VEN + AZA

AZA

BSC

VEN + LDAC

LDAC

5.42a

(2.80 to 10.50)

1.07

(0.64 to 1.78)

0.09a

(0.00 to 0.68)

6.24a

(2.98 to 14.42)

VEN + AZA

0.18a

(0.10 to 0.36)

0.20a

(0.13 to 0.30)

0.02a

(0.00 to 0.12)

1.16

(0.43 to 3.33)

AZA

0.94

(0.56 to 1.56)

5.05a

(3.30 to 7.87)

0.08a

(0.00 to 0.59)

5.84a

(2.39 to 15.22)

BSC

11.38a

(1.47 to 344.71)

61.55a

(8.23 to 1,881.53)

12.07a

(1.70 to 356.61)

73.35a

(8.05 to 2,370.88)

VEN + LDAC

0.16a

(0.07 to 0.34)

0.86

(0.30 to 2.35)

0.17a

(0.07 to 0.42)

0.01a

(0.00 to 0.12)

AZA = azacitidine; BSC = best supportive care; CR = complete remission; CRi = complete remission with incomplete blood count recovery; LDAC = low-dose cytarabine; OR = odds ratio; OS = overall survival; VEN = venetoclax.

Comparisons should be read as OR for the treatment specified in the column vs. that specified in the row. An OR < 1 favours the treatment specified in the row.

aThe 95% credible interval does not contain 1 (indicating what what be interprested as representing a treatment difference).

Source: Indirect treatment comparison report.39

Results of Propensity-Score Analyses: Venetoclax Plus Azacitidine Versus Low-Dose Cytarabine

Table 24 shows the baseline characteristics for the comparison between venetoclax plus azacitidine and LDAC for the analysis of the whole population, before and after weighting. Patients in the LDAC group with favourable cytogenetic risk or prior HMA use were excluded. The largest baseline imbalances in terms of standardized mean difference were in ECOG PS, secondary AML, and race, all of which were reduced by adjustment. This weighting was used for all efficacy analyses for this comparison in this population.

Table 24: Baseline Characteristics for Venetoclax Plus Azacitidine and LDAC Before and After Weighting

Baseline characteristics

Before weighting

After weighting

VEN + AZAa

N = 285

LDACa

N = 50

SMD

P valueb

VEN + AZAa

N = 285

LDACa

N = 50

SMD

P valueb

Age < 75

38.95%

40.00%

0.022

1.000

39.10%

38.67%

0.009

0.955

Female

40.00%

44.00%

0.081

0.708

40.62%

42.05%

0.029

0.853

Race: White

75.79%

68.00%

0.174

0.322

74.62%

74.52%

0.002

0.987

Secondary AML

25.26%

18.00%

0.177

0.354

24.19%

24.97%

0.018

0.917

AML with MRC

32.28%

28.00%

0.093

0.663

31.68%

33.71%

0.043

0.792

Antecedent history of MDS

17.19%

12.00%

0.147

0.479

16.46%

18.47%

0.053

0.765

ECOG Performance Status < 2

55.09%

46.00%

0.183

0.301

53.73%

53.97%

0.005

0.976

IVRS cytogenetic risk: poor

34.74%

32.00%

0.058

0.830

34.29%

33.51%

0.017

0.918

Bone marrow blast count, mean ± SD

52.04 ± 24.26

54.13 ± 24.21

0.086

0.573

52.39 ± 24.32

53.49 ± 24.33

0.045

0.773

AML = acute myeloid leukemia; ECOG = Eastern Cooperative Oncology Group; IVRS = interactive voice recognition system; LDAC = low-dose cytarabine; MDS = myelodysplastic syndrome; MRC = myelodysplasia-related changes; OS = overall survival; SMD = standard mean difference; VEN + AZA = venetoclax plus azacitidine; VEN + LDAC = venetoclax plus low-dose cytarabine.

aTwo patients were excluded from the analysis due to missing data. One had missing data for cytogenetic risk in the LDAC group and 1 had missing bone marrow data in the VEN + AZA arm.

bBefore weighting, categorical variables were compared using chi-square tests and continuous outcomes with analyses of variance (ANOVAs). After weighting, categorical variables were compared using weighted chi-square tests and continuous outcomes with weighted ANOVAs.

Source: Indirect treatment comparison report.39

OS, overall population: Table 25 shows the results of the weighted and unweighted comparisons for OS for the comparison of venetoclax plus azacitidine and LDAC. Figure 9 shows the survival curves for both unweighted and weighted comparisons. Venetoclax plus azacitidine was favoured over LDAC in both the unweighted (HR = 0.47; 95% CI, 0.33 to 0.67) and weighted comparisons (HR = 0.50; 95% CI, 0.35 to 0.73). Median OS in the weighted comparison was 14.69 months (95% CI, 12.12 to 19.25) for venetoclax plus azacitidine compared with 7.43 months (95% CI, 3.15 to 10.18) for LDAC.

Table 25: Comparison of OS for Overall Population, Before and After Weighting

Treatment

N

Events

Before weighting

After weighting

Median OS, months

(95% CI)

HR

(95% CI)

Median OS, months (95% CI)

HR

(95% CI)

VEN + AZA

285

161

14.69

(11.53 to 18.69)

0.47

(0.33 to 0.67)

14.69

(12.12 to 19.25)

0.50

(0.35 to 0.73)

LDACa

50

40

6.13

(2.23 to 8.90)

7.43

(3.15 to 10.18)

AZA = azacitidine; CI = confidence interval; HR = hazard ratio; LDAC = low-dose cytarabine; OS = overall survival; VEN = venetoclax.

aReference treatment.

Source: Indirect treatment comparison report.39

Figure 9: OS for Venetoclax Plus Azacitidine Versus LDAC for the Overall Population, Before and After Weighting

Two plots, placed side by side. The left-hand plot is labelled “before weighting” and the right-hand plot is labelled “after weighting.” Each depicts survival curves for venetoclax plus azacitidine and low-dose cytarabine between 0 and 30 months. The curves diverge from month 0, with venetoclax plus azacitidine as the upper and low-dose cytarabine as the lower. On the right-hand plot, the low-dose cytarabine curve is slightly shallower than the corresponding curve on the right. In both, venetoclax plus azacitidine is the smoother of the curves, while low-dose cytarabine is more stepped. The Y axes are labelled “Proportion free of event,” from 0% to 100%, and the X axes are in months, up to 35 months.

AZA = azacitidine; LDAC = low-dose cytarabine; VEN = venetoclax.

Source: Indirect treatment comparison report.39

Event-free survival, overall population: Table 26 shows the results of the weighted and unweighted comparison for EFS between venetoclax plus azacitidine and LDAC. Figure 10 shows the survival curves for both weighted and weighted comparisons. Venetoclax plus azacitidine was favoured over LDAC in both the unweighted (HR = 0.40; 95% CI, 0.28 to 0.56) and the weighted comparisons (HR = 0.40; 95% CI, 0.28 to 0.58). Median EFS in the weighted comparison was 9.79 months (95% CI, 8.41 to 11.99) for venetoclax plus azacitidine compared with 3.06 months (95% CI, 1.71 to 5.82) for LDAC.

Table 26: Comparison of EFS for Overall Population, Before and After Weighting

Treatment

N

Events

Before weighting

After weighting

Median OS, months

(95% CI)

HR

(95% CI)

Median, months

OS (95% CI)

HR

(95% CI)

VEN + AZA

285

190

9.66

(8.41 to 11.53)

0.40

(0.28 to 0.56)

9.79

(8.41 to 11.99)

0.40

(0.28 to 0.58)

LDACa

50

43

3.02

(1.45 to 5.72)

3.06

(1.71 to 5.82)

AZA = azacitidine; CI = confidence interval; EFS = event-free survival; HR = hazard ratio; LDAC = low-dose cytarabine; OS = overall survival; VEN = venetoclax.

aReference treatment.

Source: Indirect treatment comparison report.39

Figure 10: EFS for Venetoclax Plus Azacitidine Versus LDAC for the Overall Population, Before and After Weighting

Two plots, placed side by side. The left-hand plot is labelled “before weighting” and the right-hand is labelled “after weighting.” Each depicts survival curves for venetoclax plus azacitidine and low-dose cytarabine between 0 and 30 months. The curves diverge from month 0, with venetoclax plus azacitidine as the upper and low-dose cytarabine as the lower. On the right-hand plot, the low-dose cytarabine curve is slightly shallower than the corresponding curve on the right. In both, venetoclax plus azacitidine is the smoother of the curves, while low-dose cytarabine is more stepped. The Y axes are labelled “Proportion free of event” with a scale from 0% to 100%, and the X axes are in months, up to 35 months.

AZA = azacitidine; EFS = event-free survival; LDAC = low-dose cytarabine.

Source: Indirect treatment report.39

CR + CRi, overall population: Table 27 shows the results for the comparison between venetoclax plus azacitidine and LDAC for CR, CRi, and CR + CRi before and after weighting. Venetoclax plus azacitidine was favoured over LDAC for CR + CRi in both the unweighted (OR = 10.32; 95% CI, 4.67 to 22.89) and weighted comparison (OR = 10.17; 95% CI, 4.55 to 22.73). After weighting, the proportion of patients with CR + CRi was 0.66 (95% CI, 0.61 to 0.72) for venetoclax plus azacitidine and 0.16 (95% CI, 0.08 to 0.29) for LDAC.

Table 27: Comparison of CR, CRi, and CR + CRi for VEN Plus AZA Versus LDAC for the Overall Population, Before and After Weighting

Outcome

Before weighting

After weighting

VEN + AZA

% (95% CI)

LDAC

% (95% CI)

OR for VEN + AZA vs. LDAC (95% CI)

VEN + AZA

% (95% CI)

LDAC

% (95% CI)

OR for VEN + AZA vs. LDAC (95% CI)

CR

0.37

(0.31 to 0.43)

0.10

(0.04 to 0.22)

5.25

(2.02 to 13.64)

0.37

(0.31 to 0.43)

0.10

(0.04 to 0.22)

5.17

(1.97 to 13.56)

CRi

0.29

(0.24 to 0.35)

0.06

(0.02 to 0.17)

6.55

(1.98 to 21.62)

0.29

(0.24 to 0.35)

0.06

(0.02 to 0.17)

6.46

(1.94 to 21.52)

CR + CRi

0.66

(0.61 to 0.72)

0.16

(0.08 to 0.29)

10.34

(4.67 to 22.89)

0.66

(0.61 to 0.72)

0.16

(0.08 to 0.29)

10.17

(4.55 to 22.73)

AZA = azacitidine; CR = complete remission; CRi = complete remission with incomplete hematological recovery; OR = odds ratio; LDAC = low-dose cytarabine; VEN = venetoclax; vs. = versus.

Source: Indirect treatment comparison report.39

Sensitivity analysis, overall population: The sensitivity analysis included all patients enrolled in the LDAC arm in VIALE-C (n = 66), regardless of prior HMA use or good cytogenetic risk. The results for the comparison of venetoclax plus azacitidine versus LDAC were consistent with the results from the main analysis, with an after-weighting OS HR of 0.47 (95% CI, 0.34 to 0.66), an EFS HR of 0.38 (95% CI, 0.28 to 0.43), and an OR for CR + CRi of 10.52 (95% CI, 4.90 to 22.58).

OS, subgroup with baseline bone marrow blast count of 30% or greater: Table 28 shows the baseline characteristics for the comparison between venetoclax plus azacitidine and LDAC for the analysis of the subpopulation of patients with 30% or greater blasts, before and after weighting. Patients in VIALE-C with favourable cytogenetic risk or prior HMA use were excluded. The largest baseline imbalances in terms of standard mean difference were in ECOG PS, secondary AML, and race, all of which were reduced by adjustment. This weighting was used for all efficacy analyses for this comparison in this subgroup.

Table 28: Baseline Characteristics for VEN Plus AZA and LDAC Before and after Weighting, Patients With Baseline Bone Marrow Blast Count of 30% or Greater

Baseline characteristics

Before weighting

After weighting

VEN + AZAa

N = 206

LDACa

N = 36

SMD

P valueb

VEN + AZAa

N = 206

LDACa

N = 36

SMD

P valueb

Age < 75

37.86%

47.22%

0.190

0.381

39.33%

41.50%

0.044

0.813

Female

39.32%

36.11%

0.066

0.858

38.88%

40.06%

0.024

0.900

Race: White

74.27%

63.89%

0.226

0.277

72.71%

72.40%

0.007

0.968

Secondary AML

23.79%

16.67%

0.178

0.468

22.72%

22.76%

0.001

0.996

AML with MRC

26.70%

25.00%

0.039

0.993

26.47%

28.65%

0.049

0.805

Antecedent history of MDS

16.99%

11.11%

0.170

0.468

16.16%

17.23%

0.029

0.892

ECOG Performance Status < 2

55.34%

41.67%

0.276

0.182

53.23%

49.83%

0.068

0.721

IVRS cytogenetic risk: poor

32.04%

38.89%

0.144

0.539

32.97%

32.60%

0.008

0.967

Bone marrow blast count, mean ± SD

62.78 ± 19.75

65.75 ± 17.90

0.157

0.400

63.24 ± 19.77

64.91 ± 16.67

0.091

0.580

AML = acute myeloid leukemia; ANOVA = analysis of variance; AZA = azacitidine; ECOG = Eastern Cooperative Oncology Group; IVRS = interactive voice recognition system; LDAC = low-dose cytarabine; MDS = myelodysplastic syndrome; MRC = myelodysplasia-related changes; OS = overall survival; SD = standard deviation; SMD = standard mean difference; VEN = venetoclax.

aOne patient was excluded from the analysis due to missing data for cytogenetic risk in the LDAC group and 1 had missing bone marrow data in the VEN + AZA arm.

bBefore weighting, categorical variables were compared using chi-square tests and continuous outcomes with ANOVAs. After weighting, categorical variables were compared using weighted chi-square tests and continuous outcomes with weighted ANOVAs.

Source: Indirect treatment comparison report.39

OS, subgroup with baseline bone marrow blast count of 30% or greater: Table 29 shows the results of the weighted and unweighted comparison for OS for the comparison of venetoclax plus azacitidine versus LDAC in patients with a baseline bone marrow blast count of 30% or greater. Venetoclax plus azacitidine was favoured over LDAC in both the unweighted (HR = 0.47; 95% CI, 0.33 to 0.67) and the weighted comparisons (HR = 0.47; 95% CI, 0.32 to 0.69). Median OS in the weighted comparison was 14.06 months (95% CI, 10.61 to 17.15) for venetoclax plus azacitidine compared with 3.61 months (95% CI, 3.12 to 10.18) for LDAC.

Table 29: Comparison of OS for Patients With a Bone Marrow Blast Count of 30% or Greater Before and After Weighting

Treatment

N

Events

Before weighting

After weighting

Median OS, months

(95% CI)

HR

(95% CI)

Median OS, months (95% CI)

HR

(95% CI)

VEN + AZA

206

121

14.06

(10.41 to 16.95)

0.47

(0.31 to 0.70)

14.06

(10.61 to 17.15)

0.47

(0.32 to 0.69)

LDACa

36

30

3.61

(1.87 to 7.85)

3.61

(3.12 to 10.18)

AZA = azacitidine; CI = confidence interval; HR = hazard ratio; LDAC = low-dose cytarabine; OS = overall survival; VEN = venetoclax.

aReference treatment.

Source: Indirect treatment comparison report.39

EFS, subgroup with baseline bone marrow blast count of 30% or greater: Table 30 shows the results of the weighted and unweighted comparison for EFS between venetoclax plus azacitidine and LDAC in patients with a baseline bone marrow blast count of 30% or greater. Venetoclax plus azacitidine was favoured over LDAC in both the unweighted (HR = 0.41; 95% CI, 0.28 to 0.61) and the weighted comparisons (HR = 0.42; 95% CI, 0.28 to 0.61). Median EFS in the weighted comparison was 9.00 (95% CI, 7.69 to 11.53) months for venetoclax plus azacitidine compared with 3.06 (95% CI, 1.71 to 5.82) months for LDAC.

Table 30: Comparison of EFS for Patients With Bone a Marrow Blast Count of 30% or Greater Before and After Weighting

Treatment

N

Events

Before weighting

After weighting

Median OS, months

(95% CI)

HR

(95% CI)

Median, months

OS (95% CI)

HR

(95% CI)

VEN + AZA

206

136

9.00

(7.59 to 11.50)

0.41

(0.28 to 0.61)

9.00

(7.69 to 11.53)

0.42

(0.28 to 0.61)

LDACa

36

32

3.02

(1.45 to 5.72)

3.06

(1.71 to 5.82)

AZA = azacitidine; CI = confidence interval; EFS = event-free survival; HR = hazard ratio; LDAC = low-dose cytarabine; OS = overall survival; VEN = venetoclax.

aReference treatment.

Source: Indirect treatment comparison report.39

CR + CRi, subgroup with baseline bone marrow blast count of 30% or greater: Table 31 shows the results for the comparison between venetoclax plus azacitidine and LDAC for CR, CRi, and CR + CRi before and after weighting, for patients with a bone marrow blast count of 30% or greater. Venetoclax plus azacitidine was favoured over LDAC for CR + CRi in both the unweighted (OR = 10.39; 95% CI, 3.88 to 27.84) and weighted comparison (OR = 10.80; 95% CI, 3.89 to 29.94). After weighting, the proportion of patients with CR + CRi was 0.62 (95% CI, 0.55 to 0.69) for venetoclax plus azacitidine and 0.13 (95% CI, 0.05 to 0.29) for LDAC.

Table 31: Comparison of CR, CRi, and CR + CRi for Patients With a Bone Marrow Blast Count of 30% or Greater Before and After Weighting

Outcome

Before weighting

After weighting

VEN + AZA

% (95% CI)

LDAC

% (95% CI)

OR for VEN + AZA vs. LDAC (95% CI)

VEN + AZA

% (95% CI)

LDAC

% (95% CI)

OR for VEN + AZA vs. LDAC (95% CI)

CR

0.34

(0.28 to 0.41)

0.08

(0.03 to 0.23)

5.79

(1.72 to 19.51)

0.34

(0.28 to 0.41)

0.08

(0.03 to 0.24)

5.75

(1.62 to 20.48)

CRi

0.28

(0.22 to 0.35)

0.06

(0.01 to 0.20)

6.66

(1.55 to 28.63)

0.28

(0.22 to 0.35)

0.05

(0.01 to 0.18)

7.49

(1.73 to 32.52)

CR + CRi

0.63

(0.56 to 0.69)

0.14

(0.06 to 0.29)

10.39

(3.88 to 27.84)

0.62

(0.5 to 0.69)

0.13

(0.05 to 0.29)

10.80

(3.89 to 29.94)

AZA = azacitidine; CI = confidence interval; CR = complete remission; CRi = complete remission with incomplete hematological recovery; OR = odds ratio; LDAC = low-dose cytarabine; VEN = venetoclax; vs. = versus.

Source: Indirect treatment comparison report.39

Sensitivity analysis, subgroup with baseline bone marrow blast count of 30% or greater: The sensitivity analysis included all patients enrolled in the LDAC arm in VIALE-C (n = 66), regardless of prior HMA use or good cytogenetic risk. The results for the comparison of venetoclax plus azacitidine versus LDAC were consistent with the results from the main analysis, with an after-weighting OS HR of 0.47 (95% CI, 0.34 to 0.66), an EFS HR of 0.38 (95% CI, 0.28 to 0.43), and an OR for CR + CRi of 9.99 (95% CI, 3.85 to 25.94).

Results of 3-Way Propensity-Score Analyses: Venetoclax Plus Azacitidine Versus LDAC Versus Azacitidine

Table 32 shows the baseline characteristics for the 3-way comparison for venetoclax plus azacitidine versus LDAC versus azacitidine for the analysis of the whole population, before and after weighting. Patients in the LDAC group with favourable cytogenetic risk or prior HMA use were excluded. The largest baseline imbalances in terms of standard mean difference were in ECOG PS, secondary AML, and race, all of which were reduced by adjustment. This weighting was used for all efficacy analyses for this comparison in this population.

Table 32: Baseline Characteristics for VEN Plus AZA, LDAC, and AZA Before and After Weighting

Baseline characteristics

VEN + AZA

N = 286

LDACa

N = 50

AZA

N = 145

VEN + AZA vs. LDAC

AZA vs. LDAC

VEN + AZA vs. AZA

SMD

P value2

SMD

P valueb

SMD

P valueb

Before weighting

Age < 75

39.16%

40.00%

40.00%

0.017

1.000

0.000

1.000

0.017

0.949

Female

39.86%

44.00%

40.00%

0.084

0.694

0.081

0.742

0.003

1.000

Race: White

75.87%

68.00%

75.17%

0.176

0.315

0.160

0.422

0.016

0.967

Secondary AML

25.17%

18.00%

24.14%

0.175

0.360

0.151

0.484

0.024

0.907

AML with MRC

32.17%

28.00%

33.79%

0.091

0.674

0.126

0.562

0.035

0.817

History of MDS

17.13%

12.00%

18.62%

0.146

0.485

0.185

0.391

0.039

0.803

ECOG Performance Status < 2

54.90%

46.00%

55.86%

0.179

0.313

0.198

0.298

0.019

0.930

IVRS cytogenetic risk: Poor

34.97%

32.00%

36.55%

0.063

0.806

0.096

0.683

0.033

0.827

Bone marrow blast count, mean ± SDc

52.04 ± 24.26

54.13 ± 24.21

53.46 ± 24.52

0.086

0.573

0.028

0.867

0.058

0.567

After weighting

Age < 75

39.16%

39.79%

40.00%

0.013

0.937

0.004

0.980

0.017

0.949

Female

39.86%

40.08%

40.00%

0.005

0.977

0.002

0.992

0.003

1.000

Race: White

75.87%

74.97%

75.17%

0.021

0.888

0.005

0.977

0.016

0.967

Secondary AML

25.17%

25.41%

24.14%

0.005

0.976

0.029

0.876

0.024

0.907

AML with MRC

32.17%

35.65%

33.79%

0.074

0.663

0.039

0.828

0.035

0.817

History of MDS

17.13%

20.76%

18.62%

0.093

0.615

0.054

0.784

0.039

0.803

ECOG Performance Status < 2

54.90%

56.86%

55.86%

0.040

0.802

0.020

0.905

0.019

0.930

IVRS cytogenetic risk: Poor

34.97%

36.73%

36.55%

0.037

0.824

0.004

0.983

0.033

0.827

Bone marrow blast count, mean ± SDc

52.04 ± 24.26

55.51 ± 24.31

53.46 ± 24.52

0.143

0.366

0.084

0.617

0.058

0.567

AML = acute myeloid leukemia; ANOVA = analysis of variance; AZA = azacitidine; ECOG = Eastern Cooperative Oncology Group; IVRS = interactive voice recognition system; LDAC = low-dose cytarabine; MDS = myelodysplastic syndrome; MRC = myelodysplasia-related changes; OS = overall survival; SMD = standard mean difference; VEN = venetoclax; vs. = versus.

aOne patient from the LDAC arm was removed from the analysis due to missing cytogenetic risk.

bCategorical outcomes were compared using chi-square tests and continuous outcomes with ANOVAs.

cBone marrow blast count for the VEN + AZA arm of VIALE-A was calculated among 285 patients with non-missing data for that variable.

Source: Indirect treatment comparison report.39

OS, overall population: Table 33 shows the median OS for venetoclax plus azacitidine, LDAC, and azacitidine before and after weighting. Table 34 shows the weighted and weighted results for the comparisons, and Figure 11 shows the survival curves.

Venetoclax plus azacitidine was favoured over LDAC in both the unweighted (HR = 0.47; 95% CI, 0.33 to 0.66) and weighted (HR = 0.52; 95% CI, 0.36 to 0.77) comparisons. Before weighting, azacitidine was favoured over LDAC (HR = 0.69; 95% CI, 0.48 to 0.99) but, after weighting, there was no statistically significant difference (HR = 0.78; 95% CI, 0.52 to 1.17). With weighting, the median OS for LDAC increased from 6.1 months (95% CI, 2.2 to 8.9) to 7.4 months (95% CI, 3.2 to 14.3), compared with 14.7 months (95% CI, 11.9 to 18.7) for venetoclax plus azacitidine and 9.6 months (95% CI, 7.4 to 12.7) for azacitidine alone.

Table 33: OS for Venetoclax Plus Azacitidine, LDAC, and Azacitidine Before and After Weighting

Treatment

N

Events

Median OS (95% CI)

Before weighting

After weighting

VEN + AZA

286

161

14.7 (11.9 to 18.7)

14.7 (11.9 to 18.7)

LDAC

50

40

6.1 (2.2 to 8.9)

7.4 (3.2 to 14.3)

AZA

145

109

9.6 (7.4 to 12.7)

9.6 (7.4 to 12.7)

AZA = azacitidine; LDAC = low-dose cytarabine; OS = overall survival; VEN = venetoclax.

Source: Indirect treatment comparison report.39

Table 34: Comparison of OS for Venetoclax Plus Azacitidine, LDAC, and Azacitidine Before and After Weighting

Comparison

HR (95% CI)

Before weighting

After weighting

VEN + AZA vs. LDAC

0.47 (0.33 to 0.66)

0.52 (0.36 to 0.77)

VEN + AZA vs. AZA

0.64 (0.50 to 0.82)

0.64 (0.50 to 0.82)

AZA vs. LDAC

0.69 (0.48 to 0.99)

0.78 (0.52 to 1.17)

AZA = azacitidine; CI = confidence interval; HR = hazard ratio; LDAC = low-dose cytarabine; OS = overall survival; VEN = venetoclax; vs. = versus.

Source: Indirect treatment comparison report.39

Figure 11: OS for Venetoclax Plus Azacitidine, LDAC, and Azacitidine Before and After Weighting

Two plots, placed side by side. The left-hand plot is labelled “before weighting” and the right-hand plot is labelled “after weighting.” Each depicts survival curves for venetoclax plus azacitidine, azacitidine, and low-dose cytarabine between 0 and 30 months. low-dose cytarabine diverges from the other 2 from month 0, and venetoclax plus azacitidine from azacitidinearound month 3, with VEN plus azacitidine, azacitidine, and low-dose cytarabine as the upper, middle, and lower curves, respectively. On the right-hand plot, the low-dose cytarabine and azacitidine curves are slightly shallower than the corresponding curve on the right, and the azacitidine and low-dose cytarabine curves approach each other after month 15. In both, venetoclax plus azacitidine and azacitidineare the smoother of the curves, while low-dose cytarabine is more stepped. The Y axes are labelled proportion free of event with a scale from 0% to 100%, and the X axes are in months, up to 35 months.

AZA = azacitidine, LDAC = low-dose cytarabine; OS = overall survival.

Source: Indirect treatment report.39

EFS: Table 35 shows the EFS for venetoclax plus azacitidine, LDAC, and azacitidine before and after weighting. Table 35 shows the unweighted and weighted results for the comparisons, and Figure 12 shows the survival curves.

Venetoclax plus azacitidine was favoured over LDAC in both the unweighted (HR = 0.40; 95% CI, 0.28 to 0.56) and weighted comparisons (HR = 0.41; 95% CI, 0.29 to 0.59). Azacitidine was also favoured over LDAC in both weighted (HR = 0.61; 95% CI, 0.43 to 0.86) and unweighted (HR = 0.63; 95% CI, 0.43 to 0.92) comparisons. With weighting, the median EFS for LDAC increased from 3.0 months (95% CI, 1.5 to 5.7) to 3.1 months (95% CI, 1.8 to 5.8), compared with 9.8 months (95% CI, 8.4 to 11.8) for venetoclax plus azacitidine and 7.0 months (95% CI, 5.6 to 9.5) for azacitidine.

Table 35: EFS for Venetoclax Plus Azacitidine, LDAC, and Azacitidine Before and After Weighting

Treatment

N

Events

Median EFS (95% CI)

Before Weighting

After Weighting

VEN + AZA

286

191

9.8 (8.4 to 11.8)

9.8 (8.4 to 11.8)

LDAC

50

43

3.0 (1.5 to 5.7)

3.1 (1.8 to 5.8)

AZA

145

122

7.0 (5.6 to 9.5)

7.0 (5.6 to 9.5)

AZA = azacitidine; CI = confidence interval; EFS = event-free survival; LDAC = low-dose cytarabine; VEN = venetoclax.

Source: Indirect treatment comparison report.39

Table 36: EFS for Comparisons of Venetoclax Plus Azacitidine With LDAC, Venetoclax Plus Azacitidine With Azacitidine, and Azacitidine With LDAC

Treatment

HR (95% CI)

Before weighting

After weighting

VEN + AZA vs. LDAC

0.40 (0.28 to 0.56)

0.41 (0.29 to 0.59)

VEN + AZA vs. AZA

0.62 (0.49 to 0.77)

0.62 (0.49 to 0.77)

AZA vs. LDAC

0.61 (0.43 to 0.86)

0.63 (0.43 to 0.92)

AZA = azacitidine; CI = confidence interval; EFS = event-free survival; HR = hazard ratio; LDAC = low-dose cytarabine; VEN = venetoclax.

Source: Indirect treatment comparison report.39

Figure 12: EFS for Venetoclax Plus Azacitidine, LDAC, and Azacitidine Before and After Weighting

Two plots, placed side by side. The left-hand plot is labelled “before weighting” and the right-hand plot is labelled “after weighting.” Each depicts survival curves for venetoclax plus azacitidine, azacitidine, and low-dose cytarabine between 0 and 30 months. In each plot, the curves diverge from each other around month 1, with venetoclax plus azacitidine, azacitidine, and low-dose cytarabine as the upper, middle, and lower curves, respectively. Around month 20, the azacitidine and low-dose cytarabine curves approach each other. On the right-hand plot, the low-dose cytarabine and azacitidine curves are slightly shallower than the corresponding curve on the right. In both, venetoclax plus azacitidine and azacitidine are the smoother of the curves, while low-dose cytarabine is more stepped. The Y axes are labelled proportion free of event with a scale from 0% to 100%, and the X axes are in months, up to 35 months.

AZA = azacitidine, EFS = event-free survival; LDAC = low-dose cytarabine.

Source: Indirect treatment comparison report.39

CR + CRi, overall population: Table 37 shows the results for the comparison between venetoclax plus azacitidine and LDAC for CR, CRi, and CR + CRi, before and after weighting, for patients with a bone marrow blast count of 30% or greater. Venetoclax plus azacitidine was favoured over LDAC for CR + CRi in both the unweighted (OR = 10.39; 95% CI, 4.69 to 23.01) and weighted comparisons (OR = 9.69; 95% CI, 4.30 to 21.85). No statistically significant difference was seen between azacitidine and LDAC in either the unweighted (OR = 2.07; 95% CI, 0.90 to 4.78) or weighted comparison (OR = 1.93; 95% CI, 0.82 to 4.54). After weighting, the proportion of patients with CR + CRi was 0.62 (95% CI, 0.55 to 0.69) for venetoclax plus azacitidine, 0.28 (95% CI, 0.21 to 0.36) for azacitidine, and 0.13 (95% CI, 0.05 to 0.29) for LDAC.

Table 37: Comparison of CR, CRi, and CR + CRi for VEN Plus AZA, LDAC, and AZA Before and After Weighting

Outcome

VEN + AZA

% (95% CI)

LDAC

% (95% CI)

AZA

% (95% CI)

VEN + AZA vs. LDAC
OR (95% CI)

AZA vs. LDAC

OR (95% CI)

VEN + AZA vs. AZA

OR (95% CI)

Before weighting

CR

0.37

(0.31 to 0.43)

0.10

(0.04 to 0.22)

0.18

(0.12 to 0.25)

5.22

(2.01 to 13.56)

1.97

(0.71 to 5.43)

2.66

(1.63 to 4.32)

CRi

0.30

(0.25 to 0.35)

0.06

(0.02 to 0.17)

0.10

(0.06 to 0.17)

6.63

(2.01 to 21.87)

1.81

(0.50 to 6.53)

3.67

(2.03 to 6.62)

CR + CRi

0.66

(0.61 to 0.72)

0.16

(0.08 to 0.29)

0.28

(0.21 to 0.36)

10.39

(4.69 to 23.01)

2.07

(0.90 to 4.78)

5.02

(3.24 to 7.77)

After weighting

CR

0.37

(0.31 to 0.43)

0.11

(0.04 to 0.23)

0.18

(0.12 to 0.25)

4.93

(1.88 to 12.98)

1.86

(0.66 to 5.20)

2.66

(1.63 to 4.33)

CRi

0.30

(0.25 to 0.35)

0.06

(0.02 to 0.19)

0.10

(0.06 to 0.17)

6.15

(1.80 to 21.00)

1.68

(0.45 to 6.26)

3.67

(2.03 to 6.63)

CR + CRi

0.66

(0.61 to 0.72)

0.17

(0.09 to 0.31)

0.28

(0.21 to 0.36)

9.69

(4.30 to 21.85)

1.93

(0.82 to 4.54)

5.02

(3.24 to 7.77)

AZA = azacitidine; CR = complete remission; CRi = complete remission with incomplete hematological recovery; OR = odds ratio; LDAC = low-dose cytarabine; VEN = venetoclax.

Source: Indirect treatment comparison report.39

Critical Appraisal of Indirect Treatment Comparisons

NMA and propensity-score analyses are considered separately in this section.

The key limitations of the NMA include the small size and structure of the network, which had no closed loops, potential sources of heterogeneity across the trials related to differences in study design, and patient characteristics. These limitations resulted in imprecise estimates and the potential for bias.

The key limitations of the propensity-score analyses include the intrinsic high susceptibility to bias of the method due to the lack of an anchor for comparison and the possibility for unmeasured covariates and residual confounding. Relatively small numbers were involved, with the potential for unstable estimates susceptible to the influence of high weights.

Critical Appraisal of Systematic Review

The ITC (NMA) was based on a systematic literature review that identified studies according to pre-specified inclusion criteria. These included a broad selection of comparators and outcomes. The literature search was last conducted in October 2020 and appeared comprehensive in terms of the databases searched and the search strategy. Two sets of selection criteria were applied: an initial broader set of criteria and, at the full-text review step, a narrowed set of criteria intended to create a high-quality dataset for meta-analysis. The selection of comparators was not justified, but those meaningful to the Canadian context were included. Lists of studies excluded at the full-text state for both sets of criteria were provided. Screening and selection was done by 2 independent reviewers, with a third involved to reconcile differences. Data extraction was also done by 2 independent reviewers. The data were extracted to pre-designed data sheets, with differences reconciled by a third reviewer.

Critical Appraisal of the NMA

Studies included in the NMA were selected from those identified by the systematic literature review. The criteria for the inclusion of studies for the NMA were provided and are consistent with the objective. The eligible interventions were restricted further to those used in Canada for the treatment of the population of interest, which was defined as treatment-naive adult patients with AML who are ineligible for intensive chemotherapy. Only clinical efficacy outcomes were pre-specified for the NMA. Available data limited the end points further to OS and CR + CRi for the NMA and OS, EFS, and CR + CRi for the propensity-score analysis. Patient-reported QoL and safety end points were not represented.

Heterogeneity in study and patient baseline characteristics was reported and reviewed by the authors as part of the assessment of feasibility for the meta-analysis. Baseline differences were noted in the prognostic variables and potential treatment-effect modifiers of blast count at baseline, prior treatment with an HMA, and cytogenetic risk. The proportion of patients with 50% or greater bone marrow blasts at baseline ranged from 0% (AZA-001 trial) to 100% (AZA-AML-001) in the network for OS and 70.8% (VIALE-A) to 100% (AZA-AML-001) in the network for CR + CRi. Patients with prior HMA treatment were excluded from VIALE-A, AZA-001, and AZA-AML-001, but not from VIALE-C, in which 19.9% had been treated with an HMA. This might represent a group more refractory to treatment with azacitidine, affecting both OS and CR + CRi end points. Patients with poor cytogenetic risk were more represented in the azacitidine arm of VIALE-A compared with the azacitidine arm of AZA-001 (39% versus 26%). This difference potentially affects the NMA network for OS. The median length of study follow-up ranged from 17.5 (VIALE-C) to 24 months (AZA-AML-001). The variability was unlikely to affect CR + CRi, as response tended to occur early, but may affect OS, as patients may be censored before OS events in studies with short follow-up.

Four studies formed a mainly linear connected network for OS and 3 studies for CR + CRi. The end point of CR + CRi was not reported for AZA-001. There were no closed loops in the network, meaning that inconsistency within the network could not be statistically assessed. The dose and duration for azacitidine and the dose (but not duration) for LDAC was the same across trials, and BSC included the same constituents, limiting heterogeneity in dosing. In AZA-001 and AZA-AML-001, patients were preselected for the comparator therapy, so the comparison of azacitidine against LDAC was made in patients preselected for LDAC, and the comparison of azacitidine against BSC was made in patients preselected for BSC. These were treated as 2 separate contrasts, not as a 3-arm trial.

A standard Bayesian generalized linear model was used for the meta-analysis and the diagnostics and model selection were sufficiently described. The reviewers checked the proportional hazards assumption for OS for the contributing plots using log-log plots. The risk of violation of the proportional hazards assumption was low for VIALE-A and VIALE-C and low to moderate for AZA-AML-001, where the survival curves were largely overlapping and intermittently crossing. The model in the NMA used assumed constant hazards, which was an appropriate choice, given the low-to-moderate risk of violation of the proportional hazards assumption and the small number of studies available.

The networks for all analyses were small. Thus, the decision was made a priori to limit the analysis to fixed-effects models. This entailed the assumption that between-study heterogeneity was zero, which was unlikely to be the case. The small number of studies led to imprecise estimates, with the risk of not detecting a difference. In the analysis of CR + CRi, low response counts (including 0, requiring a zero-cell adjustment) led to highly uncertain estimates with wide CrIs. The small number of studies meant there was no opportunity to use statistical methods (such as meta-regression) to adjust for variability in baseline treatment-effect modifiers and correct for potential bias. Finally, non-informative prior distributions were used in the models, as is usual practice, under the assumption that the final estimates will reflect only the data. However, with a low information dataset, the prior distributions may add to the imprecision. Consideration of alternative priors was mentioned but not detailed.

Critical Appraisal of Propensity-Score Analyses

Comparisons of venetoclax plus azacitidine with azacitidine (2-way propensity score) and venetoclax plus azacitidine with azacitidine and with LDAC (3-way propensity score) were conducted by propensity-score weighting on the individual patient data from both arms of VIALE-A and the LDAC arm of VIALE-C.

VIALE-A restricted recruitment to patients with a cytogenetic risk of intermediate or poor and excluded those who previously been treated with an HMA, whereas those patients were eligible for VIALE-C. For the purposes of the analysis, only patients who met the eligibility criteria for VIALE-A were included in the LDAC comparator arm for the main analysis. A sensitivity analysis included all patients in both studies.

Three efficacy outcomes were available for the propensity score–weighting analysis, OS, EFS, and CR + CRi. Weights were generated from a logistic regression model that included pre-specified demographic and clinical covariates anticipated to affect prognosis. The demographic characteristics were age, sex, race. The clinical characteristics were AML type, AML with myelodysplasia-related changes, prior MDS, bone marrow blasts, cytogenetic risk, and ECOG PS. All were dichotomized. These represented the important covariates and the dichotomization thresholds were the same as those accepted as meaningful in clinical practice. It is possible there is residual heterogeneity within the categories and residual confounding following adjustment, although the analysis was carried out on data from 2 closely related trials from the same sponsor on a similar population. There was no reported estimate of the potential risk of bias due to unmeasured confounders.

The comparisons were not randomized and the results were highly susceptible to bias due to imbalances in unmeasured confounders. Baseline comparisons were reported for dichotomized baseline covariates before and after adjustment for each end point. Weighting was generally good, with observed reduction of standardized differences. The weights themselves were not reported, and it was not indicated which methods were needed or applied to stabilize overly large or overly small weights. Effective sample size was calculated but not reported, also limiting appraisal of the weighting process. Relatively small numbers of patients were involved, particularly in the LDAC group, limiting the number of covariates that could be included in the model. Weighted statistical tests were used appropriately for the comparison of baseline covariates after adjustment; weighted Cox models were used for the calculation of time-to-event outcomes for OS and EFS. Log-log plots suggested a low risk of bias for the proportional hazards assumption. A subgroup analysis was conducted for the 2-way propensity-score analysis for both end points that was limited to patients with blast counts of 30% or greater.

The primary analysis used the subset of patients from VIALE-C who had no previous exposure to HMA as a comparison arm for the LDAC treatment arm (n = 50, compared with n = 285 for venetoclax plus azacitidine). The LDAC group had relatively few CR + CRi events, resulting in imprecise estimates for this outcome.

Summary

Seven trials met the systematic review inclusion criteria. With the additional restriction of the comparators for the NMA inclusion criteria, removing decitabine from the comparators, 4 trials were included in the ITC. Three analysis were conducted: 1 NMA and 2 propensity score–weighting analyses. One propensity score–weighting analysis compared venetoclax plus azacitidine (VIALE-A) with LDAC (VIALE-C), and the second compared venetoclax plus azacitidine and azacitidine alone (VIALE-A) with LDAC (VIALE-C). For the NMA, data were available for OS for 4 trials in a connected network and for CR + CRi for 3 trials. For the propensity score–weighting analysis, data were available for OS, EFS, and CR + CRi.

In the NMA, venetoclax plus azacitidine had a lower hazard of death compared with azacitidine, LDAC, and BSC, with no difference seen between venetoclax plus azacitidine and venetoclax plus LDAC. For CR + CRi, venetoclax plus azacitidine was favoured over azacitidine, LDAC, and BSC, with no treatment favoured between venetoclax plus azacitidine and venetoclax plus LDAC.

In both propensity score–weighting analyses, venetoclax plus azacitidine was favoured over LDAC for OS, EFS, and CR + CRi.

The systematic review was well conducted and documented. The search was limited to efficacy end points, and data were available only for OS and CR + CRi for the NMA, and OS, CR + CRi, and EFS for the propensity score–matched comparisons; no comparisons were conducted for transfusion independence, hospitalization, QoL end points, or safety. The NMA used appropriate methods to model survival, having assessed the risk of violation of the proportional hazards assumption. There was clinical heterogeneity in potential treatment-effect modifiers of blast count at baseline, prior treatment with an HMA, and cytogenetic risk. As the network was sparse, fixed-effects models had to be used, and there was no opportunity for baseline covariate adjustments. Due to the previously mentioned limitations, the comparative efficacy estimates may be biased, and it is not possible to quantify or identify the direction of the bias. In the propensity-score analyses, weighting was generally good, but the relatively small numbers of patients in the LDAC comparator group limited the number of covariates that could be included in the model. The comparisons were not randomized and the results were highly susceptible to bias due to imbalances in unmeasured confounders. Results of these ITCs must be interpreted with caution.

Discussion

Summary of Available Evidence

One double-blind, placebo-controlled phase III RCT and 1 ITC contributed evidence to this review. The objective of the RCT was to evaluate the efficacy and safety of venetoclax plus azacitidine in adults with newly diagnosed AML who were 18 years or older and ineligible for standard induction therapy due to age or comorbidities. The trial was restricted to patients who had not previously been treated with an HMA and who had intermediate or poor risk cytogenetics. Primary outcomes were OS and composite complete remission rate (CR + CRi). Secondary outcomes were CR, CR + CRh, rate of CR + CRi by the initiation of cycle 2, transfusion-independence rate, MRD response rate, response rates and OS in molecular subgroups, fatigue and GHS/QoL, and EFS.

A total of 431 patients were randomized in a 2:1 ratio, 286 to venetoclax plus azacitidine and 144 to placebo plus azacitidine, and included in the efficacy analysis. The most common reasons given for patients to be considered ineligible for standard induction therapy were age and ECOG PS. Patients were elderly, with poor performance, and markers of severe disease. The mean age was 75.4 years, with 60.6% aged 75 years or older. The majority were male (60.1%), and almost all were White or Asian. Most (75.2%) had de novo rather than secondary AML. Nearly 2-thirds had intermediate risk cytogenetics, 1-third had poor risk, and 1-half had 50% or greater bone marrow blasts at baseline.

The study was well conducted, with no clinically meaningful imbalance in baseline characteristics, minimal loss to follow-up, and the collection of end points were standardized and meaningful to patients. Multiplicity was controlled, with pre-specified strategies for testing of end points. The rate of study discontinuation was low and the assumptions surrounding missing data were conservative for most end points.

As RCTs were not available for all comparisons of interest, the sponsor supplied an ITC that included an NMA comparing venetoclax plus azacitidine and venetoclax plus LDAC with alternative treatments, and 2 propensity score–weighting comparisons of venetoclax plus azacitidine versus LDAC. Among the studies, 4 contributed to the NMA for OS, 3 contributed to the NMA for CR + CRi, and 2 contributed to the propensity score–weighting comparisons of OS, CR + CRi, and EFS. Safety end points were not included in the search, and data for other end points were not found.

Interpretation of Results

Efficacy

Venetoclax plus azacitidine improved most of the outcome measures that were identified as being of interest to clinicians and patients. Statistically significant treatment differences were seen for OS, EFS, measures of disease response (CR + CRi, CR + CRh, CR), and post-baseline transfusion independence. Statistically significant improvements were seen for OS and CR + CRi in the subgroup of patients with IDH1 or IDH2 mutations, and for CR + CRi for patients with FLT3 mutations. No statistically significant difference was detected in OS for patients with FLT3 mutations.

At a median duration of follow-up of 20.7 months for patients randomized to venetoclax plus azacitidine (versus 20.2 months for those randomized to placebo plus azacitidine), median OS for patients randomized to venetoclax plus azacitidine was 14.7 months compared with 9.6 months for azacitidine alone. The HR for mortality was 0.662 (95% CI, 0.518 to 0.845), and the stratified log-rank P value was less than 0.001. A similar magnitude of effect was seen for EFS. A greater proportion of patients randomized to venetoclax plus azacitidine had CR + CRi (65.3%) compared with those randomized to placebo plus azacitidine (25.3%), with a stratified P value of less than 0.001. Consistent results were seen for CR and early CR + CRi (after 2 cycles). For transfusion with red blood cells, 59.8% of patients randomized to venetoclax plus azacitidine were transfusion-independent compared with 35.2% of those randomized to placebo plus azacitidine, a treatment difference of 24.6% (95% CI, 15.0 to 34.2; stratified CMH P value < 0.001). For platelets, 68.5% of patients randomized to venetoclax plus azacitidine were transfusion-independent compared with 49.7% of those randomized to placebo plus azacitidine, a treatment difference of 18.9% (95% CI, 9.1% to 28.6%; stratified CMH P value < 0.001).

In the subgroup of patients with IDH1 or IDH2 mutation, the HR for mortality was 0.345 (95% CI, 0.199 to 0.598) and the risk difference for CR + CRi was 64.70% (95% CI, 48.9% to 80.4%); both differences were statistically significant. In the subgroup of patients with FLT3 mutation, the HR for mortality was 0.664 (95% CI, 0.351 to 1.257), and the risk difference for CR + CRi was 36.05% (95% CI, 10.2 to 61.9); only the risk difference was statistically significant, but the subgroup was small. Although other subgroup comparisons were not tested, the spread between estimates for OS and CR + CRi was widest for age (< 75 years and ≥ 75 years) and cytogenetic risk (intermediate versus poor; patients with good cytogenetic risk were excluded from the study).

Change from baseline in GHS/QoL as measured by the EORTC QLQ-C30 scale and fatigue as measured by the PROMIS 7a scale were secondary end points. While clinically meaningful differences were observed at individual end points, differences between treatment groups cannot be interpreted because the sequential testing strategy failed before this level. Interpretation of patient-reported outcome data is limited due to attrition over cycles.

In the NMA, the results favoured a lower hazard of death for patients assigned to venetoclax plus azacitidine compared with azacitidine alone (HR = 0.66; 95% CrI, 0.52 to 0.85), LDAC (HR = 0.57; 95% CrI, 0.40 to 0.81), and BSC (HR = 0.37; 95% CrI, 0.24 to 0.58), with no treatment favoured between venetoclax plus azacitidine and venetoclax plus LDAC (HR = 0.81; 95% CrI, 0.50 to 1.31). For CR + CRi, venetoclax plus azacitidine was favoured over azacitidine alone (OR = 5.05; 95% CrI, 3.30 to 7.87), LDAC (OR = 5.42; 95% CrI, 2.80 to 10.50), and BSC (OR = 61.55; 95% CrI, 8.23 to 1,881.53), with no treatment favoured between venetoclax plus azacitidine and venetoclax plus LDAC (OR = 0.86; 95% CrI, 0.30 to 2.35).

In the first propensity-score analysis, venetoclax plus azacitidine was favoured over LDAC for OS, EFS, and CR + CRi. In the second propensity-score analysis for OS, venetoclax plus azacitidine was favoured over LDAC and azacitidine alone. For EFS, venetoclax plus azacitidine was favoured over both azacitidine and LDAC, and azacitidine was favoured over LDAC. For CR + CRi, venetoclax plus azacitidine was favoured over both azacitidine and LDAC.

The search for the ITCs was limited to efficacy end points, and data were available only for OS and CR + CRi for the NMA, and OS, CR + CRi, and EFS for the propensity score–matched comparisons; no comparisons were conducted for transfusion independence, hospitalization, QoL end points, or safety. In the NMA, there were important differences in variables that were potentially treatment-effect modifiers between included studies. The small number of studies limited models to fixed effects and did not allow for meta-regression to adjust for baseline differences. Estimates are imprecise and at risk of bias. Estimates from propensity-score adjustment are at high risk of bias, and the small number of available patients limited the number of covariates that could be entered in the model for weighting. The comparisons were not randomized and the results were highly susceptible to bias due to imbalances in unmeasured confounders.

Harms

All patients in both groups experienced at least 1 adverse event, and almost all experienced at least 1 adverse event of grade 3 or greater. Compared with patients who received placebo plus azacitidine, a greater proportion of patients who received venetoclax plus azacitidine experienced 1 or more SAEs, 1 or more adverse events leading to dose discontinuation or interruption for venetoclax or placebo or azacitidine, or 1 or more adverse events leading to death. Common harms in all categories are generally predictable from the known mechanism of action for venetoclax and/or azacitidine and the underlying disease. Cytopenias were common, with neutropenia, febrile neutropenia, thrombocytopenia, and anemia represented across all categories, as were gastrointestinal adverse effects. Febrile neutropenia and infections contributed substantially to the most common SAEs and were the most frequent adverse events leading to death. The product monograph for venetoclax identifies the risk of serious infections leading to hospitalization or death in its section on serious warnings and precautions.

The same section specifies the need for dose ramp-up and prophylaxis for tumour lysis syndrome, and for dose reduction of the concurrent use of strong CYP3A inhibitors during dose ramp-up. Prophylaxis for tumour lysis syndrome was included in the study, and tumour lysis syndrome was uncommon, occurring in 2.5% of patients or less.

The ITC study did not include harms.

Conclusions

One double-blind, placebo-controlled phase III RCT (VIALE-A) and 1 ITC provided evidence supporting the efficacy and safety of venetoclax plus azacitidine in adult patients ineligible for standard induction chemotherapy due to age or comorbidities. Compared with azacitidine alone, patients treated with venetoclax (400 mg daily) and azacitidine (75 mg/m2 on days 1 through 7 of a 28-day cycle) showed benefits in the important clinical end points of OS, overall and early composite complete remission (CR and CRi), EFS, CR, and transfusion independence (red blood cell or platelet transfusions). All study participants reported treatment-emergent adverse events. For most categories of adverse events, there was an overall higher proportion of patients reporting these in the venetoclax plus azacitidine group. The most common adverse events were cytopenias and infections. No firm conclusion can be drawn between groups in GHS/QoL and fatigue, and patient attrition reduced the number of observation over the cycles, which limits the interpretation for these end points. Overall, the study was well conducted.

The VIALE-A study did not include a comparison between venetoclax plus azacitidine and current standards of care of induction therapy (in patients aged 75 years and older and fit), azacitidine monotherapy, LDAC, or BSC, or the alternative combination of venetoclax plus LDAC. In an ITC, venetoclax plus azacitidine was favoured over monotherapies and BSC, but no treatment was favoured for survival and composite complete remission between venetoclax plus azacitidine and venetoclax plus LDAC. No data are available for the comparison of venetoclax plus azacitidine with induction therapy. Results for 2 propensity-score comparisons between venetoclax plus azacitidine and azacitidine and LDAC were consistent. Small study and patient numbers and the potential for bias limit the reliability of the ITC, and the propensity-score comparisons were not randomized and therefore highly susceptible to bias.

References

1.Clinical Study Report [interim]: M15-656. A randomized, double-blind, placebo controlled phase 3 study of venetoclax in combination with azacitidine versus azacitidine in treatment naive subjects with acute myeloid leukemia who are ineligible for standard induction therapy [internal sponsor's report] North Chicago (IL): AbbVie; 2020 May 8.

2.Arber DA, Orazi A, Hasserjian R, et al. The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood. 2016;127(20):2391-2405. PubMed

3.Canadian Cancer Society. Acute myelogenous leukemia statistics 2021; https://www.cancer.ca/en/cancer-information/cancer-type/leukemia-acute-myelogenous-aml/statistics/. Accessed 2021 Apr 18.

4.Hodgson CS & Associaters. Blood cancer in Canada facts & stats 2016. Toronto (ON): Leukemia & Lymphoma Society of Canada; 2016: https://www.llscanada.org/sites/default/files/National/CANADA/Pdf/InfoBooklets/Blood_Cancer_in_Canada_Facts_%26_Stats_2016.pdf. Accessed 2021 Apr 18.

5.Canadian Cancer Society. Survival statistics for acute myelogenous leukemia. 2021; https://www.cancer.ca/en/cancer-information/cancer-type/leukemia-acute-myelogenous-aml/prognosis-and-survival/survival-statistics/. Accessed 2021 Apr 18.

6.Howlander N, Noone AM, Krapcho M, et al. SEER cancer statistics review, 1975-2014. 2018; https://seer.cancer.gov/csr/1975_2014/. Accessed 2021 Apr 18.

7.Alibhai SM, Leach M, Minden MD, Brandwein J. Outcomes and quality of care in acute myeloid leukemia over 40 years. Cancer. 2009;115(13):2903-2911. PubMed

8.Brandwein JM, Zhu N, Kumar R, et al. Treatment of older patients with acute myeloid leukemia (AML): revised Canadian consensus guidelines. Am J Blood Res. 2017;7(4):30-40. PubMed

9.Stubbins RJ, Lee L, Abou Mourad Y, et al. Older adults with acute myeloid leukemia in rural areas are less likely to receive azacitidine with worsened overall survival. Blood. 2018;132(Suppl 1):3989-3989.

10.Venclexta (venetoclax): 10 mg, 50 mg, and 100 mg tablets [product monograph]. St-Laurent (QC): AbbVie Corporation; 2020 Dec 3.

11.Vidaza (azacitidine): 100 mg injection [product monograph]. Mississauga (ON): Celgene Inc; 2018 Mar 22: https://pdf.hres.ca/dpd_pm/00044422.PDF. Accessed 2021 May 17.

12.Cytarabine: 100 mg/mL (100 mg/mL, 1 g/ 10 mL, 2 g/ 20 mL) injection [product monograph]. Kirkland (QC): Pfizer Canada ULC; 2020 Mar 13: https://www.pfizer.ca/sites/default/files/202006/Cytarabine_PM_E_13Mar2020_L3.pdf. Accessed 2021 Apr 18.

13.Ram R, Amit O, Zuckerman T, et al. Venetoclax in patients with acute myeloid leukemia refractory to hypomethylating agents-a multicenter historical prospective study. Ann Hematol. 2019;98(8):1927-1932. PubMed

14.Donnellan W, Xu T, Ma E, et al. Use of venetoclax (VEN) and hypomethylating agents (HMA) in newly diagnosed acute myeloid leukemia (AML) in the United States (US) - Real World (RW) response, treatment duration, dose and schedule modifications. Blood. 2020;136(Suppl 1):11-12.

15.Mozessohn L, Cheung MC, Fallahpour S, et al. Azacitidine in the 'real-world': an evaluation of 1101 higher-risk myelodysplastic syndrome/low blast count acute myeloid leukaemia patients in Ontario, Canada. Br J Haematol. 2018;181(6):803-815. PubMed

16.Arslan S, Zhang J, Dhakal P, et al. Outcomes of therapy with venetoclax combined with a hypomethylating agent in favorable-risk acute myeloid leukemia. Am J Hematol. 2021;96(3):E59-E63. PubMed

17.Tadros M, Koehne G, Zahra T, Unzaga J, Shwin M, Chow N. Evaluating acute myeloid leukemia induction regimens in elderly patients with unfavorable risk cytogenetics that are candidates for intensive remission therapy. Blood. 2020;136(Suppl 1):17.

18.Pollyea DA, Winters A, Jordan CT, Smith C, Gutman JA. Allogeneic transplant improves AML outcomes compared to maintenance venetoclax and azacitidine following response to initial venetoclax and azacitidine therapy. Blood. 2020;136(Suppl 1):24-24.

19.Kennedy VE, Hui G, Gaut D, et al. Hypomethylating agents in combination with venetoclax as a bridge to allogeneic transplant in acute myeloid leukemia. Blood. 2020;136(Suppl 1):32-33.

20.Salhotra A, Ngo D, Zhang J, et al. Clinical outcomes of patients with secondary acute myeloid leukemia (sAML) treated with hypomethylating agent plus venetoclax (HMA-Ven) or liposomal daunorubicin cytarabine (CPX-351) [abstract]. Presented at: 62nd ASH Annual Meeting and Exposition. Dec 5-8, 2020. 2020: https://ash.confex.com/ash/2020/webprogram/Paper140538.html. Accessed 2021 May 14.

21.McGowan J, Sampson M, Salzwedel DM, Cogo E, Foerster V, Lefebvre C. PRESS Peer Review of Electronic Search Strategies: 2015 guideline statement. J Clin Epidemiol. 2016;75:40-46. PubMed

22.Grey matters: a practical tool for searching health-related grey literature. Ottawa (ON): CADTH; 2019: https://www.cadth.ca/grey-matters. Accessed 2021 Feb 11.

23.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. PubMed

24.Giesinger JM, Kieffer JM, Fayers PM, et al. Replication and validation of higher order models demonstrated that a summary score for the EORTC QLQ-C30 is robust. J Clin Epidemiol. 2016;69:79-88. PubMed

25.EORTC QLC-C30 scoring manual. Brussels (BE): EORTC; 2001: https://www.eortc.org/app/uploads/sites/2/2018/02/SCmanual.pdf. Accessed 2021 Mar 8.

26.Luo N, Fones CS, Lim SE, Xie F, Thumboo J, Li SC. The European Organization for Research and Treatment of Cancer Quality of Life Questionnaire (EORTC QLQ-c30): validation of English version in Singapore. Qual Life Res. 2005;14(4):1181-1186. PubMed

27.Davda J, Kibet H, Achieng E, Atundo L, Komen T. Assessing the acceptability, reliability, and validity of the EORTC Quality of Life Questionnaire (QLQ-C30) in Kenyan cancer patients: a cross-sectional study. J Patient Rep Outcomes. 2021;5(1):4. PubMed

28.Osoba D, Rodrigues G, Myles J, Zee B, Pater J. Interpreting the significance of changes in health-related quality-of-life scores. J Clin Oncol. 1998;16(1):139-144. PubMed

29.Snyder CF, Blackford AL, Sussman J, et al. Identifying changes in scores on the EORTC-QLQ-C30 representing a change in patients' supportive care needs. Qual Life Res. 2015;24(5):1207-1216. PubMed

30.Bedard G, Zeng L, Zhang L, et al. Minimal important differences in the EORTC QLQ-C30 in patients with advanced cancer. Asia Pac J Clin Oncol. 2014;10(2):109-117. PubMed

31.Ameringer S, Elswick Jr RK, Menzies V, et al. Psychometric evaluation of the PROMIS Fatigue-short form across diverse populations. Nurs Res. 2016;65(4):279. PubMed

32.Brooks R. EuroQol: the current state of play. Health Policy. 1996;37(1):53-72. PubMed

33.EuroQol Group. EuroQol--a new facility for the measurement of health-related quality of life. Health Policy. 1990;16(3):199-208. PubMed

34.Teckle P, Peacock S, McTaggart-Cowan H, et al. The ability of cancer-specific and generic preference-based instruments to discriminate across clinical and self-reported measures of cancer severities. Health Qual Life Outcomes. 2011;9:106. PubMed

35.Estey E, Othus M, Lee SJ, Appelbaum FR, Gale RP. New drug approvals in acute myeloid leukemia: what's the best end point? Leukemia. 2016;30(3):521-525. PubMed

36.Wolach O, Levi I, Canaani J, et al. First results from a nationwide prospective non-interventional study of venetoclax-based 1st line therapies in patients with acute myeloid leukemia (AML) - revive study. Blood. 2020;136(Suppl 1):27-28.

37.Garcia-Horton A, Maze D, McNamara CJ, Sibai H, Gupta V, Murphy T. Azacitidine and venetoclax for the treatment of accelerated and blast phase myeloproliferative neoplasms and chronic myelomonocytic leukemia: a case series. Leuk Lymphoma. 2021:1-6. PubMed

38.AbbVie Corporation response to Mar 8, 2021 to DRR request for additional information regarding Venclexta (venetoclax) DRR review: request for report of systematic review informing the ITC [internal additional sponsor's information]. Pointe-Claire (QC): AbbVie Corporation; 2021 Mar 15.

39.Study report: indirect comparisons of venetoclax combinations and other treatments in treatment naïve patients with acute myeloid leukemia (AML) who are ineligible for induction chemotherapy. In: Drug Reimbursement Review sponsor submission: Venclexta (venetoclax) in combination with azacitidine for the treatment of acute myeloid leukemia (AML), 10 mg, 50 mg, and 100 mg tablets Pointe-Claire (QC): AbbVie Corporation; 2021 Jan 8.

40.Braun DP, Gupta D, Grutsch JF, Staren ED. Can changes in health related quality of life scores predict survival in stages III and IV colorectal cancer? Health Qual Life Outcomes. 2011;9:62. PubMed

41.Clinical Study Report: M16-043. A randomized, double-blind, placebo controlled phase 3 study of venetoclax co-cdministered with low dose cytarabine versus low dose cytarabine in treatment-naive patients with acute myeloid leukemia who are ineligible for intensive chemotherapy [internal sponsor's report]. North Chicago (IL): AbbVie; 2020 Mar 20.

42.EQRTC. EORTC Quality of life: FAQs. 2021; https://qol.eortc.org/faq/. Accessed 2021 May 17.

43.Sinnott PL, Joyce VR, Barnett PG. Guidebook: preference measurement in economic analysis. Menlo Park (CA): Health Economics Research Center; 2007: https://www.herc.research.va.gov/files/BOOK_419.pdf. Accessed 2021 May 14.

Appendix 1: Literature Search Strategy

Note that this appendix has not been copy-edited.

Clinical Literature Search

Overview

Interface: Ovid

Databases:

Date of search: February 11, 2021

Alerts: Weekly search updates until project completion

Study types: No filters were applied to limit the retrieval by study type

Limits:

Table 38: Syntax Guide

Syntax

Description

/

At the end of a phrase, searches the phrase as a subject heading

MeSH

Medical Subject Heading

exp

Explode a subject heading

*

Before a word, indicates that the marked subject heading is a primary topic;

or, after a word, a truncation symbol (wildcard) to retrieve plurals or varying endings

#

Truncation symbol for 1 character

adj#

Requires terms to be adjacent to each other within # number of words (in any order)

.ti

Title

.ot

Original title

.ab

Abstract

.hw

Heading word; usually includes subject headings and controlled vocabulary

.kf

Author keyword heading word (MEDLINE)

.kw

Author keyword (Embase)

.dq

Candidate term word (Embase)

.pt

Publication type

.rn

Registry number

.nm

Name of substance word (MEDLINE)

medall

Ovid database code: MEDLINE All, 1946 to present, updated daily

oemezd

Ovid database code; Embase, 1974 to present, updated daily

Multi-Database Strategy

Search Strategy

  1. (venetoclax* or Venclexta* or Venclyxto* or ABT199 or ABT-199 or GDC0199 or GDC-0199 or RG7601 or RG-7601 or N54AIC43PW).ti,ab,kf,ot,hw,nm,rn.

  2. exp Leukemia, Myeloid, Acute/

  3. (AML or ANLL).ti,ab,kf.

  4. (Acute adj5 (granulocytic* or myeloblastic* or myelocytic* or myelogenous* or myeloid* or nonlymphoblastic* or non-lymphoblastic* or nonlymphocytic* or non-lymphocytic* or basophilic* or eosinophilic* or erythroblastic* or megakaryoblastic* or monocytic* or megakaryocytic* or myelomonocytic*) adj5 (leukemia* or leukemia*)).ti,ab,kf.

  5. (erythroleukemia* or erythroleukemia*).ti,ab,kf.

  6. ((mast-cell or promyelocytic*) adj3 (leukemia* or leukemia*)).ti,ab,kf.

  7. or/2-6

  8. 1 and 7

  9. 8 use medall

  10. *venetoclax/ or (venetoclax* or Venclexta* or Venclyxto* or ABT199 or ABT-199 or GDC0199 or GDC-0199 or RG7601 or RG-7601).ti,ab,kw,dq.

  11. exp Acute myeloid leukemia/

  12. (AML or ANLL).ti,ab,kw,dq.

  13. (Acute adj5 (granulocytic* or myeloblastic* or myelocytic* or myelogenous* or myeloid* or nonlymphoblastic* or non-lymphoblastic* or nonlymphocytic* or non-lymphocytic* or basophilic* or eosinophilic* or erythroblastic* or megakaryoblastic* or monocytic* or megakaryocytic* or myelomonocytic*) adj5 (leukemia* or leukemia*)).ti,ab,kw,dq.

  14. (erythroleukemia* or erythroleukemia*).ti,ab,kw,dq.

  15. ((mast-cell or promyelocytic*) adj3 (leukemia* or leukemia*)).ti,ab,kw,dq.

  16. or/11-15

  17. 10 and 16

  18. 17 use oemezd

  19. 18 not (conference review or conference abstract).pt.

  20. 9 or 19

  21. remove duplicates from 20

Clinical Trials Registries

ClinicalTrials.gov

Produced by the US National Library of Medicine. Targeted search used to capture registered clinical trials.

[Search terms – Venclexta (venetoclax), acute myeloid leukemia]

WHO ICTRP

International Clinical Trials Registry Platform, produced by WHO. Targeted search used to capture registered clinical trials.

[Search terms – Venclexta (venetoclax), acute myeloid leukemia]

Health Canada’s Clinical Trials Database

Produced by Health Canada. Targeted search used to capture registered clinical trials.

[Search terms – Venclexta (venetoclax), acute myeloid leukemia]

EU Clinical Trials Register

European Union Clinical Trials Register, produced by the European Union. Targeted search used to capture registered clinical trials.

[Search terms – Venclexta (venetoclax), acute myeloid leukemia]

Grey Literature

Search dates: February 8-22, 2021

Keywords: Venclexta (venetoclax), acute myeloid leukemia

Limits:

Updated: Publication years: none

Search updated before the completion of stakeholder feedback period

Relevant websites from the following sections of the CADTH grey literature checklist Grey Matters: A Practical Tool For Searching Health-Related Grey Literature (https://www.cadth.ca/grey-matters) were searched:

Appendix 2: Excluded Studies

No studies were excluded on full-text review.

Appendix 3: Detailed Outcome Data

Note that this appendix has not been copy-edited.

Table 39: Results for OS and Composite Complete Remission (CR + CRi) in Subgroups, IA2

Result

VEN + AZA

PBO + AZA

OS in subgroups

IDH1 and/or IDH2 mutation

N

61

28

Median OS, months (95% CI)

NE (12.1 to NE)

6.2 (2.3 to 12.7)

HR (unstratified Cox model) (95% CI)

0.345 (0.199 to 0.598)

P value (unstratified log-rank test)

< 0.0001*

FLT3 mutation

N

29

22

Median OS, months (95% CI)

12.7 (7.3 to 23.5)

8.6 (5.9 to 14.7)

HR (unstratified Cox model) (95% CI)

0.664 (0.351 to 1.257)

P value (unstratified log-rank test)

0.2054

NPM1 mutation

N

27

17

Median OS, months (95% CI)

15.0 (3.4 to NE)

13.0 (4.2 to 20.3)

HR (unstratified Cox model) (95% CI)

0.734 (0.357 to 1.505)

TP53 mutation

N

38

14

Median OS, months (95% CI)

5.8 (2.6 to 8.3)

5.4 (1.3 to 9.3)

HR (unstratified Cox model) (95% CI)

0.760 (0.398 to 1.450)

Age

< 75 years

N

112

58

HR (unstratified Cox model) (95% CI)

0.888 (0.591 to 1.333)

≥ 75 years,

N

174

87

HR (unstratified Cox model) (95% CI)

0.535 (0.394 to 0.727)

ECOG at baseline

ECOG < 2

N

157

81

HR (unstratified Cox model) (95% CI)

0.607 (0.440 to 0.838)

ECOG ≥ 2

N

129

64

HR (unstratified Cox model) (95% CI)

0.704 (0.483 to 1.027)

Cytogenetic risk

Intermediate

182

89

N

0.566 (0.407 to 0.786)

HR (unstratified Cox model) (95% CI)

Poor

104

56

N

0.775 (0.538 to 1.117)

HR (unstratified Cox model) (95% CI)

Type of AML

De novo

N

214

110

HR (unstratified Cox model) (95% CI)

0.674 (0.508 to 0.895)

Secondary

N

72

35

HR (unstratified Cox model) (95% CI)

0.561 (0.346 to 0.910)

AML with myelodysplasia-related changes

Yes

N

92

49

HR (unstratified Cox model) (95% CI)

0.732 (0.484 to 1.107)

No

N

194

96

HR (unstratified Cox model) (95% CI)

0.616 (0.455 to 0.834)

Bone marrow blast count

< 30%

N

85

41

HR (unstratified Cox model) (95% CI)

0.716 (0.447 to 1.148)

≥ 30% to < 50%

N

61

33

HR (unstratified Cox model) (95% CI)

0.567 (0.339 to 0.949)

≥ 50%

N

140

71

HR (unstratified Cox model) (95% CI)

0.633 (0.448 to 0.893)

CR + CRi in subgroups

IDH1 and/or IDH2 mutation

N

61

28

CR, n (%; 95% CI)

26 (42.6; 30.0 to 55.9)

1 (3.6; 0.1 to 18.3)

CR + CRi, n (%; 95% CI)

46 (75.4; 62.7 to 85.5)

3 (10.7; 2.3 to 28.2)

Risk difference % (95% CI)

64.70 (48.9 to 80.4)

P value (Fisher's exact test)

< 0.001*

FLT3 mutation

N

29

22

CR, n (%; 95% CI)

10 (34.5; 17.9 to 54.3)

3 (13.6; 2.9 to 34.9)

CR + CRi, n (%; 95% CI)

21 (72.4; 52.8 to 87.3)

8 (36.4; 17.2 to 59.3)

Risk difference (%; 95% CI)

36.05 (10.2 to 61.9)

P value (Fisher's exact test)

0.021*

NPM1 mutation

N

27

17

CR, n (%; 95% CI)

12 (44.4; 25.5 to 64.7)

3 (17.6; 3.8 to 43.4)

CR + CRi, n (%; 95% CI)

18 (66.7; 46.0 to 83.5)

4 (23.5; 6.8 to 49.9)

Risk difference (%; 95% CI)

TP53 mutation

N

38

14

CR, n (%; 95% CI)

9 (23.7; 11.4 to 40.2)

0

CR + CRi, n (%; 95% CI)

21 (55.3; 38.3 to 71.4)

0

Risk difference (%; 95% CI)

Age

< 75 years

n/N (%)

70/112 (62.5)

24/58 (41.1)

Risk difference (95% CI)

21.12 (5.6 to 36.6)

≥ 75 years,

n/N (%)

120/174

17/87

Risk difference (95% CI)

49.43 (38.6 to 60.2)

ECOG at baseline

ECOG < 2

n/N (%)

108/157 (68.8)

20/81 (24.7)

Risk difference (95% CI)

44.10 (32.2 to 56.0)

ECOG ≥ 2

n/N (%)

82/129 (63.6)

21/64 (32.8)

Risk difference (95% CI)

30.75 (16.6 to 44.9)

Cytogenetic risk

Intermediate

n/N (%)

135/182 (74.2)

28/89 (31.5)

Risk difference (95% CI)

42.72 (31.2 to 54.3)

Poor

n/N (%)

55/104 (52.9)

13/56 (23.2)

Risk difference (95% CI)

29.67 (15.0 to 44.3)

Type of AML

De novo

n/N (%)

142/214 (66.4)

33/110 (30.0)

Risk difference (95% CI)

36.36 (25.7 to 47.0)

Secondary

n/N (%)

48/72 (66.7)

8/35 (22.9)

Risk difference (95% CI)

43.81 (26.1 to 61.5)

AML with myelodysplasia-related changes

Yes

n/N (%)

56/92 (60.9)

11/49 (22.4)

Risk difference (95% CI)

38.42 (23.1 to 53.8)

No

n/N (%)

134/194 (69.1)

30/96 (31.3)

Risk difference (95% CI)

37.82 (26.5 to 49.1)

Bone marrow blast count

< 30%

n/N (%)

65/85 (76.5)

16/41 (39.0)

Risk difference (95% CI)

37.45 (20.0 to 54.9)

≥ 30% to < 50%

n/N (%)

35/61 (57.4)

9/33 (27.3)

Risk difference (95% CI)

30.10 (10.5 to 49.7)

≥ 50%

n/N (%)

90/140 (64.3)

16/71 (22.5)

Risk difference (95% CI)

41.75 (29.2 to 54.3)

AML = acute myeloid leukemia; CI = confidence interval; CR = complete remission; CRi = complete remission with incomplete blood count recovery; FLT3 = FMS-like tyrosine kinase 3; HR = hazard ratio; IA2 = second interim analysis; IDH = isocitrate dehydrogenase; NPM1 = nucleophosmin 1; OS = overall survival; PBO + AZA = placebo plus azacitidine; SD = standard deviation; TP53 = tumour protein p53; VEN + AZA = venetoclax plus azacitidine.

*Statistically significant under the preplanned testing strategy.

Note: Data cut-off was January 4, 2020.

Source: Clinical Study Report.1

Table 40: Patient-Reported Outcomes, Efficacy Population, to Cycle 13

Visit

Treatment

N

Mean

Change from baseline

LS Mean (95% CI)

Between groups

LS mean diff (95% CI)

P value

PROMIS 7a

PROMIS SF 7a treatment effect

0.758†

PROMS SF 7a treatment by visit interaction

0.264†

Baseline

VEN + AZA

264

53.86

PBO + AZA

132

54.97

Cycle 3 day 1

VEN + AZA

174

52.61

−0.167 (−1.55 to 1.21)

0.294 (−2.09 to 2.67)

PBO + AZA

79

54.43

−0.461 (−2.47 to 1.55)

Cycle 5 day 1

VEN + AZA

143

51.97

−3.036 (−4.51 to −1.56)

−2.24 (−4.89 to 0.41)

PBO + AZA

54

53.59

−0.796 (−3.06 to 1.47)

Cycle 7 day 1

VEN + AZA

110

51.92

−2.263 (−3.86 to −0.67)

−0.286 (−3.17 to 2.59)

PBO + AZA

43

53.55

−1.976 (−4.43 to 0.48)

Cycle 9 day 1

VEN + AZA

91

51.59

−3.377 (−5.07, −1.68)

−2.387 (−5.43 to 0.65)

PBO + AZA

37

54.8

−0.99 (−3.57 to 1.59)

Cycle 11 day 1

VEN + AZA

77

52.15

−2.209 (−3.99 to −0.43)

−0.464 (−3.84 to 2.91)

PBO + AZA

26

54.98

−1.745 (−4.66 to 1.17)

Cycle 13 day 1

VEN + AZA

72

52.6

−1.644 (−3.46 to 0.18)

−0.191 (−3.79 to 3.41)

PBO + AZA

21

55.04

−1.453 (−4.60 to 1.69)

EORTC QLQ-C30 GHS

EORTC QLQ-C30 GHS/QoL Treatment effect

0.246†

EORTC QLQ-C30 GHS/QoL Treatment by visit interaction

0.397†

Baseline

VEN + AZA

262

52.61

PBO + AZA

130

55.96

Cycle 3 day 1

VEN + AZA

172

56.83

7.073 (4.09 to 10.05)

1.413 (−3.74 to 6.57)

PBO + AZA

77

58.98

5.66 (1.32 to 10.00)

Cycle 5 day 1

VEN + AZA

141

58.04

10.011 (6.82 to 13.20)

5.092 (−0.72 to 10.90)

PBO + AZA

53

61.95

4.918 (−0.06 to 9.90)

Cycle 7 day 1

VEN + AZA

109

57.03

7.843 (4.36 to 11.33)

2.059 (−4.27 to 8.38)

PBO + AZA

43

61.24

5.785 (0.40 to 11.17)

Cycle 9 day 1

VEN + AZA

90

57.13

12.26 (8.53 to 15.99)

4.87 (−1.84 to 11.58)

PBO + AZA

37

61.49

7.39 (1.71 to 13.07)

Cycle 11 day 1

VEN + AZA

77

58.23

10.034 (6.10 to 13.96)

4.121 (−3.37 to 11.61)

PBO + AZA

26

60.58

5.912 (−0.56 to 12.38)

Cycle 13, day 1

VEN + AZA

72

55.09

8.833 (4.81 to 12.85)

0.76 (−7.26 to 8.78)

PBO + AZA

21

59.52

8.073 (1.05 to 15.10)

EQ-5D Health Index Score

Baseline

VEN + AZA

260

0.76

PBO + AZA

130

0.74

Cycle 3, day 1

VEN + AZA

170

0.78

0.017 (−0.01 to 0.04)

−0.006 (−0.05 to 0.03)

PBO + AZA

77

0.76

0.023 (−0.01 to 0.06)

Cycle 5, day 1

VEN + AZA

139

0.79

0.052 (0.03 to 0.08)

0.024 (−0.02 to 0.07)

PBO + AZA

53

0.77

0.028 (−0.01 to 0.07)

Cycle 7, day 1

VEN + AZA

106

0.79

0.035 (0.01 to 0.06)

0.019 (−0.03 to 0.07)

PBO + AZA

43

0.75

0.017 (−0.03 to 0.06)

Cycle 9, day 1

VEN + AZA

89

0.78

0.049 (0.02 to 0.08)

0.031 (−0.02 to 0.08)

PBO + AZA

37

0.75

0.018 (−0.03 to 0.06)

Cycle 11, day 1

VEN + AZA

77

0.77

0.031 (0.00 to 0.06)

−0.009 (−0.07 to 0.05)

PBO + AZA

26

0.75

0.039 (−0.01 to 0.09)

Cycle 13, day 1

VEN + AZA

72

0.76

0.016 (−0.02 to 0.05)

−0.026 (−0.09 to 0.04)

PBO + AZA

21

0.70

0.042 (−0.01 to 0.09)

EQ-5D VAS Score

Baseline

VEN + AZA

260

64.27

PBO + AZA

130

60.29

Cycle 3, day 1

VEN + AZA

170

0.78

3.363 (0.54 to 6.19)

2.539 (−2.33 to 7.41)

PBO + AZA

77

0.76

0.825 (−3.27 to 4.92)

Cycle 5, day 1

VEN + AZA

139

0.79

6.392 (3.38 to 9.40)

3.085 (−2.33 to 8.50)

PBO + AZA

53

0.77

3.308 (−1.32 to 7.93)

Cycle 7, day 1

VEN + AZA

106

0.79

4.933 (1.67 to 8.19)

0.779 (−5.07 to 6.63)

PBO + AZA

43

0.75

4.154 (−0.82 to 9.12)

Cycle 9, day 1

VEN + AZA

89

0.78

9.027 (5.58 to 12.48)

5.218 (−0.94 to 11.38)

PBO + AZA

37

0.75

3.810 (−1.40 to 9.02)

Cycle 11, day 1

VEN + AZA

77

0.77

5.688 (2.08 to 9.29)

3.283 (−3.52 to 10.09)

PBO + AZA

26

0.75

2.405 (−3.46 to 8.27)

Cycle 13, day 1

VEN + AZA

72

0.76

5.308 (1.63 to 8.99)

−4.860 (−12.11 to 2.39)

PBO + AZA

21

0.70

10.168 (3.83 to 16.50)

CI = confidence interval; EORTC QLQ-C30 = European Organization for Research and Treatment of Cancer Quality of Life Questionnaire in Cancer; EQ-5D-5L = EuroQol 5 Dimensions 5 Levels Health State Instrument; LS = least squares; PBO + AZA = placebo plus azacitidine; PROMIS 7a = Patient-Reported Outcomes Measurement System Cancer Fatigue SF 7a; VEN + AZA = venetoclax plus azacitidine.

†Nominal P values. Sequential testing failed at prior end point on the sequence and there is a risk increased type I error if conclusions were drawn from those results.

Data cut-off: January 4; 2020.

Source: Clinical Study Report1.

Appendix 4: Description and Appraisal of Outcome Measures

Note that this appendix has not been copy-edited.

Aim

To describe the following outcome measures and review their measurement properties (validity, reliability, responsiveness to change, and MID):

Findings

Table 41: Summary of Outcome Measures and Their Measurement Properties

Outcome measure

Type

Conclusions about measurement properties

MID

European Organization for Research and Treatment of Cancer Quality of Life Questionnaire Core 30 (EORTC QLQ-C30)40

Cancer-specific measure of HRQoL

30-item questionnaire, consisting of 4 scales; 4-item response scale: Function Scale, Symptoms Scale, Single-Item Symptom Scale,

7-item Likert scale: Global QoL Scale/GHS

Validity: Construct validity assessed through convergent and discriminative approach

Reliability: Internal consistency assessed using Cronbach alpha

Responsiveness: No relevant studies found

10 points change for the individual items and scale scores.28,29

EuroQol 5 Dimensions 5 Levels Health State Instrument (EQ-5D-5L)32,33

Patient-reported, generic quality of life instrument

Validity: Less responsive than disease-specific measures.

Moderate to poor ability to distinguish between cancer severity by 3 scales:

  • self-reported health status (effect size 0.90)

  • ECOG PS (effect size = 0.31)

  • stage of cancer (effect size = 0.06)

Reliability: Five functioning scales and global health status demonstrated acceptable consistency with Cronbach alpha ranging from 0.77 to 0.82.

  • r = 0.43 between EORTC QLQ-C30 and EQ-5D

  • r = 0.73 between EORTC QLQ-C30 and EQ VAS

  • r = 0.43 between EQ-5D and EQ VAS

Responsiveness: Uncertain in populations with colorectal cancer.

No relevant studies found for patients with AML.

Patient-Reported Outcome Measurement Information System (PROMIS) Cancer Fatigue Short Form v1.0 Fatigue 7a (PROMIS F-SF)31

7-item, patient-reported, tool that measure both the experience of fatigue and the interference of fatigue on daily activities over the past week, using 5-point Likert scales from 1 = never to 5 = always

Validity:

  • Concurrent validity exanimated through Pearson’s correlations between scores from the PROMIS F-SF, the Multidimensional Fatigue Symptom Inventory-Short Form (MFSI-SF), and the Brief Fatigue Inventory (BFI).

  • Discriminant validity evaluated by examining Pearson’s correlations between scores on the PROMIS F-SF and measures of stress and depressive symptoms.

  • Known-groups validity assessed by comparing PROMIS F-SH scores in the clinical samples to healthy controls.31

Reliability: Internal consistency assessed using Cronbach alpha.31

Responsiveness: No relevant studies found.

No relevant studies found.

3 points validated and reported in the VIALE-A and VIALE-C trials (AML patients).41

ECOG PS = Eastern Cooperative Oncology Group Performance Status; EORTC QLQ-C30 = European Organization for Research and Treatment of Cancer Quality of Life Questionnaire Core 30; GHS = global health status; HRQoL = health-related quality of life; EQ-5D-5L = European Quality of Life 5-Five Dimensions 5-Levels; MID = minimal important difference; PedsQL-Core = Pediatric Quality of Life-Core Module; PROMIS = Patient-Reported Outcome Measurement Information System.

EORTC QLQ-C30

Description

The European Organization for Research and Treatment of Cancer Quality of Life Questionnaire Core 30 (EORTC QLQ-C30), is 1 of the most commonly used patient-reported outcome measures in oncology clinical trials.24 It is a multidimensional, cancer-specific, evaluative measure of HRQoL. It was designed specifically for the purpose of assessing changes in participants’ HRQoL in clinical trials, in response to treatment.42 The core questionnaire of the EORTC QLQ-C30 consists of 30 questions that are scored to create 5 multi-item functional scales, 3 multi-item symptom scales, 6 single-item symptom scales, and a 2-item QoL scale, as outlined in Table 42. Version 3.0 of the questionnaire, used in the included trials in this report, is the most current version and has been in use since December of 1997.25 It is available in 90 different languages and is intended for use in adult populations only. Notably, the global QoL scale is also known as the GHS, which was reported in the trial mentioned previously.27

Table 42: Scales of EORTC QLQ-C30

Functional scales

(15 questions)

Symptom scales

(7 questions)

Single-item symptom scales

(6 questions)

Global quality of life

(2 questions)

Physical function (5)

Fatigue (3)

Dyspnea (1)

Global Quality of Life (2)

Role function (2)

Pain (2)

Insomnia (1)

Cognitive function (2)

Nausea and vomiting (2)

Appetite loss (1)

Emotional function (4)

Constipation (1)

Social function (2)

Diarrhea (1)

Financial impact (1)

Scoring

The EORTC QLQ-C30 uses a 1-week recall period in assessing function and symptoms. Most questions have 4 response options (“not at all,” “a little,” “quite a bit,” “very much”), with scores on these items ranging from 1 to 4.25 For the 2 items that form the global QoL scale, however, the response format is a 7-point Likert-type scale, with anchors between 1 (very poor) and 7 (excellent).25

Raw scores for each scale are computed as the average of the items that contribute to a particular scale. This scaling approach is based upon the assumption that it is appropriate to provide equal weighting to each item that comprises a scale. There is also an assumption that, for each item, the interval between response options is equal (for example, the difference in score between “not at all” and “a little” is the same as “a little” and “quite a bit,” at a value of 1 unit). Each raw scale score is converted to a standardized score that ranges from 0 to 100 using a linear transformation, with a higher score reflecting better function on the function scales, higher symptoms on the symptom scales, and better QoL (i.e., higher scores simply reflect higher levels of response on that scale). Thus, a decline in score on the symptom scale would reflect an improvement, whereas an increase in score on the function and QoL scale would reflect an improvement. According to the EORTC QLQ-C30s scoring algorithm, if there are missing items for a scale (i.e., the participant did not provide a response), the score for the scale can still be computed if there are responses for at least 1-half of the items. In calculating the scale score, the missing items are simply ignored — an approach that assumes that the missing items have values equal to the average of those items for what the respondent completed.25

Psychometric Properties

Validity: One cross-sectional study aimed to validate the EORTC QLQ-30 in a convenience sample of 57 cancer patients in Singapore.26 Most patients had breast and colorectal cancer, but leukemia, lung cancer, lymphoma, germ cell tumour, and other cancers were also reported. Construct validity was assessed by cross-sectional correlational evidence and discriminative evidence. First, convergent validity was assessed using spearman’s correlations between QLQ-30 and Short Form-36 (SF-36) scales, hypothesizing moderate to strong correlation (defined as correlation coefficient of 0.35 to 0.5, and > 0.5, respectively) between scales of these 2 instruments measuring similar dimensions of HRQoL. Results showed moderate to strong correlations between QLC-30 and SF-36 scales, ranging from 0.35 to 0.67 across the assessed scales. Next, the known-groups approach was used to compare 6 QLQ-30 scale scores between patients reporting mild and severe symptoms, as well as by stage of disease and presence of comorbid conditions. With the exception of emotional functioning, the remaining 5 scales showed better scores in patients with mild symptoms than those with severe symptoms (P < 0.05 for all other comparisons). Patients in early stages of cancer (or with no comorbid conditions) generally had better QLQ-30 scores than those in advanced disease stages (or with comorbid conditions); however, none of these differences was statistically significant.26

A recent cross-sectional study in Kenya was conducted to evaluate the psychometric properties of the EORTC QLQ-C30, using the English or Kiswahili version in 100 patients with cancer.27 Most patients had breast cancer, followed by prostate, Kaposi sarcoma, lung, and other cancers. Construct validity was assessed by examining the inter-scale correlations among the subscales of EORTC QLQ-C 30. The inter-scale correlations were weak to strong; absolute magnitude ranged from 0.07 to 0.73. Notably, with the exception of the cognitive functioning, emotional functioning, nausea and vomiting, dyspnea, appetite loss, constipation, and diarrhea domains, the GHS correlated moderately with the remaining subscales (r ≥ 0.30). Cross-cultural validity was evaluated but not reported here as not relevant.27

Reliability: The Singaporean cross-sectional study above also assessed internal consistency reliability by calculating Cronbach alpha for all QLQ-C30 scales. Cronbach alpha was 0.70 or greater for 6 of the 9 assessed QLQ-30 scales; cognitive functioning, physical functioning, and nausea and vomiting had a Cronbach alpha ranging from 0.19 to 0.68.26

The Kenyan study described above assessed the internal consistency of each scale of the questionnaire using Cronbach alpha coefficients. With the exception of the cognitive function scale, all of the scales had a Cronbach alpha of 0.70 or greater.27

Studies evaluating the responsiveness of the instrument was not found.

Minimal Important Difference

For use in clinical trials, scores on the EORTC QLQ-C30 can be compared between different groups of patients or within a group of patients over time. One study conducted in breast cancer and small-cell lung cancer patients in 1998 estimated a clinically relevant change in score on any scale of the EORTC QLQ-C30 to be 10 points.28 The estimate was based on a study that used an anchor-based approach to estimating the MID in which patients who reported “a little” change (for better or worse) on the subjective significance questionnaire had corresponding changes on a function or symptom scale of the EORTC QLQ-C30 of approximately 5 to 10 points. Participants who reported a “moderate” change had corresponding changes in the EORTC QLQ-C30 of about 10 to 20, and those who reported being “very much” changed had corresponding changes of more than 20.28

More recently in 2015, a Canadian study estimated the MIDs of EORTC QLQ-C30 scales using data from 193 newly diagnosed breast and colorectal cancer patients.29 The Supportive Care Needs Survey-Short Form-34 (SCNS-SF34) was used as an anchor; mean changes in EORTC QLQ-C30 scales associated with improvement, worsening, and no-change in supportive care based on the SCNS-SF34 was then calculated. MIDs were assessed for the following scales: Physical function, role function, emotional function, global health/QoL (i.e., GHS), pain, and fatigue. For improvement, MIDs associated with a statistically significantly improved supportive care needs ranged from 10 to 32 points. For worsening, MIDs associated with a statistically significantly worsening of supportive care needs ranged from 9 to 21 points. The range for unchanged supportive care needs was from 1-point worsening to 16-point improvement in EORTC QLQ-C30 score.29 Based on this, the authors suggested a 10-point change in EORTC QLQ-C30 score represented changes in supportive care needs, and therefore should be considered for clinical use.29

In 2014, another Canadian study estimated the MID for EORTC QLQ-C30 in 369 patients with advanced cancer, who completed the questionnaire at baseline and 1 month post radiation.30 Most common cancer type was breast cancer, followed by lung, prostate, gastrointestinal, renal cell, and others. MID was estimated using both anchor and distribution-based methods for improvement and deterioration. Two anchors of overall health and overall QoL were used, both taken directly from the EORTC QLQ-C30 (questions 29 and 30) where patients rated their overall health and QoL themselves. Improvement and deterioration were categorized as an increase or decrease by 2 units to account for the natural fluctuation of patient scoring. With these 2 anchors, the estimated MIDs across all EORTC QLQ-C30 scales ranged from 9.1 units to 23.5 units for improvement, and from 7.2 units to 13.5 units for deterioration. Distribution-based estimates were closest to 0.5 SD.30 Notably, this study used the global score as an anchor, without providing an MID for this scale, which was the scale used in the NAVIGATE trial, thereby the MIDs from this study are not applicable to this review.

EuroQol 5-Dimensions 5-Levels (EQ-5D-5L) Health State Instrument

Description

The EQ-5D is a generic HRQoL instrument that may be applied to a wide range of health conditions and treatments.32,33 The first of 2 parts of the EQ-5D is a descriptive system that classifies respondents (aged ≥ 12 years) based on the following 5 dimensions: mobility, self-care, usual activities, pain/discomfort, and anxiety/depression. The EQ-5D-5L has 5 possible levels for each domain representing “no problems,” “slight problems,” “moderate problems,” “severe problems,” and “extreme problems.” Respondents are asked to choose the level that reflects their health state for each of the 5 dimensions, corresponding with 3,125 different health states.

Scoring

A scoring function can be used to assign a value (EQ-5D-5L index score) to self-reported health states from a set of population-based preference weights.32,33 The second part is a 20 cm visual analogue scale (EQ VAS) that has end points labelled 0 and 100, with respective anchors of “worst imaginable health state” and “best imaginable health state.” Respondents are asked to rate their health by drawing a line from an anchor box to the point on the EQ VAS which best represents their health on that day. Hence, the EQ-5D produces 3 types of data for each respondent:

  1. A profile indicating the extent of problems on each of the 5 dimensions represented by a 5-digit descriptor, such as 11121, 33211

  2. A population preference-weighted health index score based on the descriptive system

  3. A self-reported assessment of health status based on the EQ VAS

The EQ-5D index score is generated by applying a multi-attribute utility function to the descriptive system. Different utility functions are available that reflect the preferences of specific populations (e.g., US or UK). The lowest possible overall score for the 3L version (corresponding to severe problems on all 5 attributes) varies depending on the utility function that is applied to the descriptive system (e.g., −0.59 for the UK algorithm and −0.109 for the US algorithm). Scores less than 0 represent health states that are valued by society as being worse than dead, while scores of 0 and 1.00 are assigned to the health states “dead” and “perfect health,” respectively. Reported MIDs for the 3L version of the scale have ranged from 0.033 to 0.074.43

Psychometric Properties

Reliability: Teckle et al. conducted a study at the Vancouver Cancer Clinic of patients (N = 184) who had either breast (36%), colorectal (31%), or lung (33%) cancer to investigate whether disease severity could be distinguished by cancer-specific and generic preference-based instruments.34 Internal consistency was calculated using Cronbach alpha and all 5 functioning scales along with global health status showed acceptable consistency (alpha > 0.7), with values ranging from 0.77 to 0.82.

Validity: Validity was assessed using Pearson’s correlation coefficient (r) where r between 0 and 0.3 demonstrated weak correlation, between 0.3 and 0.49 was moderate, and greater than 0.5 was considered strong. Teckle et al. found the following, between the EORTC QLQ-C30 and EQ-5D, r = 0.43; comparing the EORTC QLQ-C30 and EQ VAS, r = 0.73; and between EQ-5D and EQ VAS, r = 0.43. External validity was estimated between cancer severity (self-reported health status, Eastern Cooperative Oncology Group Performance Status [ECOG PS], and cancer stage). An effect size (ES) between 0.2 and 0.5 was considered small, between 0.5 and 0.8 was medium, and greater than 0.8 was large.34 The EQ-5D was able to discriminate populations based on self-reported health status (excellent/good versus fair/very poor; ES = 0.90), and somewhat based on ECOG PS (0 versus 1 to 3; ES = 0.31), but not for stage of cancer (stages I and II versus stages III and IV; ES = 0.06). The EORTC QLQ-C30 performed better in all 3 areas: self-reported health status (ES = 1.39), ECOG PS (ES = 0.65), and stage of cancer (ES = 0.49). It is worth noting that the EQ-5D was based on a non-Canadian population and the comparison with EORTC QLQ-C30 was based solely on the 2 questions asking about overall health and QoL rather than the questionnaire as a whole. Furthermore, there was no information on what type of treatment the patients were receiving when completing the questionnaires.

Responsiveness: Responsiveness was not reported.

Minimal Important Difference

There were no relevant studies reporting the MID among patients with AML.

Patient-Reported Outcome Measurement Information System Fatigue Short Form v1.0–Fatigue 7a (PROMIS F-SF)

Description

PROMIS is a set of standardized tools funded by the National Institutes of Health for measuring patient-reported outcomes.31 The PROMIS has 2 major frameworks—Adult Self-Reported Health and Pediatric Self- and Proxy-Reported Health.31 Each framework has their own physical, mental, and social health domains. Item banks and subsequent PROMIS measures were developed within each framework to assess patient-reported outcomes, such as fatigue and disease conditions.31 Fatigue is part of the PROMIS physical health domain.31 The PROMIS F-SF has 7 items to measure both the experience of fatigue and the interference of fatigue on daily activities over the past week.31

Scoring

For the 7 items, the response options are measured on a 5-point Likert scale, from 1 = never to 5 = always. One item, “How often did you have enough energy to exercise strenuously,” is reverse scored.31 The total score is the sum of the keyed scores of all items. Total scores can range from 7 to 35, with higher scores indicating greater fatigue.31

Psychometric Properties

Reliability: In a secondary analysis that compared fatigue measures in the PROMIS F-SF, the Multidimensional Fatigue Symptom Inventory-Short Form (MFSI-SF), and the Brief Fatigue Inventory (BFI) in patients with fibromyalgia (n = 72), patient with sickle cell disease (n = 60), individuals with cardiometabolic risks (n = 63), pregnant women (n = 72), and healthy controls (n = 40) in 4 studies.31 Reliability of PROMIS F-SF scores was adequate across samples, ranging from 0.72 in pregnant women to 0.88 in healthy controls.31

Validity: Concurrent validity was strong based on the correlations between the PROMIS F-SF and the MFSI-SF (r = 0.70 to 0.85) and those between the PROMIS F-SF and the BFI (r = 0.60 to 0.85).31 Discriminant correlations between the PROMIS F-SF and the Perceived Stress Scale (PSS) were from r = 0.37 to 0.62, and between the PROMIS F-SF and the CES-D ranged from r = 0.45 to 0.64.31 For known-groups validity, the samples in the 4 study had significantly higher levels of fatigue on the PROMIS F-SF than the healthy controls.31

Responsiveness: Responsiveness was not reported.31

Minimal Important Difference

The researchers that conducted the VIALE-A trial assessed the MID using anchor- and distribution-based approaches in a group of AML patients.1 A 3-point difference that fell within the range of 3 to 5 proposed in the literature was considered an appropriate MID for patients with AML.41 The 3-point difference was also applied for the patients with AML in another related trial, the VIALE-C trial.41

Appendix 5: Summary of Protocol Changes for VIALE-A

Note that this appendix has not been copy-edited.

The original protocol (dated October 25, 2016, with 2 patients enrolled globally under the amendment) had several global amendments, and 1 amendment that was specific to China, and allowed enrolment of an open-label cohort. The global amendments are summarized below:

The SAP had 7 versions with amendments to align the SAP with the protocol changes. Version 1 (March 27, 2018) described the initial efficacy and safety analysis methods. Significant amendments, involving changes to end points of interest to this review, follow:

Pharmacoeconomic Review

Abbreviations

AIC

Akaike information criteria

AML

acute myeloid leukemia

BIC

Bayesian information criteria

BSC

best supportive care

CR

complete remission

CRh

complete remission with incomplete hematological recovery

CRi

complete remission with incomplete bone marrow recovery

EFS

event-free survival

HMA

hypomethylating agent

HR

hazard ratio

ICER

incremental cost-effectiveness ratio

LDAC

low-dose cytarabine

NMA

network meta-analysis

OS

overall survival

PD/RL

progressive/relapsed disease

QALY

quality-adjusted life-year

WTP

willingness to pay

Executive Summary

The executive summary comprises 2 tables (Table 1 and Table 2) and a conclusion.

Table 1: Submitted for Review

Item

Description

Drug product

Venetoclax (Venclexta), 10 mg, 50 mg, 100 mg, tablets, oral

Submitted price

Venetoclax, 100 mg tablet: $70

Indication

In combination with azacitidine or low-dose cytarabine, is indicated for the treatment of patients with newly diagnosed acute myeloid leukemia (AML) who are 75 years or older, or who have comorbidities that preclude use of intensive induction chemotherapy.

Health Canada approval status

NOC

Health Canada review pathway

Standard

NOC date

December 4, 2020

Reimbursement request

In combination with azacitidine for the treatment of patients with newly diagnosed AML who are 75 years or older, or who have comorbidities that preclude use of intensive induction chemotherapy.

Sponsor

AbbVie Corporation

Submission history

Previously reviewed: Yes

Indication: For the treatment of patients with CLL who have received at least 1 prior therapy and have a 17p deletion

Recommendation date: December 1, 2016

Recommendation: Not recommended1

Indication: As monotherapy for the treatment of patients with CLL who have received at least 1 prior therapy and who have failed a B-cell receptor inhibitor

Recommendation date: November 30, 2017

Recommendation: Recommended on the condition of cost-effectiveness being improved to an acceptable level2

Indication: In combination with rituximab for the treatment of adult patients with CLL who have received at least 1 prior therapy, irrespective of their 17p deletion status

Recommendation date: May 31, 2019

Recommendation: Recommended on the condition of cost-effectiveness being improved to an acceptable level3

Indication: In combination with obinutuzumab for the treatment of adult patients with previously untreated CLL who are fludarabine ineligible

Recommendation date: November 17, 2020

Recommendation: Recommended on the condition of cost-effectiveness being improved to an acceptable level4

AML = acute myeloid leukemia; CLL = chronic lymphocytic leukemia; NOC = Notice of Compliance.

Table 2: Summary of Economic Evaluation

Component

Description

Type of economic evaluation

Cost-utility analysis

Partitioned survival model

Target population

Patients with newly diagnosed AML for whom IC is unsuitable or who are aged 75 years or older

Treatment

Venetoclax plus azacitidine

Comparators

Azacitidine alone

LDAC

BSC

Perspective

Canadian publicly funded health care payer

Outcomes

QALYs, LYs

Time horizon

Lifetime horizon (90 years)

Key data source

VIALE-C and VIALE-A trials and a network meta-analysis

Submitted results

  • Based on the sequential analyses, the optimal treatments (i.e., on the cost-effectiveness frontier) are BSC, LDAC, and venetoclax plus azacitidine.

  • ICER for venetoclax plus azacitidine when compared with LDAC was $105,286 per QALY gained (1.59 incremental QALYs and $167,432 incremental costs).

Key limitations

  • The sponsor excluded IC as a comparator. Clinical experts consulted for this review indicated that individuals older than 75 would be eligible to receive IC.

  • The sponsor incorporated a cure assumption for individuals who remain in the CR + CRi health state for more than 5 years. Clinical experts indicated that this assumption was unlikely to be correct.

  • A substantial portion of the QALY benefits of venetoclax plus azacitidine occurred after individuals exited the EFS state and were no longer on first-line treatment. Clinical experts indicated there was unlikely to be a substantive benefit for individuals who receive venetoclax plus azacitidine after exiting the EFS health state.

  • In the sponsor’s model, EFS and the duration of first-line treatment were estimated independently. It is likely that EFS and treatment duration are highly correlated.

  • There exists substantial uncertainty surrounding the effectiveness of venetoclax plus azacitidine beyond the follow-up of the VIALE-A trial.

CADTH reanalysis results

  • CADTH reanalyses included estimates for OS curves limiting the benefit of venetoclax plus azacitidine post EFS, and a cure assumption for those who remain in the CR + CRi health state for more than 10 years. In addition to these modifications, CADTH conducted several scenario analyses to quantify the uncertainty surrounding the CADTH base case. These scenario analyses included all individuals in the EFS health state being on treatment, and varying estimates of OS for venetoclax plus azacitidine. CADTH was not able to address the exclusion of IC as a comparator.

  • In the sequential analysis, venetoclax plus azacitidine was associated with an ICER of $125,580 per QALY compared with LDAC; LDAC was associated with an ICER of $72,232 per QALY compared with BSC. Azacitidine remained ruled out as an optimal option.

  • The probability that venetoclax plus azacitidine was cost-effective at a $50,000 WTP threshold compared with LDAC was 0%.

AML = acute myeloid leukemia; BSC = best supportive care; CR + CRi = complete remission plus complete remission with incomplete blood count recovery; EFS = event-free survival; IC = intensive chemotherapy; ICER = incremental cost-effectiveness ratio; LDAC = low-dose cytarabine; LY = life-year; OS = overall survival; QALY = quality-adjusted life-year; WTP = willingness to pay.

Conclusions

Based on the Clinical Review, patients treated with venetoclax plus azacitidine showed benefits in overall survival (OS), overall and early composite combined response (complete remission plus complete remission with incomplete blood count recovery [CR + CRi]), event-free survival (EFS), and complete remission compared with patients treated with azacitidine alone. An indirect treatment comparison comparing venetoclax plus azacitidine with low-dose cytarabine (LDAC) was found to be highly susceptible to bias due to the absence of randomized propensity-score comparisons.

CADTH undertook reanalyses to address limitations with the sponsor’s submission. These reanalyses included: a different assumption on functional form of the OS probability for venetoclax plus azacitidine (Weibull distribution) which resulted in more plausible estimates of survival post EFS for venetoclax plus azacitidine; and changing the sponsor’s assumption of disease being cured for those who remain in the CR + CRi health state from 5 years to 10 years. In the CADTH base case, best supportive care (BSC), LDAC, and azacitidine are considered optimal treatments (i.e., on the cost-effectiveness efficiency frontier). Venetoclax plus azacitidine is more effective and more costly than LDAC (incremental quality-adjusted life-year [QALY]: 1.21; incremental cost: $151,779) with an incremental cost-effectiveness ratio (ICER) of $125,580 per QALY. The probability that venetoclax plus azacitidine is cost-effective at a $50,000 willingness-to-pay (WTP) threshold compared with LDAC was 0%. There is no price reduction at which venetoclax plus azacitidine was cost-effective compared with LDAC at a WTP threshold of $50,000 per QALY, due to the cost of combination therapy and the long duration of first-line treatment. The probability that venetoclax plus azacitidine is cost-effective compared with azacitidine monotherapy at this threshold was also 0%.

The cost-effectiveness of venetoclax plus azacitidine was driven by assumptions about treatment duration and the extrapolation of OS and EFS beyond the observation period of the trial. The pharmacoeconomic model was also associated with notable structural uncertainty that appeared to confer a post-event survival benefit for venetoclax plus azacitidine that was not adequately supported by the available data. These findings taken together suggest the cost-effectiveness of venetoclax plus azacitidine compared with azacitidine monotherapy, LDAC, and BSC are uncertain and likely overestimated. The cost-effectiveness of venetoclax plus azacitidine compared with intensive chemotherapy, which the clinical experts indicated is an important comparator for those over 75 years of age, is unknown.

Stakeholder Input Relevant to the Economic Review

This section is a summary of the feedback received from the patient groups, registered clinicians, and drug plans that participated in the CADTH review process.

One patient advocacy group, the Leukemia and Lymphoma Society of Canada (LLSC) provided input on venetoclax in combination with azacitidine for the treatment of AML. LLSC collected responses from 29 Canadian patients diagnosed with acute myeloid leukemia (AML) using an online survey conducted between December 7, 2020 and January 24, 2021. Respondents reported receiving the following front-line treatments after diagnosis: chemotherapy (n = 24), stem cell transplant or bone marrow transplant (n = 16), drug therapy (n = 6), radiation therapy (n = 5), chimeric antigen receptor (CAR) T-cell therapy (n = 1), and liposomal daunorubicin and cytarabine (n = 1). Five of the respondents had experience with venetoclax in combination with azacitidine.

According to the respondents, the AML symptoms that impact quality of life included fatigue, suddenness of symptom development, anxiety, fear of relapse, and loss of eyesight. Moreover, the respondents reported a wide range of side effects under current treatments. Respondents highlighted that they were unable to work due to disease and associated symptoms as well as the impact on caregivers. Patients’ responses to venetoclax in combination with azacitidine varied greatly, including an overall great experience (n = 1), experiences with side effects (tiredness and loss of appetite, n = 1), no side effects but relapse (n = 1), significant side effects (n = 1), and transition to transplant (n = 1). Their overall opinion of this new treatment varied greatly as well, based on diverging opinions on financial costs, efficacy, and side effects.

The LLSC survey patient respondents also reported the characteristics of new treatment options that they hoped to have, in particular, those that could maintain remission, with fewer side effects, covered by public drug plans, and accessible in wider geographic regions. The opportunity to have access to other supportive options, such as meditation, hypnosis, neuro-linguistic programming support, and awareness support (thoughts, emotions, and behaviours), was also mentioned.

Feedback from registered clinicians suggested that the options for standard of care for first-line AML treatment were azacitidine, LDAC, and supportive care. Clinicians stated that the expected goal of treatment with venetoclax plus azacitidine was an improvement in survival and quality of life, as well as transfusion independence. Clinicians remarked that very frail or very elderly patients likely would not receive venetoclax plus azacitidine, and that patients would need to travel to a clinic to receive the azacitidine component of the treatment. Clinicians also noted that younger and fit patients without significant comorbidities would be better suited for intensive induction chemotherapy.

The drug plans highlighted considerations for the implementation of venetoclax plus azacitidine that are relevant to the economic analysis. One issue is the exclusion of relevant comparators, particularly for those aged 75 years and older, where many patients may be fit to tolerate intensive chemotherapy. Another issue related to whether venetoclax can be used with alternative dosing schedules for azacitidine that some provinces currently fund (e.g., 5 to 2-2, 6 consecutive days) as it differs from the schedule included in this submission. In addition, dosing regiments for azacitidine, venetoclax, and LDAC are slightly different across indications and this was flagged by the drug plans. Another concern of the drug plans related to whether venetoclax plus azacitidine would become available to individuals who have been previously treated with azacitidine as well as a possible selection bias in the VIALE-A trial of healthier patients. It was also noted that other novel therapies are under review by CADTH for the same population.

Several of these concerns were addressed in the sponsor’s model:

CADTH was unable to address the following concerns raised from stakeholder input:

Economic Review

Economic Evaluation

The current review is for venetoclax and azacitidine for newly diagnosed AML who are 75 years or older, or who have comorbidities that preclude use of intensive induction chemotherapy.

Summary of Sponsor’s Economic Evaluation

Overview

The sponsor submitted a cost-utility analysis assessing venetoclax plus azacitidine compared with azacitidine alone, LDAC, and BSC in patients who are newly diagnosed AML for whom intensive chemotherapy is unsuitable. The modelled population was consistent with the VIALE-A clinical trial did not match the reimbursement request. Two subgroup analyses were conducted according to a bone marrow blast count of 20% to 30% with azacitidine as a comparator, and a greater than 30% blast count with azacitidine and LDAC as comparators. The cost-utility analysis was conducted from the perspective of the Canadian publicly funded health system.5

The recommended dose of venetoclax when used in combination with azacitidine consists of 100 mg on day 1, 200 mg on day 2, 400 mg on days 3 to 28 for the first 28-day cycle, and 400 mg administered daily on subsequent 28-day cycles.6 The recommended dose of azacitidine when used alone or in combination with venetoclax consisted of 75 mg/m2 on days 1 to 7 of each 28-day cycle. The recommended dose for LDAC was 20 mg/m2 on days 1 to 10 of each 28-day cycle. BSC was not explicitly defined in the submitted report, but no drug administration was assumed for that strategy.5

Administration costs for venetoclax consist of pharmacy dispensing fees and physician fees for management of oral chemotherapy.5 The administration costs for azacitidine and LDAC were associated with inpatient IV therapy administration. The total drug-acquisition cost per patient for the first 28-day cycle of venetoclax plus azacitidine is $11,724 (venetoclax: $5,585; azacitidine: $6,139) and $11,877 (venetoclax: $5,739; azacitidine: $6,139) for subsequent 28-day cycles, based on a venetoclax unit price of $70 per 100 mg tablet. The total drug-acquisition cost per patient for each 28-day cycle of azacitidine was $7,581. The total drug-acquisition cost per patient for each 28-day cycle of LDAC was $48 based on a price per vial of $4.90. The sponsor assumed no drug-acquisition costs associated with BSC.

The clinical outcomes modelled included QALYs and life-years. The economic analysis was undertaken over a lifetime horizon using a 28-day cycle length. The economic evaluation was conducted from the perspective of a publicly funded health care system and discounting (1.5% per year) was applied to both costs and outcomes.5

Model Structure

A partitioned survival model (PSM) was developed in Microsoft Excel. The PSM consisted of 3 mutually exclusive health states: EFS, progressive/relapsed disease (PD/RL), and death (Figure 1). EFS was defined as the time from treatment initiation to first progression or relapse from complete remission/complete remission with incomplete blood count recovery (CR/CRi), or treatment failure or death due to any cause. All patients enter the model in the EFS health state. Within EFS a proportion of time was assumed to be spent with CR + CRi and the remaining time in EFS without CR or CRi. Duration of first-line treatment was modelled independently from EFS, and patients could stop treatment without transitioning to another state. Patients then transition to the PD/RL state included alive patients who progressed or relapsed. After transitioning to PD/RL patients undergo subsequent treatment. Individuals remain in PD/RL until experiencing death either due to AML related mortality or due to other cause mortality. It was assumed that individuals who remain in the EFS health state with CR + CRi for more than 5 years were “cured” and no longer at risk of transitioning to PD/RL or experiencing disease-related mortality. Patients could also experience treatment-related adverse events, which were assumed to occur during the first model cycle.

Parametric survival models in combination with hazard ratios (HRs) were used to inform OS and EFS. EFS was assumed to be less than or equal to OS at all time points. The proportion of patients in the EFS health state of the model was set to be equal to the EFS curve of each treatment. The proportion of patients in the PD/RL health state was set to be equal to the difference between the proportion of living patients, which was based on the OS curve, and the proportion of EFS patients. During each cycle, the cohort of patients was redistributed among the 3 health states, with death being the absorbing state.

Model Inputs

Baseline patient characteristics for the modelled population and the clinical efficacy of venetoclax plus azacitidine and azacitidine were sourced from the VIALE-A trial (data cut-off: January 4, 2020), while clinical efficacy of LDAC was based on the placebo arm data from the VIALE-C trial. The VIALE-A and VIALE-C trials were multi-centre and randomized double-blind placebo-controlled phase III trials in which patients were assigned in a 2:1 ratio either to venetoclax plus azacitidine or a comparator.7,8 The baseline characteristics of the patient population in the VIALE-A trial consisted of a median age of 76, 21% with prior use of hypomethylating agents (HMAs) and 76% having bone marrow blast counts greater than 30%. The baseline characteristics of the patient population in the placebo arm of the VIALE-C trial consisted of a median age of 76, 21% with prior HMA use, and 76% having bone marrow blast counts greater than 30%.8 Information on BSC efficacy in the network meta-analysis was based on NCT01074047 (Dombret) a multi-centre, randomized, open-label, phase III trial that evaluated azacitidine efficacy and safety versus conventional care regimens that consisted of patients aged 65 years and older with newly diagnosed AML who were not considered eligible for hematopoietic stem cell transplant (HSCT).9 The median age of this arm was 78, with 100% of the 48 patients having a bone marrow blast count of greater than 30% and 0% having previously used an HMA.

Overall survival and EFS for venetoclax plus azacitidine and azacitidine were obtained using propensity score–matched parametric survival models on individual patient-level data from the VIALE-A trial, which was then extrapolated beyond the trial period. Exponential, Weibull, log-logistic, log-normal, Gompertz, and generalized gamma models were considered, and Akaike information criterion (AIC) and Bayesian information criterion (BIC) tests, visual inspection, examination of log-cumulative hazard plots, Schoenfeld residuals tests, and clinical input and external validation were used in the survival model selection process. Graphical representation of the fitted parametric distributions for EFS and OS extrapolations are shown in figures 2 to 5). For the LDAC arm, the HR method was used to evaluate their comparative effectiveness versus venetoclax plus azacitidine. To adjust for the potential difference in patient population between venetoclax plus azacitidine and LDAC, the HR was calculated via a propensity-score analysis using individual patient-level data from the VIALE-A and VIALE-C trials. The OS for BSC was estimated using a network meta-analysis with venetoclax plus azacitidine as the reference. The network meta-analysis was conducted using Bayesian mixed treatment comparison techniques. Bayesian Markov Chain Monte Carlo methods were used to estimate the posterior probability distribution and generate pairwise comparisons for treatments of interest by outcome. The proportion of time in CR + CRi for venetoclax plus azacitidine, azacitidine alone, and LDAC was estimated by the CR + CRi rate in the VIALE-A and VIALE-C trials. For the subgroup analysis of 20% to 30% blasts, parametric survival models were fitted on the VIALE-A trial data. For the subgroup analysis of greater than 30% blasts, parametric survival models were used to inform the venetoclax plus azacitidine and azacitidine estimates of OS and EFS, with the OS and EFS for LDAC estimated using an HR measure similar to the base-case analysis. The survival for the patients assumed to be cured was modelled using general population mortality based on the 2019 Canadian life table.

HRQoL in the VIALE-A and VIALE-C trials was measured using the EuroQol 5-Dimensions 5-Levels questionnaire (EQ-5D-5L).7,8 It was administered at cycle 1 day 1 and on day 1 of every other cycle as well as on the last visit after patients discontinue the treatment. The final visit was defined as the last assessments on or after the date of disease progression, relapse from CR + CRi, or treatment failure. EQ-5D-5L utility scores were estimated from pooled VIALE-A and VIALE-C trial data based on individual dimension scores and using Canada preference weights.7,10,11 A linear mixed-effects model was developed to estimate patient utility scores with a robust variance estimator to account for correlation within patients’ repeated assessments. The linear model adjusted for the grade 3 or 4 adverse events that occurred at a prevalence rate of 5% or greater in the VIALE-A and VIALE-C trials. Adverse event utility and disutility inputs were derived from Wehler.12 For adverse events that were not reported in the literature, values were assumed to be equal to those under the same adverse event category or the average disutility of all the adverse events. The model assumed patients could receive subsequent HSCT after initial treatment. Patients receiving subsequent HSCT were assumed to have additional HSCT disutility that would last for 365 days.13

The dosing schedule, dose intensity, and treatment duration for venetoclax plus azacitidine, azacitidine alone, and LDAC were obtained from the VIALE-A and VIALE-C trials. Venetoclax had a dose intensity of 73%. Azacitidine had a dose intensity of 73% when used in combination with venetoclax, and 90% when used alone. LDAC had a dose intensity of 98%. The median treatment durations for venetoclax plus azacitidine, azacitidine alone, and LDAC were obtained from the VIALE-A and VIALE-C trials and an exponential model was used to extrapolate the time on treatment beyond the trial observation period. The proportion of patients receiving subsequent treatments for each comparator were obtained from a Canadian key opinion leader. For BSC, all patients are assumed to receive subsequent treatment of hydroxycarbamide, also based on Canadian key opinion leader input. Only the subsequent treatments with a prevalence rate greater than or equal to 5% in any of the treatment arms were considered. The dosing schedule for subsequent treatments was sourced from the VIALE-A and VIALE-C trials and Cancer Care Ontario.14,15The mean treatment duration of azacitidine as subsequent treatment was derived from a retrospective database study and was used as treatment duration for all subsequent therapies.5,16 The adverse event rates for BSC were based on Dombret.17 Only adverse events that were grade 3 or 4 with a greater than 5% prevalence rate in any of the arms were considered. The proportion of grade 3 or 4 adverse events managed on either an inpatient or outpatient basis were established based on input from a Canadian key opinion leader.5

The model considered the following cost components: initial treatment costs (including drug and administration), subsequent HSCT costs, subsequent pharmacological treatment costs (including drug and administration), adverse event costs associated with initial treatments, and terminal care costs. The unit drug costs of venetoclax and all other treatments were obtained from IQVIA price list (October 2020). Resource utilization and unit costs were sourced from the overall population in the VIALE-C trial, the literature, public databases, and a Canadian key opinion leader. An inpatient hospitalization cost of $1,817.86 was sourced from the Patient Cost Estimator provided by the Canadian Institute for Health Information (CIHI).18 (A daily cost of being in an intensive care unit of $3,927.67 was sourced from a CIHI 2019 report.19) All patients who transitioned to death were assumed to incur terminal care costs of $86,582.31 during the last cycle before death.20 The inpatient length of stay per cycle, the number of red blood cell transfusions per cycle and number of platelet transfusion per cycle were sourced from the key opinion leader. Monitoring costs were mostly obtained from the Ontario Schedule of Benefits for Physician Services and the Schedule of Benefits for Laboratory Services.21 The Ontario Care Costing Initiative (OCCI) was also used to retrieve the procedure costs for bone marrow aspirates and biopsies. The cost per adverse event for both outpatient and inpatient adverse event management was obtained from the OCCI.22 All of these costs were inflated to 2020 Canadian dollars using the all-item Consumer Price Index.

Summary of Sponsor’s Economic Evaluation Results

The sponsor presented probabilistic analyses (5,000 iterations for the base case).

Base-Case Results

In the sponsor’s base-case analysis for the overall population, BSC was found to have the lowest expected cost ($39,324), but also the lowest expected QALYs (0.58). The cost-effectiveness efficiency frontier included LDAC and venetoclax plus azacitidine but not azacitidine monotherapy, since azacitidine was found to be costlier and less effective than LDAC and was therefore dominated by LDAC. Compared with BSC, LDAC costs an extra $58,582 per expected QALY gained while, compared with LDAC, venetoclax plus azacitidine costs an expected extra $105,286 per QALY gained.

The initial treatment costs were the key cost driver for venetoclax plus azacitidine and azacitidine (63% and 55% of total costs respectively), while medical costs were the key cost drivers for LDAC and BSC (79% and 91% of total costs respectively). Medical costs, particularly in the PD/RL health state, were also a major cost driver for venetoclax plus azacitidine (17% of total costs), since patients on venetoclax plus azacitidine experienced longer OS compared with other treatment options. Consequently, patients in the venetoclax plus azacitidine arm experienced more QALYs than the comparators (2.53 QALYs for venetoclax plus azacitidine compared with 0.58 to 0.94 QALYs for the other treatments). At a WTP threshold of $50,000 per QALY, there was a 0% probability that venetoclax plus azacitidine is cost-effective compared with LDAC. The probability that venetoclax plus azacitidine is cost-effective compared with azacitidine monotherapy at this threshold was also 0%.

Table 3: Summary of the Sponsor’s Economic Evaluation Results

Drug

Total costs ($)

Total QALYs

Sequential ICER ($/QALY)

Best supportive care

39,324

0.58

Reference

Low-dose cytarabine

60,259

0.94

58,582 vs. best supportive care

Azacitidine

95,629

0.88

Dominated by low-dose cytarabine

Venetoclax plus azacitidine

227,691

2.53

105,286 vs. low-dose cytarabine

ICER = incremental cost-effectiveness ratio; QALY = quality-adjusted life-year.

Source: Sponsor’s pharmacoeconomic submission.5

Note: Dominated refers to a treatment having a higher total cost and lower total QALYs when compared with the previous less costly treatment.

Additional results from the sponsor’s submitted economic evaluation base case are presented in Appendix 4 (Table 11).

Sensitivity and Scenario Analysis Results

The sponsor conducted a subgroup analysis on the subgroup of patients with a bone marrow blast count of between 20% and 30% and patients with a blast count greater than 30%. The incremental cost per QALY gained for venetoclax plus azacitidine when compared with azacitidine in the 20% to 30% subgroup analysis was $85,091 per QALY. In the greater than 30% subgroup analysis, azacitidine was dominated, and the incremental cost per QALY gained for venetoclax plus azacitidine when compared with LDAC was $96,294 per QALY. The sponsor performed scenario analyses related to the duration of treatment, the inclusion of cure assumption, and time horizon. The results of these analyses are presented in Appendix 4 (Table 12).

CADTH Appraisal of the Sponsor’s Economic Evaluation

CADTH identified several key limitations to the sponsor’s analysis that have notable implications on the economic analysis:

Additionally, the following key assumptions were made by the sponsor and have been appraised by CADTH (See Table 4).

Table 4: Key Assumptions of the Submitted Economic Evaluation (Not Noted as Limitations to the Submission)

Sponsor’s key assumption

CADTH comment

Gamma distribution not considered.

The sponsor did not implement the gamma distribution in the submitted model. CADTH was not able to assess the impact of not including that distribution on the outcomes of the economic analysis. However, the gamma distribution did not provide the best-fitting curve according to BIC or AIC for any of the treatments.

The EFS was artificially restricted such that it remained under OS. This is a by-product of OS and EFS being independently modelled.

When a partitioned survival model is used, the OS and EFS curves are typically modelled independently. In situations where either of the 2 probabilities is non-zero by the end of the trial follow-up, this assumption is particularly problematic, as it can result in biased estimates. The bias is amplified in the context of a probabilistic analysis where restrictions that are introduced in the model, such as the EFS being artificially restricted to be lower than OS, can amplify the bias. This is a structural assumption shared by all partitioned survival models.

The sponsor did not define what BSC consisted of in the submission.

This limits the usefulness of the model with regard to the comparator arm in decision-making. However, experts agreed that BSC is an unlikely treatment option.

Incomplete administration costs.

According to the product monograph, treatment with venetoclax requires preparatory steps, including anti-hyperuricemic drugs, cytoreduction before treatment, assessment and monitoring of blood chemistry, and laboratory monitoring. These additional steps are associated with additional administration costs. The sponsor’s model assumed the administration costs for venetoclax were limited to pharmacy dispensing fees and physician monitoring for chemotherapy regimens. This is likely to underestimate the initial treatment costs for venetoclax and the estimates of the cost-effectiveness of venetoclax plus azacitidine as a result.

The sponsor did not consider an alternative reference treatment when estimating the OS under BSC using NMA input.

When estimating an absolute effect size (e.g., probability of event) using estimates from an NMA, a reference treatment needs to be assumed. In the submitted model, the reference treatment when estimating BSC OS was assumed to be venetoclax plus azacitidine. However, the choice of venetoclax plus azacitidine as a reference treatment is arbitrary. Ideally, the sponsor would want to assess the sensitivity of the results on that reference treatment assumption by choosing a different reference treatment. However, the sponsor did not conduct such a sensitivity analysis on this assumption.

Hospitalization costs were accrued based on time in state, not on treatment-specific.effects

The sponsor assumed that hospitalizations were dependent on time in a specific health state, not treatment-specific risks of inpatient hospitalization. However, experts agreed there is limited evidence on the inpatient hospitalization risks for the treatments considered.

Did not consider vial sharing.

The sponsor assumed no vial sharing, generating uncertainty in the treatment cost estimates.

AIC = Akaike information criterion; BIC = Bayesian information criterion; BSC = best supportive care; EFS = event-free survival; NMA = network meta-analysis; OS = overall survival.

CADTH Reanalyses of the Economic Evaluation

Base-Case Results

To address limitations identified within the economic model, the CADTH base case was derived by making changes in model parameter values and assumptions, in consultation with clinical experts (Table 5).

Table 5: CADTH Revisions to the Submitted Economic Evaluation

Stepped analysis

Sponsor’s value or assumption

CADTH value or assumption

Corrections to sponsor’s base case

LDAC drug-acquisition costs: The sponsor used a lower drug cost for LDAC based on an expired wholesale price. CADTH has selected the available pricing in the IQVIA database for the concentration of LDAC based on its product monograph (100 mg/mL).

Changes to derive the CADTH base case

1. Cure assumption for those who remain in the CR + CRi state for more than 5 years

Cure assumption for those who remain in the CR + CRi state for more than 5 years.

Cure assumption for those who remain in the CR + CRi state for more than 10 years.

2. Substantial benefit of venetoclax plus azacitidine occurring after EFS

OS distribution for venetoclax plus azacitidine: Log-normal

OS distribution for venetoclax plus azacitidine (Weibull)

CADTH base case

Combined revisions 1 + 2

CR + CRi = complete remission plus complete remission with incomplete blood count recovery; EFS = event-free-survival; LDAC = low-dose cytarabine; OS = overall survival.

CADTH’s base-case results for the main population are presented in Table 6 and stepped reanalysis in Table 13. Disaggregated results of the CADTH reanalysis are presented in Table 14. In CADTH’s base case, venetoclax plus azacitidine was associated with the highest total discounted costs ($205,367) and QALYs (1.97) over the lifetime time horizon. According to the sequential analysis, BSC is preferred for WTP thresholds below $72,232, LDAC for WTP thresholds of between $72,232 and $125,580, and venetoclax plus azacitidine for WTP thresholds above $125,580. Azacitidine was extendedly dominated by LDAC and venetoclax plus azacitidine. The probability that venetoclax plus azacitidine was a cost-effective strategy compared with LDAC was 0% at a WTP threshold of $50,000 per QALY. In the CADTH base case, 44% of the QALYs were accrued after the duration of the VIALE-A trial for venetoclax plus azacitidine (0.87 QALYs).

CADTH did not consider any subgroup analysis, given the minimal differences in the point estimates of the identified subgroups.

Table 6: Summary of the CADTH Reanalysis Results

Drug

Total costs ($)

Total QALYs

ICER vs. BSC ($/QALY)

Sequential ICER

($/QALY)

CADTH base case

BSC

36,180

0.52

Reference

Reference

LDAC

53,588

0.76

72,232

72,232 vs. BSC

Venetoclax plus azacitidine

205,367

1.97

116,680

125,580 vs. LDAC

Azacitidine

95,659

0.87

169,939

Extendedly dominated

BSC = best supportive care; ICER = incremental cost-effectiveness ratio; LDAC = low-dose cytarabine; QALY = quality-adjusted life-year.

Note: Reanalyses are based on publicly available prices of the comparator treatments.

Scenario Analysis Results

Price-reduction analyses were conducted using both the sponsor and CADTH base case (Table 7). In the price-reduction scenarios, CADTH varied the price of venetoclax, keeping the price of azacitidine constant. Within the CADTH base case and the sponsors base case, there was no price reduction that resulted in venetoclax plus azacitidine being considered cost-effective at a WTP threshold of $50,000 per QALY. This is due to the cost of combination therapy and the long duration of first-line treatment for individuals who receive venetoclax plus azacitidine. In particular, the significant cost of azacitidine implied that even if venetoclax was offered at a price of $0, the cost of combination therapy would not be low enough for venetoclax plus azacitidine to be considered cost-effective at a WTP threshold of $50,000 per QALY. Exploratory price-reduction analyses were performed to estimate the necessary price reduction to reach a $50,000 per QALY threshold for the combination of venetoclax plus azacitidine (72%), and the additional reduction of the azacitidine price (45%) if the price of venetoclax were reduced by 100%.

Table 7: CADTH Price-Reduction Analyses

ICERs for venetoclax plus azacitidine vs. comparators

ICERs for venetoclax plus azacitidine vs. LDAC ($/QALY)

Price reduction (%)

Sponsor base case ($)

CADTH reanalysis ($)

No price reduction

103,995

131,933

10

100,021

126,227

20

96,048

120,521

30

92,074

114,816

40

88,101

109,110

50

84,128

103,404

60

80,154

97,699

70

76,181

91,993

80

72,208

86,287

90

68,234

80,582

100

64,261

74,876

BSC = best supportive care; ICER = incremental cost-effectiveness ratio; LDAC = low-dose cytarabine; QALY = quality-adjusted life-year.

Note: Only non-dominated strategies are presented. Reanalyses are based on publicly available prices for the comparator treatments.

CADTH also performed a set of scenario analyses. The scenarios included selecting the exponential distribution for venetoclax plus azacitidine OS, assuming that all patients in the EFS state were on active therapy, using the second best–fitting OS and EFS curves (according to BIC). Additionally, CADTH conducted exploratory analyses considering venetoclax plus LDAC as a comparator as well as using a shortened time horizon. Detailed results are presented in Appendix 4 (Table 15).

Based on the sequential analysis, all the scenarios considered altered the ICER for venetoclax plus azacitidine versus other comparators. The 2 largest impacts were assuming that all individuals who are in the EFS health state were on first-line treatment (ICER for venetoclax plus azacitidine versus LDAC = $273,764 per QALY) and setting the model time horizon equal to that of the VIALE-A trial (ICER for venetoclax plus azacitidine versus azacitidine = $411,828 per QALY).

Taken together, the findings within the CADTH base-case reanalysis and scenario analyses suggest that in the absence of long-term data, the cost-effectiveness of venetoclax plus azacitidine remains highly uncertain. The CADTH base-case and scenario results suggest that the magnitude of incremental effectiveness appears to be driven by 2 principal factors: the benefit of venetoclax plus azacitidine after EFS, and the duration an individual can remain in the EFS health state while being off treatment. The model findings were sensitive to changes in the parametric extrapolation assumptions for OS and EFS, as seen by the second best–fit scenario analysis. In particular, although most of the distributions for OS and EFS that were implemented in the submitted model fitted the observed data well, they diverged considerably in extrapolations beyond the trial follow-up time. The distributional assumptions made by CADTH ensured that post-event survival is similar between strategies, since an assumption of a post-event survival benefit for venetoclax plus azacitidine was not supported by the submitted evidence or by the clinical feedback from experts consulted by CADTH.

Issues for Consideration

Overall Conclusions

Based on the CADTH Clinical Review of the VIALE-A study results and a sponsor-submitted indirect treatment comparison, treatment with venetoclax plus azacitidine increased OS and EFS compared with LDAC and BSC among patients with newly diagnosed AML who have comorbidities that preclude the use of intensive induction chemotherapy over the trial’s follow-up (median 20 months). The extrapolated difference in EFS and OS between venetoclax plus azacitidine and both LDAC and BSC were the key drivers of incremental effectiveness in the economic analysis. The duration of first-line treatment was a key driver of costs in the economic analysis. The CADTH Clinical Review found that the OS benefit beyond progression that was observed in the economic analysis is not supported by evidence or clinical experience. Intensive chemotherapy was excluded as a comparator, despite the indication from clinical experts that a notable proportion (upward of 30%) of those 75 years or older would receive intensive chemotherapy in Canada.

CADTH undertook reanalyses to address limitations with the sponsor’s submission. These reanalyses included: a different assumption on the functional form of the OS probability for venetoclax plus azacitidine (Weibull distribution) that limits the benefit of venetoclax plus azacitidine post EFS, and changing the sponsor’s assumption of disease being cured for those who remain in the CR + CRi health state from 5 years to 10 years. In the CADTH base case, BSC, LDAC, and venetoclax plus azacitidine were considered optimal treatments (i.e., on the cost-effectiveness efficiency frontier). The results of the CADTH reanalysis were broadly aligned with the sponsor’s submission. Venetoclax plus azacitidine was more effective and more costly than LDAC (incremental QALY: 1.21; incremental cost: $151,779), with an ICER of $125,580 per QALY. The probability that venetoclax plus azacitidine was cost-effective at a $50,000 WTP threshold compared with LDAC was 0%. There was no price reduction at which venetoclax plus azacitidine was cost-effective compared with LDAC at a WTP threshold of $50,000 per QALY, due to the cost of combination therapy and the long duration of first-line treatment. The probability that venetoclax plus azacitidine is cost-effective compared with azacitidine monotherapy at this threshold was also 0%.

The CADTH base-case results are associated with substantial uncertainty for multiple reasons. First, the modelling approach followed by the sponsor did not address the dependence between EFS and treatment duration. This, in turn, resulted in uncertainty in the extrapolation of the treatment duration. The sponsor provided limited evidence on the probability of stopping treatment over time, so CADTH was not able to adequately assess what the duration of treatment would be. In a scenario analysis where individuals were assumed to be on treatment throughout the EFS, the ICER of venetoclax plus azacitidine versus LDAC increased to $273,764 per QALY.

The model had several further limitations that prevented CADTH from estimating an unbiased estimate of cost-effectiveness. The exclusion of intensive chemotherapy as a comparator and the benefit accrued after EFS, as noted earlier, provided insufficient clinical evidence to support such a finding. The EFS and OS were estimated independently, which likely resulted in unrealistic scenarios in the extrapolation of the model (e.g., EFS probability > OS probability). Taken together, these findings suggest the cost-effectiveness results were driven primarily by assumptions about the relationship between time to treatment discontinuation, EFS, and OS, which were uncertain within the trial data.

The cost-effectiveness of venetoclax plus azacitidine compared with intensive chemotherapy is unknown.

References

1.pCODR Expert Review Committee (pERC) initial recommendation: venetoclax (Venclexta - AbbVie Corporation). Ottawa (ON): CADTH; 2016 Dec 1: https://cadth.ca/sites/default/files/pcodr/pcodr_venetoclax_venclexta_cll_17pdel_in_rec.pdf. Accessed 2021 Feb 18.

2.CADTH pCODR Expert Review Committee (pERC) initial recommendation: venetoclax (Venclexta - AbbVie Corporation). Ottawa (ON): CADTH; 2017 Nov 30: https://cadth.ca/sites/default/files/pcodr/pcodr_venetoclax_venclexta_cll_in_rec.pdf. Accessed 2021 Feb 18.

3.CADTH pCODR Expert Review Committee (pERC) final recommendation: venetoclax (Venclexta - AbbVie Corporation). Ottawa (ON): CADTH; 2019 May 31: https://cadth.ca/sites/default/files/pcodr/Reviews2019/10162VenetoclaxRituximabCLL_FnRec_approvedbyChair_REDACT_Post_31May2019-final.pdf. Accessed 2021 Feb 18.

4.CADTH pCODR Expert Review Committee (pERC) final recommendation: venetoclax (Venclexta - AbbVie Corporation). Ottawa (ON): CADTH; 2020 Nov 17: https://cadth.ca/sites/default/files/pcodr/Reviews2020/10212VenetoclaxObinutuzumabCLL_fnRec_EC_Post17Nov2020_final.pdf. Accessed 2021 Feb 18.

5.Pharmacoeconomic evaluation [internal sponsor's report]. In: Drug Reimbursement Review sponsor submission: Venclexta (venetoclax) in combination with azacitidine for the treatment of acute myeloid leukemia (AML), 10 mg, 50 mg, and 100 mg tablets. Pointe-Claire (QC): AbbVie Corporation; 2021 Jan 8.

6.Venclexta (venetoclax): 10 mg, 50 mg, and 100 mg tablets [product monograph]. St-Laurent (QC): AbbVie Corporation; 2020 Dec 3.

7.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. PubMed

8.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. PubMed

9.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. PubMed

10.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. PubMed

11.Xie F, Pullenayegum E, Gaebel K, et al. A time trade-off-derived value set of the EQ-5D-5L for Canada. Med Care. 2016;54(1):98-105. PubMed

12.Wehler E, Storm, M., Kowal, S., Campbell, C., Boscoe, A. A health state utility model estimating the impact of ivosidenib on quality of life in patients with relapsed/refractory acute myeloid leukemia. Presented at: 23rd Congress of the European Hematology Associtation, 2018 Jun 14-18, Stockholm, Sweden. 2018: https://investor.agios.com/static-files/25de7161-9a10-4d8c-8a03-8bd5e7d0a5d3. Accessed 2021 Feb 18.

13.Guadagnolo BA, Punglia RS, Kuntz KM, Mauch PM, Ng AK. Cost-effectiveness analysis of computerized tomography in the routine follow-up of patients after primary treatment for Hodgkin's disease. J Clin Oncol. 2006;24(25):4116-4122. PubMed

14.Cancer Care Ontario. Azctvene regimen. 2020: https://www.cancercareontario.ca/en/drugformulary/regimens/monograph/66881. Accessed 2021 Feb 18.

15.Cancer Care Ontario. CYTA(LD) regimen. 2019; https://www.cancercareontario.ca/en/drugformulary/regimens/monograph/51351. Accessed 2021 Feb 18.

16.Stahl M, DeVeaux M, Montesinos P, et al. Hypomethylating agents in relapsed and refractory AML: outcomes and their predictors in a large international patient cohort. Blood Adv. 2018;2(8):923-932. PubMed

17.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. PubMed

18.Canadian Institute for Health Information. Patient cost estimator. 2020; https://www.cihi.ca/en/patient-cost-estimator. Accessed 2021 Feb 18.

19.Care in Canadian ICUs. Ottawa (ON): Canadian Institute for Health Information; 2016: https://secure.cihi.ca/free_products/ICU_Report_EN.pdf. Accessed 2021 Feb 18.

20.de Oliveira C, Pataky R, Bremner KE, et al. Phase-specific and lifetime costs of cancer care in Ontario, Canada. BMC Cancer. 2016;16(1):809. PubMed

21.Schedule of benefits for laboratory services: effective July 1, 2020. Toronto (ON): Ontario Ministry of Health; 2020: https://www.health.gov.on.ca/en/pro/programs/ohip/sob/lab/lab_mn2020.pdf. Accessed 2021 Feb 18.

22.Ontario Ministry of Health and Long Term Care. Ontario care costing analysis tool. 2020: https://hsimi.ca/occp/occpreports/. Accessed 2021 Feb 18.

23.Vidaza (azacitidine): 100 mg injection [product monograph]. Mississauga (ON): Celgene Inc; 2018 Mar 22: https://pdf.hres.ca/dpd_pm/00044422.PDF. Accessed 2021 Feb 18.

24.BCCA protocol summary for therapy of acute myeloid leukemia using low-dose cytarabine. Vancouver (BC): BC Cancer Agency; 2015: http://www.bccancer.bc.ca/chemotherapy-protocols-site/Documents/Leukemia-BMT/LKAMLCYT_Protocol.pdf Accessed 2021 Feb 18.

25.3+7 regimen. Toronto (ON): Cancer Care Ontario; 2019. Accessed 2021 Feb 18.

26.FLAG+IDA regimen. Fludarabine-cytarabine-filgrastim-idarubicin. Toronto (ON): Cancer Care Ontario; 2019: https://www.cancercareontario.ca/sites/ccocancercare/files/FLAGIDA_HEM_AML_A.pdf. Accessed 2021 Feb 18.

27.Ontario Ministry of Long-Term Care. Ontario drug benefit formulary/comparative drug index. 2020; https://www.formulary.health.gov.on.ca/formulary/. Accessed 2021 Feb 18.

28.Budget Impact Analysis [internal sponsor's report]. Drug Reimbursement Review sponsor submission: Venclexta (venetoclax) in combination with azacitidine for the treatment of acute myeloid leukemia (AML), 10 mg, 50 mg, and 100 mg tablets. Pointe-Claire (QC): AbbVie Corporation; 2021 Jan 8.

Appendix 1: Cost Comparison Table

Note that this appendix has not been copy-edited.

The comparators presented in the following table have been deemed to be appropriate based on feedback from clinical expert(s) and drug plans. Comparators may be recommended (appropriate) practice or actual practice. Existing Product Listing Agreements are not reflected in the table and as such, the table may not represent the actual costs to public drug plans.

Table 8: CADTH Cost Comparison Table for Acute Myeloid Leukemia

Treatment

Strength/concentration

Form

Price ($)

Recommended dosage

28-day cycle cost ($)

Average annual cost ($)

Venetoclax (VENCLEXTA) + low-dose cytarabine (CYTOBAR)

Venetoclax (Venclexta)b

10 mg

50 mg

100 mg

Tablet

7.0000a

35.0000a

70.0000a

100 mg on day 1; 200 mg on day 2; 400 mg on day 3; 400 mg on day 4 and onward

Cycle 1: 7,490

Cycle 2+: 7,840

101,850 to 102,200

Azacitidineb,c

100 mg

Vial for powdered suspension

599.9900

(5.9999 per mg)

75 mg/m2 on days 1 to 7

8,400

109,498

Venetoclax + azacitidine

Cycle 1: 15,890

Cycle 2+: 16,240

211,348

Non-intensive chemotherapies

Azacitadinec

100 mg

Powdered suspension

599.9900

(5.9999 per mg)

75 mg/m2 daily for days 1 to 7

8,400

109,498

Low-dose cytarabined

100 mg/mL

(5 mL vial)

Injectable solution

76.8500

(15.37 per mL)

20 mg/m2, days 1 to 10

769

10,018

100 mg/mL

(20 mL vial)

Injectable solution

306.5000

(15.3250 per mL)

Induction therapy (7 + 3)e

Cytarabine

100 mg/mL

(5 mL vial)

Injectable solution

76.8500

(15.37 per mL)

100 mg/m2, days 1 to 7

200 mg/m2, days 1 to 7 f

538

NA

100 mg/mL

(20 mL vial)

Injectable solution

306.5000

(15.3250 per mL)

Daunorubicin

20 mg

Powdered solution

91.0000

60 mg/m2 IV days 1 to 3e

1,638

NA

Idarubicin

1 mg/mL

(5 mL vial)

IV solution

211.5200 (42.304 per mL)

12 mg/m2 on days 1, 2, and 3e,f

3,173

NA

7 + 3 induction therapy (cytarabine 100 or 200 mg/m2 + daunorubicin 60 mg/m2)f

2,176

NA

7 + 3 induction therapy (cytarabine 200 mg/m2 + idarubicin 12mg/m2)f

3,711

NA

FLAG-IDA (first-line and salvage therapy)

Filgrastim

0.30 mg/0.5 mL

Pre-filled syringe

144.3135 (per 0.5 mL pre-filled syringe)

0.30 mg days 1 to 4

577

7,525

0.30 mg/mL

Vial

176.1330

0.480 mg/0.8 mL

Pre-filled syringe

230.9000

230.9017

0.480 mg/1.6 mL

Vial

230.9000

0.600 mg/mL

Vial

352.2650 (mL in 10 × 0.8 mL pen)

352.2660 (mL in 10 × 0.5 mL pen)

Idarubicin

1 mg/mL

(5 mL vial)

IV solution

211.5200

(42.3040 per mL)

10 mg/m2 days 1 to 2

1,692

22,059

Fludarabine

10 mg

Tablet

40.0760h

30 mg/m2 days 1 to 4

962

12,538

Cytarabine

100 mg/mL

(5 mL vial)

Injectable solution

76.8500

(15.37 per mL)

2,000 mg/m2 days 1 to 4

2,452

31,964

100 mg/mL

(20 mL vial)

Injectable solution

306.5000

(15.3250 per mL)

FLAG-IDA (first-line and salvage therapy)

5,683

74,082

AML = acute myeloid leukemia; FLAG = fludarabine, cytarabine, granulocyte colony-stimulating factor; FLAG-IDA = fludarabine, cytarabine, granulocyte colony-stimulating factor plus idarubicin; LDAC = low-dose cytarabine; NA = not applicable (due to being a single cycle for induction —see regimen monograph).

Note: All prices are from the IQVIA (DeltaPA database) (accessed March 26, 2021), unless otherwise indicated, and do not include dispensing fees. Where applicable, assumes 1.81 m2 and no vial sharing.

aSponsor-submitted price.

bBased on 28-day cycles as per the Venclexta product monograph.6

cAzacitidine product monograph.23

dCytarabine dosing as per British Columbia Cancer Agency protocol.24

e3 + 7 protocol as per Cancer Care Ontario.25

fAs per clinical expert input from CADTH’s review of Vyxeos.

gEvery 28 days as per Cancer Care Ontario regimen monograph FLAG-IDA.26

hPrice obtained from the Ontario Drug Benefit Formulary.27

Appendix 2: Submission Quality

Note that this appendix has not been copy-edited.

Table 9: Submission Quality

Description

Yes/No

Comments

Population is relevant, with no critical intervention missing, and no relevant outcome missing

No

Model population does not match reimbursement request. The reimbursement request is for venetoclax plus azacitidine for newly diagnosed AML who are 75 years or older, or who have comorbidities that preclude use of intensive induction chemotherapy. The modelled population was patients who are newly diagnosed with AML for whom IC is unsuitable.

The model excludes IC as a comparator. Clinical experts indicated that those over the age of 75 would be eligible to receive IC.

Model has been adequately programmed and has sufficient face validity

No

The sponsor used numerous IFERROR statements in their model. IFERROR statements lead to situations in which the parameter value is overwritten with an alternative value without alerting the user to the automatized overwriting. The systematic use of IFERROR statements makes thorough auditing of the sponsor’s model impossible, as it remains unclear whether the model is running inappropriately by overriding errors. Best programming practices are such that any errors alert the user to a specific error.

Model structure is adequate for decision problem

No

The PSM has a structural assumption that EFS and OS are independent, this can result in substantial benefits after individuals have exited the event-free state and are no longer on first-line treatment.

Data incorporation into the model has been done adequately (e.g., parameters for probabilistic analysis)

Yes

NA

Parameter and structural uncertainty were adequately assessed; analyses were adequate to inform the decision problem

Yes

NA

The submission was well organized and complete; the information was easy to locate (clear and transparent reporting; technical documentation available in enough detail)

Yes

NA

AML = acute myeloid leukemia; EFS = event-free survival; IC = intensive chemotherapy; OS = overall survival; PSM = partitioned survival model; NA = not applicable

Appendix 3: Additional Information on the Submitted Economic Evaluation

Note that this appendix has not been copy-edited.

Figure 1: Model Structure

Model structure from sponsor’s economic submission.

Source: Sponsor’s economic submission.5

Figure 2: Observed and Extrapolated Event-Free Survival — Venetoclax Plus Azacitidine

A graph showing observed event-free survival with different predicted extrapolations overlaid.

Source: Sponsor’s economic submission.5

Figure 3: Observed and Extrapolated Event-Free Survival: Azacitidine

A graph showing observed event-free survival with different predicted extrapolations overlaid.

Source: Sponsor’s economic submission.5

Figure 4: Observed and Extrapolated Event-Free Survival — Venetoclax Plus Azacitidine

A graph showing observed overall survival with different predicted extrapolations overlaid.

Source: Sponsor’s economic submission.5

Figure 5: Observed and Extrapolated Event-Free Survival: Azacitidine

A graph showing observed overall survival with different predicted extrapolations overlaid.

Source: Sponsor’s economic submission.5

Table 10: Total Drug-Acquisition and Administration Cost per Treatment

Treatment

Median treatment duration (cycle)

Source of treatment duration

Drug and administration costs for the first cycle ($)

Drug and administration costs for subsequent cycles ($)

VEN-AZA

8.26

VIALE-A trial84

12,627.20

12,780.92

AZA

4.67

VIALE-A trial84

8,449.85

8,449.85

LDAC

1.85

VIALE-A trial84

1,441.82

1,441.82

AZA = azacitidine; LDAC = low-dose cytarabine; VEN-AZA = venetoclax in combination with azacitidine.

Table 11: Disaggregated Summary of Sponsor’s Submitted Economic Evaluation Results

Treatment

Component

Value

Incremental (vs. BSC)

Discounted LY

BSC

Event-free survival

0.32

NA

PD/RL

0.44

NA

Total LYs

0.75

NA

LDAC

Event-free survival

0.52

0.20

PD/RL

0.62

0.18

Total LYs

1.14

0.39

VEN-AZA

Event-free survival

1.77

1.45

PD/RL

0.89

0.45

Total LYs

2.66

1.91

AZA

Event-free survival

0.80

0.48

PD/RL

0.32

−0.12

Total LYs

1.12

0.37

Discounted QALYs

BSC

Event-free survival with CR/CRi

0.00

NA

Event-free survival without CR/CRi

0.25

NA

PD/RL

0.32

NA

Total QALYs

0.57

NA

LDAC

Event-free survival with CR/CRi

0.06

0.06

Event-free survival without CR/CRi

0.36

0.11

PD/RL

0.45

0.13

Total QALYs

0.87

0.3

VEN-AZA

Event-free survival with CR/CRi

0.91

0.91

Event-free survival without CR/CRi

0.52

0.27

PD/RL

0.65

0.33

Total QALYs

2.07

1.50

AZA

Event-free survival with CR/CRi

0.16

0.16

Event-free survival without CR/CRi

0.48

0.23

PD/RL

0.23

−0.09

Total QALYs

0.87

0.30

Discounted costs ($)

BSC

Initial treatment costs

0

NA

Subsequent treatment costs

880

NA

Subsequent HSCT costs

0

NA

Adverse event

2,599

NA

Medical costs

35,492

NA

Total costs

38,972

NA

LDAC

Initial treatment costs

3,117

3,117

Subsequent treatment costs

4,837

3,957

Subsequent HSCT costs

0

0

Adverse event costs

4,496

1,897

Medical costs

45,033

9,541

Total costs

57,484

18,512

VEN-AZA

Initial treatment costs

144,001

144,001

Subsequent treatment costs

369

−511

Subsequent HSCT costs

1,296

1,296

Adverse event costs

5,743

3,144

Medical costs

64,170

28,678

Total costs

215,579

176,607

AZA

Initial treatment costs

52,414

52,414

Subsequent treatment costs

831

−49

Subsequent HSCT costs

1,294

1,294

Adverse event costs

3,743

1,144

Medical costs

37,320

1,828

Total costs

95,603

56,631

Sequential ICER ($/QALY)

ICER vs. SOC ($/QALY)

BSC

Reference

Reference

LDAC

61,707 vs. BSC

61,707

VEN-AZA

131,746 vs. LDAC

117,738

AZA

Extendedly dominated

188,770

AZA = azacitidine; HSCT = hematopoietic stem cell transplant; ICER = incremental cost-effectiveness ratio; LY = life-year; QALY = quality-adjusted life-year; VEN-AZA = venetoclax in combination with azacitidine; vs. = versus ; NA = not applicable

Table 12: Sponsor’s Submitted Scenario Analysis Results

Scenario

ICER for LDAC vs. BSC ($/QALY)

ICER for AZA vs. BSC ($/QALY)

ICER for VEN-AZA vs. BSC ($/QALY)

Base case

61,707

188,770

117,738

1

Median treatment duration

66,636

Dominated

98,807

2

Excluding cure assumption

60,206

Dominated

108, 374

3

10-year time horizon

61,774

Extendedly dominated

131,640

AZA = azacitidine; BSC = best supportive care; CR = complete remission; ICER = incremental cost-effectiveness ratio; LDAC = low-dose cytarabine; QALY = quality-adjusted life-year; VEN = venetoclax; vs. = versus .

Appendix 4: Additional Details on the CADTH Reanalyses and Sensitivity Analyses of the Economic Evaluation

Note that this appendix has not been copy-edited.

Detailed Results of the CADTH Base Case

Table 13: Summary of the Stepped Analysis of the CADTH Reanalysis Results

Scenario

Drug

Total costs ($)

Total QALYs

Sequential ICER
($/QALY)

Sponsor’s base case

BSC

39,324

0.58

Reference

LDAC

60,259

0.94

58,582 vs. BSC

VEN-AZA

227,691

2.53

105,286 vs. LDAC

AZA

95,629

0.88

Dominated

1. Cure assumption = 10 year

BSC

39,119

0.58

Reference

LDAC

61,477

0.93

64,494 vs. BSC

VEN-AZA

232,333

2.54

105,796 vs. LDAC

AZA

95,659

0.87

Dominated

2. OS VEN-AZA Weibull distribution

BSC

36,180

0.52

Reference

LDAC

53,478

0.76

71,617 vs. BSC

VEN-AZA

203,857

2.02

119,775 vs. LDAC

AZA

95,641

0.88

Extendedly dominated

3. CADTH base case (1 + 2)

BSC

36,180

0.52

Reference

LDAC

53,588

0.76

72,232 vs. BSC

VEN-AZA

205,367

1.97

125,580 vs. LDAC

AZA

95,659

0.87

Extendedly dominated

AZA = azacitidine; BSC = best supportive care ICER = incremental cost-effectiveness ratio; LDAC = low-dose cytarabine; LY = life-year; OS = overall survival; QALY = quality-adjusted life-year; Reference = this treatment was used as the reference; VEN-AZA = venetoclax in combination with azacitidine; vs. = versus.

Table 14: Disaggregated Summary of CADTH’s Economic Evaluation Results

Treatment

Component

Value

Incremental (vs. BSC)

Discounted LY

BSC

Event-free survival

0.32

NA

PD/RL

0.36

NA

Total LYs

0.67

NA

LDAC

Event-free survival

0.53

0.21

PD/RL

0.46

0.10

Total LYs

0.98

0.31

VEN-AZA

Event-free survival

2.00

1.68

PD/RL

0.49

0.14

Total LYs

2.49

1.82

AZA

Event-free survival

0.79

0.47

PD/RL

0.33

−0.03

Total LYs

1.12

0.45

Discounted QALYs

BSC

Event-free survival with CR/CRi

0.00

NA

Event-free survival without CR/CRi

0.25

NA

PD/RL

0.26

NA

Total QALYs

0.52

NA

LDAC

Event-free survival with CR/CRi

0.05

0.05

Event-free survival without CR/CRi

0.37

0.11

PD/RL

0.34

0.07

Total QALYs

0.76

0.24

VEN-AZA

Event-free survival with CR/CRi

1.03

1.03

Event-free survival without CR/CRi

0.58

0.32

PD/RL

0.36

0.10

Total QALYs

1.97

1.45

AZA

Event-free survival with CR/CRi

0.16

0.16

Event-free survival without CR/CRi

0.47

0.22

PD/RL

0.24

−0.02

Total QALYs

0.87

0.36

Discounted costs ($)

BSC

Initial treatment costs

$0

NA

Subsequent treatment costs

$892

NA

Subsequent HSCT costs

$0

NA

Adverse event costs associated with initial treatment

$2,612

NA

Medical costs

$32,676

NA

Total costs

$36,180

NA

LDAC

Initial treatment costs

$4,648

$4,648

Subsequent treatment costs

$4,887

$3,995

Subsequent HSCT costs

$0

$0

Adverse event costs associated with initial treatment

$4,537

$1,925

Medical costs

$39,516

$6,839

Total costs

$53,588

$17,408

VEN-AZA

Initial treatment costs

$144,002

$144,002

Subsequent treatment costs

$387

$-505

Subsequent HSCT costs

$1,304

$1,304

Adverse event costs associated with initial treatment

$5,708

$3,096

Medical costs

$53,965

$21,288

Total costs

$205,367

$169,186

AZA

Initial treatment costs

$52,414

$52,414

Subsequent treatment costs

$833

$-58

Subsequent HSCT costs

$1,306

$1,306

Adverse event costs associated with initial treatment

$3,716

$1,104

Medical costs

$37,389

$4,712

Total costs

$95,659

$59,479

Sequential ICER ($/QALY)

ICER vs. BSC ($/QALY)

BSC

Reference

Ref.

LDAC

$72,232 vs. BSC

$72,532

VEN-AZA

$125,580 vs. LDAC

$116,680

AZA

Extendedly dominated

$169,939

AZA = azacitidine; BSC = best supportive care; EFS = event-free-survival; HSCT = hematopoietic stem cell transplant; ICER = incremental cost-effectiveness ratio; LDAC = low-dose cytarabine; LY = life-year; OS = overall survival; QALY = quality-adjusted life-year; VEN-AZA = venetoclax in combination with azacytidine; NA = not applicable.

Scenario Analyses

Table 15: Summary of the CADTH Scenario Analysis

Scenario

Drug

Sequential ICER ($/QALY)

1

VEN-AZA OS survival estimate: Exponential not Weibull.

BSC

LDAC

$76,594

VEN-AZA

$133,637

AZA

Extendedly dominated

2

For all treatments, time on first-line treatments is the same as time event-free.

BSC

LDAC

$114,062

VEN-AZA

$273,764

AZA

Extendedly dominated

3

Second best–fitting curves according to BIC (except for VEN-AZA consider only subset of candidate curves which generate < 1 LY benefit post EFS). PFS exponential for VEN-AZA and AZA. OS: Weibull VEN-AZA, exponential VEN.

BSC

LDAC

$74,875

VEN-AZA

$168,363

AZA

Extendedly dominated

AZA = azacitidine; BIC = Bayesian information criterion; BSC = best supportive care; EFS = event-free survival; ICER = incremental cost-effectiveness ratio; LDAC = low-dose cytarabine; LY = life-year; OS = overall survival; QALY = quality-adjusted life-year; VEN-AZA = venetoclax in combination with azacitidine.

Additionally, CADTH conducted 2 exploratory scenario analyses. The first set the model time horizon to that of the pivotal trial to quantify the amount of health and cost outcomes incurred during that period. The second exploratory analysis included venetoclax in combination with LDAC as a comparator, as CADTH experts indicated there may be potential overlap in the population that would receive either venetoclax plus azacitidine or venetoclax plus LDAC. The results of these analyses are presented below.

Table 16: Summary of the CADTH Exploratory Analyses

Exploratory Scenario

Drug

Sequential ICER ($/QALY)

1

Considering VEN + LDAC a comparator.

BSC

VEN + LDAC

$62,231 vs. BSC

VEN-AZA

$180,591 vs. VEN-LDAC

LDAC

Extendedly dominated

AZA

Dominated

2

Time horizon is equal to that of the pivotal trial (2 years).

BSC

LDAC

$95,159

AZA

$282,233

VEN-AZA

$411,827

AZA = azacitidine; BSC = best supportive care; ICER = incremental cost-effectiveness ratio; LDAC = low-dose cytarabine; LY = life-year; QALY = quality-adjusted life-year; VEN-AZA = venetoclax in combination with azacitidine; VEN-LDAC = venetoclax in combination with LDAC vs. = versus .

Appendix 5: Submitted Budget Impact Analyses and CADTH Appraisal

Note that this appendix has not been copy-edited.

Table 17: Summary of Key Takeaways

Key Takeaways of the BIA

  • CADTH identified the following key limitations with the sponsor’s analysis:

    • There was uncertainty with several epidemiological inputs used to derive the market size.

    • The sponsor’s market share uptake assumptions of venetoclax in the new drug scenario does not reflect the expectations of the clinical experts consulted for this review. The estimated market shares remain uncertain with the potential availability of venetoclax in combination with low-dose cytarabine.

  • The CADTH reanalyses included revising market share estimates for venetoclax in the new drug scenario, revising the epidemiological inputs to derive the market size, allowing for drug wastage; removing patient co-payments, and aligning BIA model inputs to those applied in the pharmacoeconomic analysis.

  • Based on the CADTH reanalysis, the budget impact from the venetoclax in combination with azacitidine would result in an incremental budget impact of $16,784,064 in year 1, $21,182,961 in year 2, and $32,039,516 in year 3, for a total budget impact of $70,006,541. The results were primarily driven by the market share uptake of venetoclax plus azacitidine, number of patients eligible for treatment, and proportion of patients ineligible for induction chemotherapy.

Summary of Sponsor’s BIA

In the submitted budget impact analysis (BIA), the sponsor assessed the venetoclax in combination with azacitidine (VEN-AZA)for adults with newly diagnosed AML who are 75 years or older, or who are between the ages of 18 and 74 who have comorbidities that preclude the use of intensive induction chemotherapy.28 The BIA was undertaken from the perspective of the public health care payer in the Canadian setting (excluding Quebec) over a 3-year time horizon.28 In the reference scenario, the sponsor assumed that these patients would be eligible to receive either azacytidine monotherapy, or LDAC. In the new drug scenario, (VEN-AZA) was assumed to displace market share from azacitidine monotherapy.28

By leveraging data from multiple sources in the literature and assumptions based on clinical expert input, the sponsor estimated the eligible population size using an epidemiological approach. Only drug-acquisition costs were considered, and no drug wastage was assumed for azacitidine monotherapy and LDAC.28

Key inputs to the BIA are documented in Table 18.

Table 18: Summary of Key Model Parameters

Parameter

Sponsor’s estimate (reported as year 1 / year 2 / year 3 if appropriate)

Target population

Incidence

0.004%

Proportion ineligible for induction chemotherapy

50%

Percentage of patients aged less than 65 years

12%

Percentage of patients aged less than 65 years, covered by public drug plans

58.9%

Percentage of patients aged 65 years and over

88%

Percentage of patients aged 65 years and over, covered by public drug plans

100%

Number of patients eligible for drug under review

544 / 552 / 559

Market Uptake (3 years)

Uptake (reference scenario)

Azacitidine monotherapy

LDAC

BSC

Other

83.8% / 83.8% / 83.8%

9.5% / 9.5% / 9.5%

4.8% / 4.8% / 4.8%

1.9% / 1.9% / 1.9%

Uptake (new drug scenario)

VEN + AZA Azacitidine monotherapy

LDAC

BSC

Other

20.0% / 40.0% / 55.0%

63.8% / 43.8% / 28.8%

9.5% / 9.5% / 9.5%

4.8% / 4.8% / 4.8%

1.9% / 1.9% / 1.9%

Cost of treatment (per patient)

Cost of treatment per treatment coursea

Venetoclax plus azacitidine

Azacitidine monotherapy

LDAC

BSC

Other

$84,008.40

$42,735.06

$356.53

$0

$0

BSC = best supportive care; LDAC = low-dose cytarabine.

aBased on mean number of treatment cycles, as per the sponsor’s base case.28

Summary of the Sponsor’s BIA Results

Results of the sponsor’s base-case analysis suggested that venetoclax in combination with azacitidine (VEN + AZA) in patients with newly diagnosed AML who are 75 years or older, or who are between the ages of 18 and 74 who have comorbidities that preclude the use of intensive induction chemotherapy would result in incremental costs of $11,367,049 in year 1, $23,043,115 in year 2, $32,114,958 in year 3, for a total incremental cost of $66,525,123 over the 3-year time horizon.28

CADTH Appraisal of the Sponsor’s BIA

CADTH identified several key limitations to the sponsor’s analysis that have notable implications on the results of the BIA:

CADTH Reanalyses of the BIA

A table noting the changes made to the sponsor’s BIA as part of CADTH’s reanalysis is available in Table 19.

Table 19: CADTH Revisions to the Submitted BIA

Stepped analysis

Sponsor’s value or assumption

CADTH value or assumption

Corrections to sponsor’s base case

None

Changes to derive the CADTH base case

1. Market share estimates in the new drug scenario (across years 1 to 3)

VEN-AZA: 20% / 40% / 55%

AZA: 63.8% / 43.8% / 28.8%

LDAC: 9.5% / 9.5% / 9.5%

BSC: 4.8% / 4.8% / 4.8%

Other: 1.9% / 1.9% / 1.9%

VEN + AZA: 40% / 50% / 75%

AZA: 45.7% / 35.7% / 10.7%

LDAC: 9.5% / 9.5% / 9.5%

BSC: 4.8% / 4.8% / 4.8%

Other: 0.0% / 0.0% / 0.0%

2. Approach to derive market size

Proportion of newly diagnosed patients ineligible for induction chemotherapy = 50%

Proportion of newly diagnosed patients ineligible for induction chemotherapy = 40%

3. Alignment of drug cost inputs

a. Lower cost of LDAC = $6.75 per mL (20 mg/mL in 5 mL vial)

b. Drug wastage = excluded

c. Daily dose of LDAC = 100 mg/m2

d. Time on treatment based on the mean treatment duration

a. Cost of LDAC = $76.85 per vial (or $15.37 per mL [100 mg/mL in 5 mL vial])

b. Drug wastage = included

c. Daily dose of LDAC = 20 mg/m2

d. Time on treatment revised to reflect the median treatment duration

CADTH base case

Reanalysis 1 + 2 + 3

AZA = azacitidine; BSC = best supportive care; LDAC = low-dose cytarabine.

The results of the CADTH stepwise reanalysis are presented in summary format in Table 20 and Table 21.

Table 20: Summary of the CADTH Reanalyses of the BIA

Stepped analysis

Three-year total ($)

Submitted base case

66,525,123

CADTH reanalysis 1

96,678,220

CADTH reanalysis 2

53,220,098

CADTH reanalysis 3

60,204,555

CADTH base case

70,006,541

BIA = budget impact analysis.

CADTH also conducted additional scenario analyses to address the remaining uncertainty regarding the potential size of the eligible population:

  1. Assumed fewer patients less than the age of 65 years may be eligible for public drug plan coverage by decreasing the proportion by (a) 10% and (b) 25%.

  2. Assumed that (a) 30% and (b) 50% of newly diagnosed AML patients may be ineligible for induction chemotherapy.

  3. Explored the impact of varying the estimated market size by +/− 10%.

  4. Assumed that the treatment duration was reflected by the mean time on treatment to calculate drug-acquisition costs.

  5. Applied a price reduction of 72% for venetoclax and a price reduction of 72% for azacitidine.

Table 21: Detailed Breakdown of the CADTH Reanalyses of the BIA

Stepped analysis

Scenario

Year 0 (current situation) ($)

Year 1 ($)

Year 2 ($)

Year 3 ($)

Three-year total ($)

Submitted base case

Reference

19,248,209

19,509,844

19,775,036

20,043,831

59,328,711

New drug

19,248,209

30,876,893

42,818,151

52,158,790

125,853,834

Budget impact

0

11,367,049

23,043,115

32,114,958

66,525,123

CADTH base case

Reference

14,156,118

14,348,538

14,543,573

14,741,259

43,633,370

New drug

14,156,118

31,132,602

35,726,534

46,780,775

113,639,911

Budget impact

0

16,784,064

21,182,961

32,039,516

70,006,541

CADTH scenario analysis 1a

Reference

13,977,486

14,167,477

14,360,051

14,555,243

43,082,772

New drug

13,977,486

30,739,748

35,275,710

46,190,461

112,205,919

Budget impact

0

16,572,270

20,915,659

31,635,218

69,123,147

CADTH scenario analysis 1b

Reference

13,709,486

13,895,835

14,084,717

14,276,166

42,256,718

New drug

13,709,486

30,150,354

34,599,346

45,304,821

110,054,521

Budget impact

0

16,254,519

20,514,629

31,028,655

67,797,803

CADTH scenario analysis 2a

Reference

10,617,089

10,761,403

10,907,680

11,055,944

32,725,027

New drug

10,617,089

23,349,451

26,794,901

35,085,581

85,229,933

Budget impact

0

12,588,048

15,887,221

24,029,637

52,504,906

CADTH scenario analysis 2b

Reference

17,695,148

17,935,672

18,179,466

18,426,574

54,541,712

New drug

17,695,148

38,915,752

44,658,168

58,475,969

142,049,889

Budget impact

0

20,980,080

26,478,702

40,049,395

87,508,176

CADTH scenario analysis 3 ( + 10%)

Reference

14,156,118

15,783,392

15,997,930

16,215,385

47,996,707

New drug

14,156,118

34,245,862

39,299,188

51,458,852

125,003,902

Budget impact

0

18,462,470

23,301,257

35,243,467

77,007,195

CADTH scenario analysis 4 (−10%)

Reference

14,156,118

12,913,684

13,089,216

13,267,133

39,270,033

New drug

14,156,118

28,019,342

32,153,881

42,102,697

102,275,920

Budget impact

0

15,105,658

19,064,665

28,835,564

63,005,887

CADTH scenario analysis 5

Reference

22,103,496

22,403,942

22,708,472

23,017,141

68,129,555

New drug

22,103,496

32,259,906

42,688,338

50,862,879

125,811,122

Budget impact

0

9,855,964

19,979,866

27,845,738

57,681,568

CADTH scenario analysis

Reference

14,156,118

14,348,538

14,543,573

14,741,259

43,633,370

New drug

14,156,118

14,371,483

14,490,350

14,493,514

43,355,347

Budget impact

0

22,945

−53,223

−247,745

−278,023

BIA = budget impact analysis.