Abstract
Multiple myeloma (MM) is an incurable plasma cell malignancy that, despite an unprecedented increase in overall survival, lacks truly risk-adapted or targeted treatments. A proportion of patients with MM depend on BCL-2 for survival, and, recently, the BCL-2 antagonist venetoclax has shown clinical efficacy and safety in t(11;14) and BCL-2 overexpressing MM. However, only a small proportion of MM patients rely on BCL-2 (approx. 20%), and there is a need to broaden the patient population outside of t(11;14) that can be treated with venetoclax. Therefore, we took an unbiased screening approach and screened epigenetic modifiers to enhance venetoclax sensitivity in 2 non-BCL-2 dependent MM cell lines. The demethylase inhibitor 5-azacytidine was one of the lead hits from the screen, and the enhanced cell killing of the combination was confirmed in additional MM cell lines. Using dynamic BH3 profiling and immunoprecipitations, we identified the potential mechanism of synergy is due to increased NOXA expression, through the integrated stress response. Knockdown of PMAIP1 or PKR partially rescues cell death of the venetoclax and 5-azacytidine combination treatment. The addition of a steroid to the combination treatment did not enhance the cell death, and, interestingly, we found enhanced death of the immune cells with steroid addition, suggesting that a steroid-sparing regimen may be more beneficial in MM. Lastly, we show for the first time in primary MM patient samples that 5-azacytidine enhances the response to venetoclax ex vivo across diverse anti-apoptotic dependencies (BCL-2 or MCL-1) and diverse cytogenetic backgrounds. Overall, our data identify 5-azacytidine and venetoclax as an effective treatment combination, which could be a tolerable steroid-sparing regimen, particularly for elderly MM patients.
Introduction
The median age of onset of multiple myeloma (MM), an aggressive cancer of terminally differentiated plasma cells, is approximately 69 years of age.1 While triplet and quadruplet combinations of novel therapies, including immunomodulatory agents and cellular therapies, have improved the therapeutic landscape for MM, outcomes remain suboptimal. Survival rates vary by age and cytogenetic status.2 Patients eligible for autologous stem cell transplant (ASCT) have a median overall survival (OS) of eight years,3 while for patients >75 years, this falls to five years.4 Therefore, there is an urgent need to identify tolerable targeted agents for the treatment of elderly MM patients.
Reliance on the anti-apoptotic proteins BCL-2, MCL-1 and BCL-XL has emerged as a vulnerability for MM cells.5 The anti-apoptotic BCL-2 proteins (BCL-2, MCL-1, BCL-XL, BFL-1) function by binding and inhibiting the activity of pro-apoptotic proteins (BIM, BAX, etc.).6,7 Due to their function in inhibiting cell death, the anti-apoptotic proteins are attractive therapeutic targets. Previously, it was thought that the pro-survival protein MCL-1 was the main anti-apoptotic protein maintaining the viability of MM cells.8 However, selective MCL-1 inhibitors (AMG-176, S68345) are associated with potential cardiac toxicity, therefore limiting their clinical utility.9,10 The selective BCL-2 inhibitor, ABT-199 / venetoclax (ven), is the first BH3 mimetic to be approved by the Food and Drug Administration (FDA) for the treatment of chronic lymphocytic leukemia (CLL) and acute myeloid leukemia (AML) in combination with a demethylating agent.11-13
BCL-2 dependence in MM was initially identified in a subset of patients, characterized by the presence of a translocation in cyclin D1 (CCND1) t(11;14).14 This was confirmed across a panel of MM cell lines and primary MM samples; sensitivity to Ven was associated with t(11;14) and correlated with BH3 profiling.15 In the phase III BELLINI trial evaluating the combination of Ven, dexamethasone (Dex) and bortezomib in relapsed/refractory (R/R) MM, the patients with t(11;14) translocation were particularly sensitive.16 However, a proportion of patients without the t(11;14) translocation were also sensitive to Ven, specifically those with high expression of BCL-2.17 This highlights the need to identify combination treatments that could induce BCL-2 dependence in MM and broaden the patient population that could be treated with Ven, as only 16-24% of patients have a t(11;14) translocation.18 Venetoclax is FDA-approved for the treatment of AML in combination with the demethylating agent 5-azacytidine (5-Aza).13 5-Aza is a cytosine analog that is incorporated into both DNA and RNA, where it acts as an epigenetic modifier inhibiting DNA methyltransferases, resulting in hypomethylation.19 In AML, Ven in combination with 5-Aza is well tolerated in elderly patients who were ineligible for intensive induction therapy.13,20 Previously, it was shown that methylation regulated BCL-2 expression in mixed lineage leukemia (MLL).21 Whole exome sequencing analysis of MM revealed mutations in a series of epigenetic modifier genes, including UTX, MLL, MLL2, and HOX9.22 This led us to hypothesize that epigenetic modifiers may induce BCL-2 dependence in MM. Our aim was to perform an unbiased screening approach to identify epigenetic modifiers that could induce BCL-2 dependence in MM cells, thereby sensitizing MM cells to Ven, to expand the cohort of patients that could be treated with Ven.
Methods
For a more detailed description of the materials and methods used in the present study, see the Online Supplementary Appendix.
CD138+ cell isolation from primary multiple myeloma bone marrow samples
Primary MM samples were attained by informed consent from patients from Beaumont Hospital, Dublin, Ireland. Ethical approval was granted from the Beaumont Hospital and the RCSI ethics committee (Study N 19/32). For detailed methods of isolation, see the Online Supplementary Appendix.
Cell culture
Multiple myeloma cell lines (JJN3, KMS18, RPMI-8226, H929, MM1S, KMS27 and U266) were cultured in RPMI 1640 medium supplemented with 10% fetal bovine serum; KMS-12BM is supplemented with 20% fetal bovine serum (Gibco, Invitrogen, Carlsbad, CA, USA), 1% L-Glutamine (2 mM) (Gibco, Invitrogen, Carlsbad, CA, USA), and 1% v/v penicillin/ streptomycin (50 units/mL) (Gibco, Invitrogen, Carlsbad, CA, USA). Primary MM bone marrow stroma cells (MM-BMSC) were cultured in DMEM medium supplemented with 10% fetal bovine serum (Gibco, Invitrogen, Carlsbad, CA, USA), 1% L-Glutamine (2 mM) (Gibco, Invitrogen, Carlsbad, CA, USA), and 1% v/v penicillin/streptomycin (50 units/mL) (Gibco, Invitrogen, Carlsbad, CA, USA).
Dynamic BH3 profiling
For BH3 profiling, JJN3 cells were seeded at 3x106 cells in a T25 flask for 24 hours (hr), cells were treated with 3 μM 5-Aza for 20 hr. Cells were resuspended in 100 μM of buffer (1% FBS 0.4% EDTA [2 mM] in PBS, final pH 7.4) and incubated with a panel of BH3 peptides for 60 min at 21oC (Table 1). Details of the methods are provided in the Online Supplementary Appendix.
Statistical analysis
GraphPad Prism 9.0 software was used for all statistical analyses. Dose-response curves and IC50 values were calculated using linear regression curve fit (Log inhibitor vs. normalized response, variable slope). Unless otherwise stated, results are expressed as the mean + Standard Error of the Mean (SEM) of 3 independent experiments. Webb’s fractional product method was used to calculate the synergistic activity of pairs of drugs / inhibitors denoted A and B on cell viability using the following equation:
where CI >1.0 indicates antagonistic activity
CI = 1.0 indicates additive interactions
CI <0.8 indicates strong synergy
Results
Epigenetic modifier screen to induce BCL-2 dependence and sensitivity to venetoclax
Taking an unbiased screening approach, we screened 21 epigenetic modifiers in 2 non-BCL-2 dependent MM cell lines to try and induce BCL-2 dependency and enhance sensitivity to Ven. We confirmed that JJN3 mainly relied on MCL-1 for survival, as they were most sensitive to AMG-176, a BH3 mimetic that selectively inhibits MCL-1.23 In the BH3 profile, the A12 BH3 peptide that antagonizes MCL-124 caused a large reduction in mitochondrial membrane potential (Figure 1A).25 The KMS18 cell line was mixed BCL-2- / BCL-XL-dependent, as evidenced by sensitivity to ABT-263 that antagonizes BCL-2, BCL-XL, and BCL-w,26 and the BH3 peptides BAD and HRK induced loss of mitochondrial membrane potential (Figure 1B).27 Table 2 contains the list of the epigenetic compounds that were screened in both the KMS18 and JJN3 cell lines using CellTiter-Glo®. The classes of epigenetic modifiers included histone deacetylase inhibitors (HDACi), histone methyltransferase inhibitors (HMTi), DNA methyltransferase inhibitors (DNMTi), histone acetylase inhibitors (HATi), and bromodomain and extra terminal inhibitors (BETi) (Table 2). The 21 epigenetic modifier drugs were screened alone or in combination with Ven in the 2 MM cell lines, at 2 doses: 100 and 500 nM (Figure 1C, D). Based on the initial screen, the top 12 hits were assessed for an ability to induce cell death, as assessed by Annexin V / propium iodide (PI), in combination with Ven in the JJN3 cell line (Online Supplementary Figure S2) and the KMS-18 cell line (Online Supplementary Figure S3). The hits were ranked based on changes in the area under the curve (AUC) ratio of the epigenetic modifier alone versus the epigenetic modifier and Ven (Figure 1E). The lead hits from the cell death analysis in both the JJN3 and KMS18 cell lines were the histone methyltransferase inhibitors (JIB-04 and GSK-J4), the DNA methyltransferase inhibitor (5-aza), and the HDAC inhibitor (panobinostat) (Figure 1F, G). However, as evidenced by the dose-response curves (Online Supplementary Figures S2, S3), the top 4 hits all showed similar enhanced cell death and changes in IC50 values upon combination treatment.
5-azacytidine enhanced venetoclax-induced cell death even in the presence of bone marrow stromal cells
While the lead hits from the screen were the histone demethylase inhibitors, those compounds have only been assessed pre-clinically. Excitingly, the combination of 5-Aza and Ven has not been previously tested in MM; it has been shown to have acceptable tolerability in elderly AML patients and is FDA-approved as a combination treatment.13 As shown in the JJN3 cells, while Ven caused very little cell death alone (IC50 14.2 μM), when combined with 5-Aza, there is a log fold change in IC50 value (IC50 0.4 μM) (Figure 2A). This is also evident from live cell imaging of the JJN3 cells, with enhanced cell killing in the combination treatment and loss of mitochondrial staining with PK Mito Orange (PKMO)28 (Figure 2B). Interestingly, although Ven did not induce cell death, it did cause a phenotypic change to the mitochondrial structure, as determined by PKMO staining. Next, we assessed the combination of Ven + 5-Aza in a panel of 5 additional MM cell lines (Online Supplementary Figure S4A-F). Remarkably, across the panel of cell lines tested, 5-Aza significantly altered the IC50 value when combined with Ven in all 5 cell lines (Figure 2C). The KMS27 cell line was most sensitive to Ven treatment alone, probably because this cell line has a t(11;14) translocation, but the addition of 5-Aza reduced the IC50 by half. The combination index score for synergy is represented as a heatmap in Online Supplementary Figure S4G, H. We also assessed the reverse combination measuring the effect of Ven treatment on a 5-Aza dose-response curve, with 4/5 of the cell lines showing major changes in IC50 values upon the addition of Ven (Online Supplementary Figure S4J-N), with combination index scores of synergy graphed as a heatmap (Online Supplementary Figure S4 O). The complex interplay between MM cells and the bone marrow microenvironment has been shown to influence cell behavior, particularly MM survival and drug resistance.29
To assess the impact of bone marrow stroma cells on the 5-Aza and Ven combination, we performed co-culture experiments using HS-5, a bone marrow stroma cell line, and primary MM bone marrow stromal cells (MM-BMSC) (Figure 2D). Surprisingly, co-culture of JJN3 cells with HS-5 cells conferred significant increase in cell death with 5-Aza and Ven, compared to JJN3 cells alone (Figure 2E). Of interest, when MM1S cells were co-cultured with HS-5 cells, they were slightly less sensitive to the 5-Aza and Ven combination (Figure 2F). Next, we co-cultured JJN3 cells with MM-BMSC isolated from a MM patient bone marrow sample (Figure 2G). The JJN3 cells were still sensitive to 5-Aza and Ven combination when co-cultured with the MM-BMSC. In summary, while the MM1S cells benefit from a slight protective effect and the JJN3 cells are slightly sensitized following co-culture with the HS-5 cells, 5-Aza and Ven is still an effective treatment combination in the presence of bone marrow stroma cells across multiple cell lines with diverse anti-apoptotic dependencies.
5-azacytidine induces NOXA through the integrated stress response antagonizing MCL-1, sensitizing multiple myeloma cells to venetoclax treatment
To understand the mechanism of action of how 5-Aza may enhance sensitivity to Ven in MM cell lines, a series of approaches were taken. Firstly, we used dynamic BH3 profiling to determine changes in BH3 peptide-induced cytochrome c release from mitochondria, following 5-Aza treatment (Figure 3A). As is clear from the BH3 profile, there was a statistically significant increase in the amount of cytochrome c released following the addition of the A12 BH3 peptide, that selectively binds MCL-1, following treatment with 5-Aza. In addition, direct Ven treatment, as part of the BH3 profile on permeabilized cells, caused a statistically significant increase in the release of cytochrome c, following 5-Aza treatment in the JJN3 cells (Figure 3A). Next, we confirmed the dynamic BH3 profile by assessing the pro-apoptotic proteins that might be interacting with MCL-1 using immunoprecipitation. Following 5-Aza treatment, there was an increase in the amount of both BIM EL and NOXA bound to MCL-1 (Figure 3B). Previously, it was shown in AML cells, that 5-Aza induced NOXA by the integrated stress response.30 Indeed, we found a similar induction of NOXA following 5-Aza treatment in the JJN3 cells, which was even greater in the combination of 5-Aza and Ven (Figure 3C). We also assessed if this induction of NOXA was dependent on the integrated stress response. Pre-treatment with ISRIB, which blocks the integrated stress response by targeting eIF1B,31 also blocked the induction of the NOXA protein following the combination treatment (Figure 3C). Ven alone induces a small amount of NOXA protein, but it enhances the amount of NOXA induced by 5-Aza (Figure 3C). We confirmed that ISRIB did not alter eIF2α phosphorylation but prevented the induction of activating transcription factor 4 (ATF-4) by 5-Aza and 5-Aza + Ven (Figure 3D). Confirming the importance of the integrated stress response in the cell death induced by Ven + 5-Aza, there was a significant reduction in cell death upon pre-treatment with ISRIB (Figure 3E). There is evidence in the literature that 5-Aza can induce dsRNA, through reactivation of endogenous retrovirus.32 Therefore, we assessed the amount of dsRNA in JJN3 cells treated with 5-Aza, Ven or the combination (Figure 4A). While all treatments induced some dsRNA, the combination of Ven + 5-Aza induced the most significant amount, as assessed by pixel count (Figure 4B). Next, we assessed if the knockdown of the dsRNA sensor PKR, which can activate the integrated stress response, could protect from the cell death induced by the combination treatment. Knockdown of PKR significantly reduced the cell death induced by the combination treatment (Figure 4C). The densitometry on PKR knockdown is shown in Online Supplementary Figure S5C. Validating the importance of NOXA in the cell death mechanism, siRNA knockdown of PMAIP1 (NOXA gene), also significantly protected against the combination induced cell death (Figure 4D). Similarly, densitometry for NOXA knockdown is shown in Online Supplementary Figure S5D. A proposed model is shown in Figure 4E. Combined, the data suggest the mechanism of synergy following 5-Aza + Ven is due, at least in part, to an increase in dsRNA, activating PKR, activating the integrated stress response, with an increase in eIF2α phosphorylation, inducing NOXA protein that antagonizes MCL-1. By antagonizing MCL-1, the cells are more sensitive to Ven (Figure 4E).
Assessing if a steroid enhances the cell death induced by 5-azacytidine and venetoclax in multiple myeloma and immune cells
It is standard practice for a steroid, such as Dex, to be added to a combination treatment regimen in MM.33 Therefore, we assessed the combination of Ven, 5-Aza and Dex in a panel of MM cells. Interestingly, we did not find any significant increase in cell death upon the addition of 1 μM Dex to the combination of Ven and 5-Aza, across 3 different MM cell lines (Figure 5A-C). Indeed, the combination of Ven and 5-Aza caused significantly more cell death than Dex (1 μM) and Ven in both the JJN3 and MM1S cell lines (Figure 5A, B). Next, we measured the effect of the combination treatment on immune cells, Ven has been shown to induce cell death in B34 and T cells.35 Using peripheral blood mononuclear cells from healthy donors, we treated the cells ex vivo for 16 hr and measured cell death of CD3+ T cells by annexin V/PI staining (Figure 5D). In the CD3+ T cells, 5-Aza caused a minimal amount of cell death alone. Ven induced some cell death in CD3+ T cells, as was found previously in mouse CD8+ T cells,35 and additive cell death was seen with the combination treatment of 5-Aza and Ven (Figure 5E). The combination of Dex and Ven caused slightly more cell death, which was significant, than Ven and 5-Aza (Figure 5E). Dex combined with Ven caused a dose-dependent increase in T-cell death (Figure 5F). Potentially, the reason for this enhanced cell death is due to Dex inducing a BCL-2 expression in T cells. There was a dose-dependent increase in BCL-2 expression following Dex treatment, as measured by intercellular BCL-2 staining by flow cytometry (Figure 5G, H). Our data potentially suggest that the addition of a steroid may induce more cell death in the immune cells while having little effect on the tumor cell killing, at least ex vivo. Based on this result, there is a potential to use a steroid-sparing regimen to test the combination of Ven/5-Aza in the clinic. This steroid effect on T cells may also have implications for the efficacy of many other therapies in MM.
Venetoclax and 5-azacytidine show enhanced killing of primary multiple myeloma samples irrespective of stage or cytogenetics
As the combination treatment had only been assessed in MM cell lines, we next wanted to confirm our findings that the combination of 5-Aza and Ven was effective in heterogeneous MM patient samples. Following isolation of CD138+ patients’ cells, the cells were seeded and treated with either increasing doses of BH3 mimetics for 16 hr or the combination of 5-Aza and Ven and cell death was measured using Annexin V/PI staining on the flow cytometer (Figure 6A, Online Supplementary Figures S6A-K, S7A-K). Of the 11 primary MM bone marrow samples analyzed ex vivo (Figure 6A), 8 were non-t(11;14) patients (Online Supplementary Table S1). The combination of 5-Aza and Ven showed enhanced cell killing in 6 out of the 8 non-t(11;14) (Figure 6B) and 2 out of 3 t(11;14) samples (Figure 6C), with 6 samples showing a CI score of less than 1 (Online Supplementary Figure S6A-K). Patient cytogenetics were not known at the time of Ven treatment. The dose of Ven used induced substantial cell death alone in the t(11;14) patient samples, with a statistical difference in the ex vivo sensitivity of non-t(11;14) samples versus the t(11;14) samples (Figure 6D). These data highlight the use of BH3 mimetic profiling as a biomarker to identify patients who may respond to Ven. There was a diverse anti-apoptotic dependence across the MM patient sample (Online Supplementary Figure S6). Four of the primary samples CD138+ patient cells were more BCL-2 dependent, 4 were more MCL-1 dependent, and 3 samples were a mix, with the BCL-XL dependent samples showing the lowest cell death following combination treatment (Figure 6E). Lastly, there was no statistical difference in the combination treatment of Ven and 5-Aza in upfront versus relapsed patient samples (Figure 6F) or in patients with normal or altered cytogenetics (Figure 6G). Importantly, here we have shown for the first time that 5-Aza and Ven are effective at killing primary MM samples ex vivo, over-coming high-risk cytogenetic features and multiple lines of prior therapies.
Discussion
Multiple myeloma is a disease with a diverse genetic background and, to date, the development of molecularly targeted therapies has proven elusive. Our aim was to take an unbiased screening approach to identify epigenetic modifiers that could induce BCL-2 dependence in MM cells to broaden the patient population that could benefit from treatment with Ven.15 The DNA methyltransferase inhibitor 5-Aza emerged as one of the top hits from our screen. Ven in combination with 5-Aza has never been tested in MM, even though it is an approved combination regimen in AML.36
We identified a mechanism of synergism by which 5-Aza in combination with Ven induces NOXA protein inhibiting MCL-1, sensitizing MM cells to Ven. The induction of NOXA is reliant on the integrated stress response, with evidence of phosphorylation of eIF2-α. A similar mechanism was identified for 5-Aza in AML, ATF4 was responsible for inducing NOXA expression.30 Importantly, we demonstrated that PMAIP1 (NOXA) knockdown partially rescued the cell death induced by the combination treatment, as did inhibition of the integrated stress response with ISRIB. We also showed for the first time, the translational potential of 5-Aza and Ven in combination, which demonstrated strong cell killing effect in primary CD138+ MM cells ex vivo, irrespective of cytogenetic background.
In the case of AML, either 5-Aza or decitabine were both effective at enhancing the response to Ven treatment.13,20 In contrast, in our study, while 5-Aza enhanced the cell death induced by Ven, decitabine did not cause a similar enhanced cell death. Decitabine is a deoxyribonucleotide and is incorporated into DNA, whereas 5-Aza is a ribonucleoside and is incorporated into both DNA and RNA.19,37 Potentially, this difference could explain why 5-Aza enhances cell death in MM and may suggest the integration into mRNA may lead to activation of the integrated stress response. A series of studies have also shown evidence that 5-Aza acts through activation of endogenous retroviral elements (ERV), inducing viral mimicry and activating the dsRNA sensing pathways (e.g., MDA5, TLR3).32,38 We showed that the combination treatment induced dsRNA in the cells, and that knockdown of the ds-RNA sensor PKR partially rescued the cell death induced by the combination. We did not investigate if the dsRNA also activates RIG-1 to signal through the mitochondrial antiviral-signaling protein (MAVS), to induce type 1 interferons and proinflammatory cytokines.32,39 The combination of Ven + 5-Aza works very effectively in the clinic in AML.13,36 Activation of the dsRNA pathway by the combination treatment, as shown in our data, could also activate an immune response; this could potentially play an important role in the mechanism of action in vivo. However, this was not assessed in this study. Steroid utilization in MM is standard practice.40 Therefore, we assessed if the addition of the steroid Dex enhanced the cell death induced by the combination treatment (Ven + 5-Aza). We did not detect any enhanced killing in 3 different MM cell lines, with diverse anti-apoptotic dependence, following the addition of Dex to the combination. The combination of 5-Aza + Ven was more effective at killing than Dex + Ven, which was previously identified as an effective combination in MM.41,42 Potentially, our data may help to explain the latest phase III CANOVA trial data with Ven and Dex in t(11;14) patients, which did not reach a significant endpoint in comparison to pomalidomide + Dex, despite evidence of longer progression-free survival.43 Next, we assessed the combination on immune cell killing ex vivo on T cells, as it was previously identified that mature T cells express high BCL-22 7, 4 4 and are sensitive to Ven treatment alone.35,45
We found a similar induction of cell death in human CD3 T cells with Ven treatment alone, and the addition of 5-Aza did enhance cell killing. However, Dex + Ven caused significantly more death of the T cells than 5-Aza + Ven, and Dex induced BCL-2 expression in the CD3 cells. These data would suggest that a steroid has little therapeutic benefit, as it did not enhance the MM cell killing; but it may enhance the killing of immune cells, potentially limiting the capacity of immune cells to kill tumor cells.17 In a randomized trial comparing lenalidomide with high-dose versus low-dose Dex, the low-dose Dex regimen was associated with better short-term overall survival.46 Indeed, a high-dose Dex regimen was associated with toxicities, particularly in frail, elderly patients. In a recent abstract, the results of a phase III trial comparing daratumumab and lenalidomide without longterm Dex to lenalidomide and Dex were described.47 This trial showed that the overall response rates were higher in the Dex-sparing regimen. This study highlighted that there is a case to not add Dex to MM regimens, particularly in frail, elderly patients with MM, in the situation where the patient is receiving highly effective therapy.
To assess the translational relevance of 5-Aza and Ven, we screened the combination in a panel of primary MM CD138+ cells from MM patients, with different cytogenetic backgrounds. In total, 11 patient samples were assessed: 4 were MCL-1 dependent, 3 were mixed anti-apoptotic dependent, and 4 samples were BCL-2 dependent. The 2 samples that did not respond to the combination treatment had a higher sensitivity to the BCL-XL antagonist. Given the mechanism of the synergy described, induction of the NOXA protein binding to MCL-1 could potentially explain the lack of response in these 2 patient samples. Of the 4 samples that were BCL-2 dependent, it was later revealed by cytogenetics that 3 had t(11;14) translocation. Gomez-Bougie et al. looked at the anti-apoptotic dependence in MM samples.48 They saw that BCL-2 dependence was mostly found in the t(11;14) subgroup and in 35% of the samples there was a co-dependency on either BCL-2/MCL-1 or BCL-XL/MCL-1. While we only assessed a small cohort of samples, the results corroborated previous studies, confirming that there are diverse anti-apoptotic dependencies in MM primary samples.25 In our study, we used patient-specific sensitivity measurements by carrying out ex vivo functional cell death measurements of primary MM samples treated with BH3 mimetics to determine the anti-apoptotic dependence. Our group has previously identified BCL-2 dependence in a patient with secondary plasma cell leukemia.49 BH3 profiling can also be used to identify resistance mechanisms to Ven, including switching of anti-apoptotic dependence.50-53 An important study by Gupta et al. used ex vivo assessment of MM patients’ samples to Ven as an indicator of PFS.54 As we have also shown here, assays such as precision testing and BH3 profiling are dynamic and give a functional measurement of cellular response to treatment; as such, they can be useful for identifying sensitive patients a priori.
Collectively, our data identify 5-Aza and Ven as an effective therapeutic combination for the treatment of MM, including in patients with non-t(11;14) disease with high-risk cytogenetics, such as 17p and 1q amplification. This combination has the potential to be effective across a broad patient population and should be considered in a steroid-free regimen for assessment in a clinical trial setting.
Footnotes
- Received June 29, 2023
- Accepted March 8, 2024
Correspondence
Disclosures
TNC received research funding from AbbVie. JQ has received Honoraria from Janssen and Takeda, and travel funding from Roche. MOD has no competing interest but has employment and equity in ONK Therapeutics. All the other authors have no conflicts of interest to disclose.
Funding
The authors would like to thank the following funding agencies for their support: the Leukemia Research Foundation (LRF-RCSI-2020) and Science Foundation Ireland (19/FFP/6461) (to TNC) and Irish Research Council (IRCLA/2022/2822) Breakthrough Cancer Research (BCR-RCSI-2020) (to LF).
References
- Cancer Factsheet Multiple Myeloma (ICD-10 C90) NCRI. 2023. Publisher Full TextGoogle Scholar
- Rajkumar SV. Multiple myeloma: 2020 update on diagnosis, risk-stratification and management. Am J Hematol. 2020; 95(5):548-567. Google Scholar
- Landgren O, Weiss BM. Patterns of monoclonal gammopathy of undetermined significance and multiple myeloma in various ethnic / racial groups: support for genetic factors in pathogenesis. Leukemia. 2009; 23(10):1691-1697. Google Scholar
- Moreau P, Attal M, Facon T. Frontline therapy of multiple myeloma. Blood. 2015; 125(20):3076-3084. Google Scholar
- Maji S, Panda S, Samal SK. Bcl-2 antiapoptotic family proteins and chemoresistance in cancer. Adv Cancer Res. 2018; 137:37-75. Google Scholar
- Ni Chonghaile T, Letai A. Mimicking the BH3 domain to kill cancer cells. Oncogene. 2008; 27(Suppl 1):S149-S157. Google Scholar
- Cory S, Adams JM. The Bcl2 family: regulators of the cellular life-or-death switch. Nat Rev Cancer. 2002; 2(9):647-656. Google Scholar
- Wuilleme-Toumi S, Robillard N, Gomez P. Mcl-1 is overexpressed in multiple myeloma and associated with relapse and shorter survival. Leukemia. 2005; 19(7):1248-1252. Google Scholar
- Thomas RL, Roberts DJ, Kubli DA. Loss of MCL-1 leads to impaired autophagy and rapid development of heart failure. Genes Dev. 2013; 27(12):1365-1377. Google Scholar
- Wang X, Bathina M, Lynch J. Deletion of MCL-1 causes lethal cardiac failure and mitochondrial dysfunction. Genes Dev. 2013; 27(12):1351-1364. Google Scholar
- Roberts AW, Davids MS, Pagel JM. Targeting BCL2 with venetoclax in relapsed chronic lymphocytic leukemia. N Engl J Med. 2016; 374(4):311-322. Google Scholar
- DiNardo CD, Stein EM, de Botton S. Durable remissions with ivosidenib in IDH1-mutated relapsed or refractory AML. N Engl J Med. 2018; 378(25):2386-2398. Google Scholar
- DiNardo CD, Pratz K, Pullarkat V. Venetoclax combined with decitabine or azacitidine in treatment-naive, elderly patients with acute myeloid leukemia. Blood. 2019; 133(1):7-17. Google Scholar
- Bodet L, Gomez-Bougie P, Touzeau C. ABT-737 is highly effective against molecular subgroups of multiple myeloma. Blood. 2011; 118(14):3901-3910. Google Scholar
- Touzeau C, Dousset C, Le Gouill S. The Bcl-2 specific BH3 mimetic ABT-199: a promising targeted therapy for t(11;14) multiple myeloma. Leukemia. 2014; 28(1):210-212. Google Scholar
- Kumar S, Kaufman JL, Gasparetto C. Efficacy of venetoclax as targeted therapy for relapsed/refractory t(11;14) multiple myeloma. Blood. 2017; 130(22):2401-2409. Google Scholar
- Kumar S, Harrison SJ, Cavo M. Final overall survival results from BELLINI, a phase 3 study of venetoclax or placebo in combination with bortezomib and dexamethasone in relapsed/ refractory multiple myeloma. Blood. 2021; 138(Suppl 1):84. Google Scholar
- Bal S, Kumar SK, Fonseca R. Multiple myeloma with t(11;14): unique biology and evolving landscape. Am J Cancer Res. 2022; 12(7):2950-2965. Google Scholar
- Stresemann C, Lyko F. Modes of action of the DNA methyltransferase inhibitors azacytidine and decitabine. Int J Cancer. 2008; 123(1):8-13. Google Scholar
- DiNardo CD, Pratz KW, Letai A. Safety and preliminary efficacy of venetoclax with decitabine or azacitidine in elderly patients with previously untreated acute myeloid leukaemia: a non-randomised, open-label, phase 1b study. Lancet Oncol. 2018; 19(2):216-228. Google Scholar
- Benito JM, Godfrey L, Kojima K. MLL-rearranged acute lymphoblastic leukemias activate BCL-2 through H3K79 methylation and are sensitive to the BCL-2-specific antagonist ABT-199. Cell Rep. 2015; 13(12):2715-2727. Google Scholar
- Palumbo A, Anderson K. Multiple myeloma. N Engl J Med. 2011; 364(11):1046-1060. Google Scholar
- Caenepeel S, Brown SP, Belmontes B. AMG 176, a selective MCL1 inhibitor, is effective in hematologic cancer models alone and in combination with established therapies. Cancer Discov. 2018; 8(12):1582-1597. Google Scholar
- Foight GW, Ryan JA, Gulla SV, Letai A, Keating AE. Designed BH3 peptides with high affinity and specificity for targeting Mcl-1 in cells. ACS Chem Biol. 2014; 9(9):1962-1968. Google Scholar
- Touzeau C, Ryan J, Guerriero J. BH3 profiling identifies heterogeneous dependency on Bcl-2 family members in multiple myeloma and predicts sensitivity to BH3 mimetics. Leukemia. 2016; 30(3):761-764. Google Scholar
- Tse C, Shoemaker AR, Adickes J. ABT-263: a potent and orally bioavailable Bcl-2 family inhibitor. Cancer Res. 2008; 68(9):3421-3428. Google Scholar
- Chonghaile TN, Roderick JE, Glenfield C. Maturation stage of T-cell acute lymphoblastic leukemia determines BCL-2 versus BCL-XL dependence and sensitivity to ABT-199. Cancer Discov. 2014; 4(9):1074-1087. Google Scholar
- Liu T, Stephan T, Chen P. Multi-color live-cell STED nanoscopy of mitochondria with a gentle inner membrane stain. Proc Natl Acad Sci U S A. 2022; 119(52):e2215799119. Google Scholar
- Vincent T, Mechti N. Extracellular matrix in bone marrow can mediate drug resistance in myeloma. Leuk Lymphoma. 2005; 46(6):803-811. Google Scholar
- Jin S, Cojocari D, Purkal JJ. 5-azacitidine induces NOXA to prime AML cells for venetoclax-mediated apoptosis. Clin Cancer Res. 2020; 26(13):3371-3383. Google Scholar
- Sidrauski C, Acosta-Alvear D, Khoutorsky A. Pharmacological brake-release of mRNA translation enhances cognitive memory. Elife. 2013; 2:e00498. Google Scholar
- Chiappinelli KB, Strissel PL, Desrichard A. Inhibiting DNA methylation causes an interferon response in cancer via dsRNA including endogenous retroviruses. Cell. 2015; 162(5):974-986. Google Scholar
- Shah UA, Mailankody S. Emerging immunotherapies in multiple myeloma. BMJ. 2020; 370:m3176. Google Scholar
- Khaw SL, Merino D, Anderson MA. Both leukaemic and normal peripheral B lymphoid cells are highly sensitive to the selective pharmacological inhibition of prosurvival Bcl-2 with ABT-199. Leukemia. 2014; 28(6):1207-1215. Google Scholar
- Ludwig LM, Hawley KM, Banks DB. Venetoclax imparts distinct cell death sensitivity and adaptivity patterns in T cells. Cell Death Dis. 2021; 12(11):1005. Google Scholar
- DiNardo CD, Rausch CR, Benton C. Clinical experience with the BCL2-inhibitor venetoclax in combination therapy for relapsed and refractory acute myeloid leukemia and related myeloid malignancies. Am J Hematol. 2018; 93(3):401-407. Google Scholar
- Yoo CB, Jones PA. Epigenetic therapy of cancer: past, present and future. Nat Rev Drug Discov. 2006; 5(1):37-50. Google Scholar
- Roulois D, Loo Yau H, Singhania R. DNA-demethylating agents target colorectal cancer cells by inducing viral mimicry by endogenous transcripts. Cell. 2015; 162(5):961-973. Google Scholar
- Patel JR, Jain A, Chou YY. ATPase-driven oligomerization of RIG-I on RNA allows optimal activation of type-I interferon. EMBO Rep. 2013; 14(9):780-787. Google Scholar
- Rajkumar SV, Kumar S. Multiple myeloma current treatment algorithms. Blood Cancer J. 2020; 10(9):94. Google Scholar
- Matulis SM, Gupta VA, Nooka AK. Dexamethasone treatment promotes Bcl-2 dependence in multiple myeloma resulting in sensitivity to venetoclax. Leukemia. 2016; 30(5):1086-1093. Google Scholar
- Matulis SM, Gupta VA, Neri P. Functional profiling of venetoclax sensitivity can predict clinical response in multiple myeloma. Leukemia. 2019; 33(5):1291-1296. Google Scholar
- AbbVie presents results from phase 3 CANOVA study of venetoclax in patients with relapsed or refractory multiple myeloma. News release. AbbVie. September 29. 2023. Publisher Full TextGoogle Scholar
- Veis DJ, Sentman CL, Bach EA, Korsmeyer SJ. Expression of the Bcl-2 protein in murine and human thymocytes and in peripheral T lymphocytes. J Immunol. 1993; 151(5):2546-2554. Google Scholar
- Rohner L, Reinhart R, Iype J. Impact of BH3-mimetics on human and mouse blood leukocytes: a comparative study. Sci Rep. 2020; 10(1):222. Google Scholar
- Rajkumar SV, Jacobus S, Callander NS. Lenalidomide plus high-dose dexamethasone versus lenalidomide plus low-dose dexamethasone as initial therapy for newly diagnosed multiple myeloma: an open-label randomised controlled trial. Lancet Oncol. 2010; 11(1):29-37. Google Scholar
- Manier S, Corre J, Hulin C. A dexamethasone-sparing regimen with daratumumab and lenalidomide in frail patients with newly-diagnosed multiple myeloma: efficacy and safety analysis of the phase 3 IFM2017-03 trial. Blood. 2022; 140(Suppl 1):1369. Google Scholar
- Gomez-Bougie P, Maiga S, Tessoulin B. BH3-mimetic toolkit guides the respective use of BCL2 and MCL1 BH3-mimetics in myeloma treatment. Blood. 2018; 132(25):2656-2669. Google Scholar
- Glavey SV, Flanagan L, Bleach R. Secondary plasma cell leukaemia treated with single agent venetoclax. Br J Haematol. 2020; 190(4):e242-e245. Google Scholar
- Di Grande A, Peirs S, Donovan PD. The spleen as a sanctuary site for residual leukemic cells following ABT-199 monotherapy in ETP-ALL. Blood Adv. 2021; 5(7):1963-1976. Google Scholar
- Sullivan GP, Flanagan L, Rodrigues DA, Ni Chonghaile T. The path to venetoclax resistance is paved with mutations, metabolism, and more. Sci Transl Med. 2022; 14(674):eabo6891. Google Scholar
- Ni Chonghaile T, Sarosiek KA, Vo TT. Pretreatment mitochondrial priming correlates with clinical response to cytotoxic chemotherapy. Science. 2011; 334(6059):1129-1133. Google Scholar
- Bhatt S, Pioso MS, Olesinski EA. Reduced mitochondrial apoptotic priming drives resistance to BH3 mimetics in acute myeloid leukemia. Cancer Cell. 2020; 38(6):872-890. Google Scholar
- Gupta VA, Matulis SM, Barwick BG. Venetoclax ex vivo functional profiling predicts improved progression-free survival. Blood Cancer J. 2022; 12(8):115. Google Scholar
Data Supplements
Figures & Tables
Article Information
This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.