Introduction: The Evolution of Precision Medicine in Multiple Myeloma
For decades, the treatment landscape for multiple myeloma (MM) has been characterized by a broad-spectrum approach, utilizing proteasome inhibitors, immunomodulatory drugs (IMiDs), and monoclonal antibodies. However, as our understanding of the molecular heterogeneity of plasma cell dyscrasias deepens, the focus has shifted toward precision oncology—identifying specific genetic drivers that can be therapeutically exploited. Among these, the t(11;14)(q13;q32) translocation stands out as a unique biological subset, occurring in approximately 15% to 20% of patients with MM. This translocation leads to the juxtaposition of the CCND1 gene with the immunoglobulin heavy chain locus, resulting in the overexpression of cyclin D1 and a distinct reliance on the antiapoptotic protein BCL-2.
Venetoclax, a first-in-class, highly selective oral BCL-2 inhibitor, has already revolutionized the treatment of chronic lymphocytic leukemia and acute myeloid leukemia. In the context of MM, preclinical and early clinical data suggested that the t(11;14) translocation serves as a biomarker for BCL-2 dependency, making these patients particularly sensitive to venetoclax-induced apoptosis. The Phase III CANOVA study was designed to rigorously test this hypothesis by comparing venetoclax-dexamethasone (Ven-Dex) against the standard-of-care pomalidomide-dexamethasone (Pom-Dex) in a molecularly defined population of patients with relapsed or refractory multiple myeloma (RRMM).
Study Rationale: Targeting BCL-2 Dependency
The biological rationale for the CANOVA study rests on the observation that t(11;14)-positive myeloma cells exhibit high levels of BCL-2 mRNA and a high BCL-2 to MCL-1/BCL-XL ratio. Unlike most myeloma cells, which depend on MCL-1 for survival, t(11;14) cells are uniquely primed for BCL-2-mediated survival. Furthermore, the addition of dexamethasone has been shown to enhance this sensitivity. Dexamethasone upregulates BCL-2 and the pro-apoptotic protein BIM, while simultaneously decreasing MCL-1 levels, thereby deepening the dependency on BCL-2 and facilitating venetoclax-induced cell death.
The CANOVA Trial: Methodology and Study Population
CANOVA (NCT03539744) was a randomized, open-label, multicenter, phase III study. It enrolled 263 adult patients with t(11;14)-positive RRMM who had received at least two prior lines of therapy, including a proteasome inhibitor and an IMiD. Patients were randomized 1:1 to receive either venetoclax (800 mg daily) plus dexamethasone (40 mg weekly, or 20 mg for those >75 years) or pomalidomide (4 mg on days 1–21 of a 28-day cycle) plus dexamethasone.
The primary end point was progression-free survival (PFS) as assessed by an independent review committee (IRC). Secondary end points included overall response rate (ORR), very good partial response (VGPR) or better rate, minimal residual disease (MRD) negativity rate (threshold of 10^-5), overall survival (OS), and safety. The study utilized a stratified log-rank test for the primary analysis, with a two-sided alpha of 0.05.
Primary Results: Progression-Free Survival Analysis
In the intention-to-treat population, the median PFS for patients receiving venetoclax-dexamethasone was 9.9 months (95% CI, 6.9 to 12.6), compared with 5.8 months (95% CI, 3.8 to 9.2) for those in the pomalidomide-dexamethasone arm. Despite the numerical difference favoring venetoclax, the hazard ratio (HR) was 0.823 (95% CI, 0.596 to 1.136), and the p-value was 0.24. Consequently, the study did not meet its primary end point of demonstrating a statistically significant improvement in PFS.
The lack of statistical significance, despite a four-month difference in median PFS, suggests several possibilities. The study might have been underpowered to detect the specific effect size observed, or the performance of the control arm (pomalidomide) in this specific cytogenetic subgroup may have been more robust than historical benchmarks predicted. Subgroup analyses did not reveal a specific cohort that derived a significantly discordant benefit, suggesting the effect was relatively uniform across the enrolled population.
Deepening Responses: Secondary Endpoints and MRD
While the primary endpoint remained elusive, the secondary endpoints provided compelling evidence of the clinical activity of venetoclax in this population. The overall response rate was significantly higher in the venetoclax-dexamethasone group at 62%, compared to 35% in the pomalidomide-dexamethasone group. Furthermore, the quality of responses was deeper with venetoclax; the rate of VGPR or better was 39% versus 14%, respectively.
Perhaps the most striking finding was the minimal residual disease (MRD) negativity rate. In the Ven-Dex arm, 8% of patients achieved MRD negativity at the 10^-5 level, whereas 0% of patients in the Pom-Dex arm reached this milestone. These data suggest that for patients who do respond to venetoclax, the responses are not only more likely to occur but are also significantly deeper than those achieved with standard IMiD-based therapy in the third-line setting.
Overall Survival Trends
At the time of the primary analysis, the median overall survival (OS) was 32.4 months (95% CI, 26.4 to 40.7) for venetoclax-dexamethasone and 26.9 months (95% CI, 20.4 to 38.9) for pomalidomide-dexamethasone. The hazard ratio for death was 0.856 (95% CI, 0.612 to 1.197). While not statistically significant, the numerical trend toward improved OS is encouraging, particularly given that patients in the control arm were permitted to receive subsequent lines of therapy that may have included other BCL-2 directed approaches or newer agents like CAR-T cell therapies and bispecific antibodies.
Safety and Tolerability Profile
The safety profile observed in CANOVA was consistent with the known toxicities of both venetoclax and pomalidomide. Interestingly, the rate of Grade 3 or higher treatment-emergent adverse events (TEAEs) was lower in the venetoclax-dexamethasone arm (67%) compared to the pomalidomide-dexamethasone arm (83%). Neutropenia and infections remained the most common high-grade toxicities associated with venetoclax.
A point of clinical concern was the mortality data. There were 16 treatment-emergent deaths (12%) in the venetoclax arm compared to 8 (6%) in the pomalidomide arm. This mirrors observations from the earlier Phase III BELLINI trial, where venetoclax was added to bortezomib and dexamethasone in an unselected RRMM population, resulting in an increased risk of fatal infections. In CANOVA, the deaths were largely attributed to disease progression or infectious complications. This underscores the necessity of vigilant infectious disease monitoring and the potential use of prophylactic antibiotics or intravenous immunoglobulin (IVIG) in patients treated with BCL-2 inhibitors.
Expert Commentary: Interpreting the CANOVA Findings
The CANOVA study presents a nuanced challenge for clinicians. On one hand, the failure to meet the primary PFS endpoint limits the path toward immediate, broad regulatory approval for venetoclax in this specific setting. On the other hand, the doubling of response rates and the achievement of MRD negativity in a subset of patients demonstrate that venetoclax is a potent tool for t(11;14)-positive disease.
From a mechanistic perspective, the data reinforce the importance of the t(11;14) biomarker. However, they also suggest that t(11;14) alone might not be sufficient to ensure long-term disease control with venetoclax-dexamethasone monotherapy. The future of venetoclax in MM likely lies in its combination with other agents, such as anti-CD38 monoclonal antibodies (daratumumab) or proteasome inhibitors, which can further sensitize cells to apoptosis.
Furthermore, the trial highlights the difficulties of conducting Phase III studies in genetically defined subgroups. As the MM treatment landscape becomes increasingly crowded with highly effective therapies, demonstrating a statistically significant PFS benefit over a potent comparator like pomalidomide becomes an increasingly high bar, especially in later lines of therapy where clonal evolution and secondary resistance mechanisms are prevalent.
Conclusion: The Path Forward for BCL-2 Inhibition
The CANOVA study confirms that venetoclax-dexamethasone is a highly active regimen for patients with t(11;14)-positive RRMM, offering superior response rates and deeper remissions compared to pomalidomide-dexamethasone. While the primary PFS objective was not met, the numerical improvements in PFS and OS, combined with a manageable (though infection-prone) safety profile, suggest that venetoclax remains a critical therapeutic consideration for this specific molecular subset.
For clinicians, the takeaway is twofold: first, the routine testing for t(11;14) by FISH at diagnosis and relapse is mandatory to identify potential candidates for BCL-2 inhibition. Second, when using venetoclax, aggressive supportive care and infection prophylaxis are paramount to mitigate the risks observed in this and previous trials. As we move toward more personalized myeloma care, the lessons from CANOVA will be instrumental in refining how we integrate targeted therapies into the broader therapeutic armamentarium.
Funding and ClinicalTrials.gov
This study was funded by AbbVie and Roche. ClinicalTrials.gov Identifier: NCT03539744.
References
1. Popat R, Beksac M, Dimopoulos MA, et al. Venetoclax-Dexamethasone Versus Pomalidomide-Dexamethasone in t(11;14)-Positive Relapsed/Refractory Multiple Myeloma: Primary Results of the Randomized, Phase III CANOVA Study. J Clin Oncol. 2026;44(3):164-175.
2. Moreau P, Chanan-Khan A, Roberts AW, et al. Promising efficacy and acceptable safety of venetoclax plus bortezomib and dexamethasone in relapsed or refractory multiple myeloma. Blood. 2017;130(22):2392-2400.
3. Kumar SK, Harrison SJ, Cavo M, et al. Venetoclax or placebo in combination with bortezomib and dexamethasone in patients with relapsed or refractory multiple myeloma (BELLINI): a randomised, double-blind, multicentre, phase 3 trial. Lancet Oncol. 2020;21(12):1630-1642.
4. Touzeau C, Maciag P, Amiot M, et al. The BCL-2 specific inhibitor venetoclax (ABT-199) sensitizes colorectal cancer cells to chemotherapies. Sci Rep. 2014;4:4706.

