Abstract
BCL-XL and BCL-2 are key anti-apoptotic proteins and validated cancer targets. 753B is a novel BCL-XL/BCL-2 proteolysis targeting chimera (PROTAC) that targets both BCL-XL and BCL-2 to the von Hippel-Lindau (VHL) E3 ligase, leading to BCLX L/BCL-2 ubiquitination and degradation selectively in cells expressing VHL. Because platelets lack VHL expression, 753B spares on-target platelet toxicity caused by the first-generation dual BCL-XL/BCL-2 inhibitor navitoclax (ABT-263). Here, we report pre-clinical single-agent activity of 753B against different leukemia subsets. 753B effectively reduced cell viability and induced dose-dependent degradation of BCL-XL and BCL-2 in a subset of hematopoietic cell lines, acute myeloid leukemia (AML) primary samples, and in vivo patient-derived xenograft AML models. We further demonstrated the senolytic activity of 753B, which enhanced the efficacy of chemotherapy by targeting chemotherapy-induced cellular senescence. These results provide a pre-clinical rationale for the utility of 753B in AML therapy, and suggest that 753B could produce an added therapeutic benefit by overcoming cellular senescence-induced chemoresistance when combined with chemotherapy.
Introduction
Acute myeloid leukemia (AML) is a hematopoietic malignancy involving clonal hematopoiesis and defects in differentiation, proliferation, and cell death, resulting in the accumulation of immature blasts, marrow failure, and rapid death of patients if not treated. Therapeutic progress has been slow despite the recent identification of genomic and epigenetic alterations. Fewer than half of adults with AML (and less than 10-20% of elderly [>60 years old] patients with AML) survive long-term. Over-expression of anti-apoptotic BCL-2 family proteins is a core oncogenic property of leukemia, and is associated with disease progression and resistance to chemotherapy by protecting tumor cells from apoptosis.1
Targeting BCL-2 family proteins has been successfully explored as a therapeutic strategy for leukemia, and several small-molecule inhibitors of BCL-2 proteins have been identified.2,3 Venetoclax (ABT-199), a BCL-2–selective inhibitor, has been approved by the US Food and Drug Administration for treatment of chronic lymphocytic leukemia (CLL) and of AML when combined with low-intensity chemotherapy. However, despite high response rates, the majority of the patients treated with hypomethylating agents and venetoclax eventually relapse.4 The upregulation of anti-apoptotic proteins other than BCL-2, such as BCL-XL or MCL-1, have been identified as major determinants of venetoclax resistance in CLL and AML,2,5-7 consistent with the high efficacy of the dual BCL-2/BCL-XL inhibitor navitoclax in killing venetoclax-resistant CLL cells.7
However, the clinical utility of navitoclax was hampered by the on-target and dose-limiting thrombocytopenia due to dependence of platelet survival on BCL-XL.8 We have previously reported that DT2216, a von Hippel-Landau (VHL)-recruiting proteolysis-targeting chimera (PROTAC) derived from navitoclax, is able to overcome this on-target thrombocytopenia.9 DT2216 selectively ubiquitinated and degraded BCL-XL in a VHL E3 ligase- and proteasomedependent manner in VHL-expressing cells, and was highly effective against tumors that primarily depend on BCL-XL for survival, such as T-cell acute lymphoblastic leukemia (T-ALL).9 Since platelets do not express VHL, DT2216 importantly spares platelets. Although DT2216 binds both BCL-2 and BCL-XL with high affinity, DT2216 degrades only BCL-XL, not BCL-2. Consequently, DT2216 showed minimal efficacy in cancers that co-depend on both BCL-XL and BCL-2 for survival, such as certain subsets of leukemia and mantle cell lymphoma (MCL), unless combined with venetoclax or conventional chemotherapy.10
We recently reported the first-in-class dual BCL-XL/BCL-2 PROTAC 753B that induces both BCL-XL and BCL-2 ubiquitination and degradation selectively in cells expressing VHL, and demonstrated its pre-clinical activity in tumor cells that co-depend on BCL-2 and BCL-XL.10 In this study, we characterized its broad activity in AML. In addition, recent findings indicate that chemoresistance in AML is associated with chemotherapy-induced cellular senescence.11,12 Upregulation of both BCL-XL and BCL-2 has been reported essential for senescent cell survival.13 Accordingly, inhibition of BCL-XL and BCL-2 activity may facilitate clearance of senescent cells.14 Here, we sought to evaluate the efficacy of 753B as a senolytic agent in leukemia after chemotherapy. Our study showed that 753B effectively eliminates leukemia cells both in vitro and in vivo, and enhances the efficacy of chemotherapy by targeting senescent cells.
Methods
Drugs and reagents
The drugs 753B, DT2216, and QVD (a pan-caspase inhibitor) were kindly provided by Prof. Zhou at the University of Texas Health Science Center and Prof. Zheng at the University of Florida. Venetoclax (ABT-199), Navitoclax (ABT-263), cytarabine (Ara-C) and S63845 were purchased from Selleckchem (Pittsburgh, PA, USA).
Cell lines and primary acute myeloid leukemia samples
Leukemia cell lines were obtained from ATCC (Manassas, VA, USA) or DSMZ (Braunschweig, Germany) and maintained in RPMI 1640 supplemented with 10-20% heat-in-activated fetal bovine serum (Gibco, Thermo-Scientific). All cell lines were frozen and low passage cells were used for all experiments.
Peripheral blood samples were collected from patients with AML; informed consent was obtained in accordance with the requirements of the Institutional Review Board of the University of Texas MD Anderson Cancer Center. The clinical features of the patients are listed in Online Supplementary Table S1.
Western bloting
Cells were lysed and 10-15 mg were prepared,15 resolved by electrophoresis on NuPAGE 4-12% SDS-PAGE gradient gels (Invitrogen), and transferred to polyvinylidene fluoride membranes (Invitrogen). Immunoblotting was performed with primary antibodies: BCL-2 (DAKO), MCL-1 (Santa Cruz Biotechnology), PARP, Caspase-3, Cleaved caspase-3, BCL-XL (Cell Signaling Technology), and tubulin (Sigma-Aldrich). Blots were scanned with an Odyssey Infrared Imaging System (LI-COR Biosciences, Lincoln, NE, USA).
Fluorogenic senescence-associated β-galactosidase activity assay using 5-dodecanoylaminofluorescein diβ-D-galactopyranoside
Senescence-associated β-galactosidase (SA-β-al) activity was assayed as previously described16 using 20 μM 5-dodecanoylaminofluorescein di-β-D-galactopyranoside (C12-FDG) (Thermo Fisher Scientific). Briefly, C12-FDG was prepared in pre-warmed media and added to cells with gentle mixing of the cell suspension. Cells were then incubated with C12-FDG and protected from light for 2-4 hours (h) at 37°C and 5% CO2. SA-β-gal-positive cells were detected and quantified by flow cytometry.
Animal studies
All mouse experiments were approved by the Institutional Animal Care and Use Committee at the University of Texas MD Anderson Cancer Center. Eight-week-old NOD scid gamma (NSG) mice (The Jackson Laboratory, Bar Harbor, Maine, USA) were injected with 3x106 AML patient-derived xenograft (PDX) #4138550 cells (which have FLT3-ITD, DNMT3A, IDH1, KIT, and NPM1 mutations) via the tail vein. After leukemia engraftment was confirmed by flow cytometry 33 days after injection, mice were randomized to receive vehicle or 753B (5 mg/kg intraperitoneally, every 4 days) for three weeks. Mouse livers, spleens, and femurs were collected after the treatment. Leukemia burden was quantified by hCD45+ flow cytometry.
Statistical analysis
Statistical analyses were performed using GraphPad Prism version 8. Results are expressed as mean ± Standard Error of Mean (SEM) of three separate replicate experiments, unless otherwise indicated. Two-sided unpaired Student t-test was used for comparisons between the means of two groups. Kaplan-Meier test was used to analyze the survival rate in the in vivo study and the data were statistically analyzed using log rank (Mantel-Cox) test.
Results
753B is more potent in reducing cell viability in a subset of hematologic cell lines compared with other BCL-XL and/or BCL-2 targeting agents via degradation of BCL-XL and BCL-2
We first evaluated the sensitivity of 24 genetically diverse hematologic cell lines to venetoclax, navitoclax, DT2216 (a BCL-XL selective PROTAC), and 753B (a dual BCL- XL/BCL-2 PROTAC). Our cohort of cell lines included 17 AML, five T-ALL, and two AML secondary to myeloproliferative neoplasms (MPN-AML) cell lines. Treatment with 753B for 24 h caused a dose-dependent but variable reduction of cell viability in all leukemia cell lines tested, as determined by CellTiter-Glo© (CTG) assay, with half-maximal inhibitory concentration (IC50) values ranging from 0.01 μM to 27.35 μM (Online Supplementary Table S2). Both BCL-XL-dependent cell lines (CCRF-CEM, TF-1) and BCL-XL/BCL-2-co-dependent cell lines (Kasumi-1, KG-1) were sensitive to 753B (Figure 1A). Median IC50 values of 753B (0.35 μM) were lower than those of venetoclax (7.69 μM) and navitoclax (0.61 μM) across all cell lines tested (Online Supplementary Figure S1A), indicating that 753B on a molar basis is more potent than other BCL-XL and/or BCL-2 targeting agents. Notably, 12 AML cell lines (KG-1, Kasumi-1, TF-1, HEL 92.1.7, MV4-11, Kasumi-3, UCSD-AML1, HNT37, MOLM-1, CMK, M-07e, F-36P) and three T-ALL cell lines (Jurkat, PF832, CCRF-CEM) showed greater sensitivity to 753B than to navitoclax (median value: 0.10 μM for 753B vs. 0.48 μM for navitoclax; P=0.0005) (Figure 1B). The IC50 of 753B in these cell lines was well correlated with that of navitoclax (correlation coefficient: r2=0.6673) (Online Supplementary Figure S1B). Of note, the top seven highly sensitive cell lines to 753B included three AML-EVI-1 (MECOM) rearranged cell lines (Kasumi-3, UCSD-AML1 and HNT37), which represent AML with extremely poor prognosis17 and are resistant to venetoclax (Online Supplementary Table S2, Online Supplementary Figure S1C).
We next analyzed the baseline (pre-treatment) expression of the anti-apoptotic proteins BCL-XL, BCL-2 and MCL-1 in 17 leukemia cell lines by western blotting. BCL-2 was in general highly expressed in AML cell lines with a notable exception for the MPN-AML cell lines SET-2 and HEL 92.1.7 and all T-ALL lines, in which BCL-XL expression was more prevalent. EVI-1 rearranged lines (e.g., Kasumi-3 and UCSD-AML1) co-expressed BCL-XL and BCL-2 (Online Supplementary Figure S1D, E). To quantify BCL-XL/BCL-2 protein degradation by 753B, we performed western blotting and densitometry analyses in 22 leukemia cell lines treated with 753B. Notably, 753B effectively and potently induced dose-dependent BCL-XL degradation in all tested lines within 24 h, with the concentrations at which 50% of the protein was degraded (DC50) ranging from 0.01 μM to 0.54 μM; in 21 of 22 tested cell lines, the DC50 of BCL-XL to 753B was less than 0.15 μM. BCL-2 was also degraded in 21 of the 22 cell lines, but this generally required higher doses of 753B, with DC50 ranging from 0.02 μM to more than 1 μM, with the exception of the T-ALL Loucy cell line (no degradation at 24 h) (Online Supplementary Table S2, Online Supplementary Figure S1E).
To further understand 753B pro-apoptotic effects, we analyzed apoptosis in 753B-treated Kasumi-1 cells harboring AML-ETO rearrangement (BCL-XL and BCL-2 co-dependent). Consistent with rapid degradation of BCL-XL that we observed as early as 4 h (Figure 1C), 753B treatment rapidly induced apoptosis in Kasumi-1 cells as determined by Annexin-V staining after 4 h of treatment (Figure 1D). The results were confirmed by analyzing the cleavage of caspase-3 and PARP after 8 h (Figure 1C). The activation of caspase-3 by 753B was also observed in the HL60 cell line (Online Supplementary Figure S1F). Moreover, the decrease in cell viability induced by 753B was effectively rescued by the pan-caspase inhibitor Q-VD-OPh (QVD) (Figure 1E, Online Supplementary Figure S1G), suggesting that 753B-induced cell death is largely mediated by caspase-dependent apoptosis.
753B is a more potent antitumor agent than DT2216 in both leukemia cell lines and primary patient samples
To determine whether 753B is more potent than the first-generation BCL-XL-targeted PROTAC DT2216, we exposed 22 cell lines, including AML, T-ALL, and MPN-AML cell lines, to increasing concentrations of DT2216 and 753B for 24 h and then determined their IC50 values. We observed that the sensitivity to DT2216 generally tracked with responsiveness to 753B (Figure 2A). We found that the IC50 of 753B in these cell lines strongly correlated with but were lower than the IC50 of DT2216 (correlation coefficient: r2=0.8732) (Figure 2B). It is notable that in BCL-XL-dependent leukemia cell lines, 753B was more potent than DT2216 (median IC50 : 0.10 vs. 1.16 μM) (Online Supplementary Figure S2A), consistent with higher potency of 753B as BCL-XL degrader due to changing the link-out position on navitoclax and allowing additional lysines on BCL-XL (K20) and BCL-2 (K17) to interact with the E2 enzyme for ubiquitination.10 To better evaluate the antitumor efficacy of 753B, we tested 753B efficacy in the BCL-XL/BCL-2 codependent AML cell line KG-1. Compared with DT2216, 753B more potently induced BCL-XL degradation (DC50, 0.01 vs. 0.06 μM) and additionally induced BCL-2 degradation in KG-1 cells (Figure 2C). Moreover, 753B was the most potent tested inhibitor inducing apoptosis (Figure 2D).
We next tested the sensitivity of 16 primary AML samples to 753B, DT2216, venetoclax, and navitoclax. Our cohort included ten AML samples from patients who were relapsed or refractory (R/R) to venetoclax-based therapies. Consistent with the results in leukemia cell lines, 753B more potently reduced cell viability than DT2216. The median IC50 value of 753B was 0.23 μM and ranged from 0.02 μM to 2.29 μM with IC50 values <0.50 μM in 13 of 16 primary AML patient samples (Online Supplementary Table S1). Of note, the IC50 of 753B in primary samples was lower than that in AML cell lines. 753B was more potent than DT2216 as evident by the extent of BCL-XL degradation, higher apoptosis induction, lower IC50 with antileukemia potency comparable to that of navitoclax in all tested AML samples, including seven venetoclax-resistant samples (defined as IC50 >1 μM) (Figure 2E-G). 753B degraded BCL-XL in all six tested primary samples, and BCL-2 in 4 out of 6 tested samples, with BCL-XL DC50 of 0.01 to approximately 0.18 μM, and BCL-2 DC50 of 0.18 to approximately 3.57 μM (Figure 2E, H, Online Supplementary Figure S2B-E). 753B induced apoptosis associated with cleavage of caspase-3 and PARP (Figure 2H).
753B enhances the efficacy of chemotherapy by eliminating senescent leukemia cells
Recent studies suggested that chemotherapy-induced cellular senescence-mediated chemoresistance and disease relapse in AML.18,19 Tumor cells can undergo senescence in response to stress and therapy, characterized by cell-cycle arrest, increased SA-β-gal activity and acquisition of a Senescence-Associated Secretory Phenotype (SASP) that comprises pro-inflammatory cytokines, chemokines and growth factors. Senescent cells can accumulate, especially under chemotherapy-induced immune suppression, and exhibit a deleterious effect on the tissue microenvironment that drives aging phenotype and potentially promotes tumor growth, relapse, metastasis, and resistance to chemotherapy.11,19,20 Navitoclax and other BCL-XL/BCL-2 inhibitors, like ABT-737 and A1331852 are known to function as potent senolytics, small molecules that selectively clear senescent cells in a variety of tissues and solid tumors,13,14,21 help maximize the efficacy of chemotherapy, and prevent tumor relapse and metastasis.13 Based on these findings, we hypothesized that 753B might exhibit senolytic activity in leukemia.
To confirm the induction of cellular senescence by Ara-C, one of the most used chemotherapy agents in AML, we treated MOLM-14 cells with increasing concentrations of Ara-C for three days. Ara-C-treated cells increased in size and cytoplasmic granularity by flow cytometry (Figure 3A), consistent with previous reports. SA-β-gal is a sensitive marker to identify cells in senescence state,22 and its fluorogenic substrate C12-FDG is widely used to quantify senescent cells.16 Consistent with prior reports, the activity of SA-β-gal and C12-FDG median fluorescence intensity (MFI) was drastically increased in viable cells after 0.05 μM Ara-C treatment for 72 h, indicating that Ara-C induced cellular senescence11 (Figure 3B, C). 753B not only inhibited baseline cell senescence (Online Supplementary Figure S3A-C), but also reversed Ara-C-induced cellular senescence, as indicated by reduced SA-β-gal staining and lower MFI of C12-FDG (Figure 3D, E).
To further validate the effect of 753B on cell senescence, we used immunoblotting to analyze the expression of cell cycle regulators such as p16 (CDKN2A), p21 (CDKN1A), and p53 (TP53), which are known to be associated with senescence.23-25 Expression levels of p16 and p21 were increased after Ara-C treatment but abrogated by 753B (Figure 3B). Consistently, Ara-C treatment also increased the expression levels of pro-inflammatory cytokines and chemokines including interleukin (IL)-8, IL-6, IL-1β, and CCR5, which are SASP markers of senescent cells.26,27 In contrast, 753B co-treatment prevented the Ara-C-induced expression of these cytokines and chemokines (Figure 3G). The inhibitory effect of 753B on chemotherapy-induced senescence was further confirmed in an additional AML cell line, Kasumi-1 (Online Supplementary Figure S3D-K). Taken together, these data indicate that 753B can target chemotherapy-induced senescent AML cells.
Chemotherapy-induced senescent cells express higher levels of BCL-XL, representing a therapeutic target for 753B
To explore the molecular mechanism behind the chemotherapy-induced senescence phenotype, we FACS-sorted viable C12-FDG-high (senescent) and C12-FDG-low MOLM-14 cells after three days exposure to low doses of Ara-C (0.05 μM or 0.10 μM) (Online Supplementary Figure S4A). Compared to that in the C12-FDG-low cells, the expression of BCL-XL was significantly higher in the C12-FDG-high senescent cells (Figure 4A). Consistently high expression of BCL-XL was also observed in C12-FDG-high, Ara-C-treated Kasumi-1 cells (Online Supplementary Figure S4B). This finding suggested that BCL-XL is a key anti-apoptotic protein in senescent cells.
To analyze the clearance of senescent cells upon 753B treatment, MOLM-14 cells were incubated with Ara-C (0.05 μM) for three days to induce senescence, followed by 24 h of treatment with 753B. Compared with control cells without Ara-C treatment (non-senescent cells), senescent cells induced by Ara-C were significantly more sensitive to treatment with 753B, with a 75% death rate at the highest concentration tested (Figure 4B). Besides, the combination of Ara-C and 753B induced higher levels of cleaved caspase-3 and cleaved PARP, indicating increased apoptosis in these cells (Figure 4C). Furthermore, the combination of 753B and Ara-C treatment showed a synergistic growth-inhibitory effect (Figure 4D, E), suggesting that targeting BCL-XL with 753B in combination with chemotherapy may boost chemotherapeutic efficacy in leukemia.
BCL-XL degradation by 753B is associated with MCL-1 upregulation in selected leukemia cells, and the combination of 753B and an MCL-1 inhibitor induced synergistic cell death
MCL-1 is a member of BCL-2 family and a known resistance factor to venetoclax.6,28,29 We have previously reported that in subsets of AML cells, short-term BCL-2 inhibition causes MCL-1 upregulation associated with activation of MAPK signaling.30 The degradation of BCL-XL and BCL-2 by 753B at the intermediate dose (0.11 μM) was similarly associated with MCL-1 upregulation in 13 of 22 cell lines tested. Out of 13 cell lines, we observed increased expression of MCL-1 at all applied concentrations of 753B in five AML lines (Online Supplementary Table S3, Online Supplementary Figure S1D). MCL-1 dose-dependent upregulation was observed as early as 4 h after 753B exposure (Figure 5A). Co-immunoprecipitation (Co-IP) and BH3 profiling were performed to explore the functional consequences of an increased level of MCL-1. 753B induced an increase in Bim binding to MCL-1 compared with vehicle treated cells as demonstrated by Co-IP (Figure 5B, C). Dynamic BH3 profiling demonstrated higher release of cytochrome C by the MCL-1-specific peptide MS-1 and the MCL-1 inhibitor S63845, indicating increased MCL-1 dependence following 753B exposure (Figure 5D). To further characterize the relationship between expression of BCL-2 family proteins and sensitivity to 753B, we quantified the expression levels of BCL-XL, BCL-2 and MCL-1 by western blotting by densitometry of the bands in ten AML cell lines (Online Supplementary Figure S1D). Spearman correlation analysis was performed to evaluate the relationship between IC50 values and the protein change level of BCL-XL, BCL-2 and MCL-1 by 753B. We found that the sensitivity to 753B inversely correlated with MCL-1 level when treated with 753B at the concentration of 1 μM at 24 h (r=-0.8182, P=0.0038) but did not correlate with the degradation level of BCL-XL or BCL-2 at the same concentrations (Figure 5E). These data further support MCL-1 dependence following 753B treatment. 753B induced cell death in 50% of OCI-AML-2 cells at a concentration of 0.64 μM and in nearly 100% of the cells, when combined with a low dose of S63845 (0.005 μM) at 24 h by CTG assay; induction of cell death was synergistic at multiple concentrations tested (Figure 5F). These data suggest that the dual targeting of MCL-1 and BCL-XL is highly effective in inducing cell death in AML.
Anti-leukemia efficacy of 753B in vivo in acute myeloid leukemia patient-derived xenograf model
To investigate the antileukemia activity of 753B in vivo, we developed a PDX model by injecting NSG mice with AML PDX #4138550 (generated from an AML patient harboring FLT3-ITD, DNMT3A, IDH1, KIT, and NPM1 mutations). After confirming engraftment by peripheral blood flow cytometry analysis, mice were randomized into two groups to receive either vehicle or 753B (5 mg/kg intraperitoneally every 4 days) for three weeks. Mice tolerated 753B therapy well with no significant changes in body weight and no significant normal hematopoietic cells, platelet or white blood cell (WBC) toxicity as measured by blood counts (Figure 6A, Online Supplementary Figure S5A-C). 753B treatment reduced the circulating leukemia cell burden measured by flow cytometry, reduced liver and spleen weight, and extended overall survival (Figure 6B, C, Online Supplementary Figure S5D). The histological evaluation of Hematoxylin & Eosin (HE) stained sections of the bone marrow (BM), liver, and spleen showed reduction in the tumor burden of the BM, spleen and liver after 753B treatment, with no microscopic evidence of liver or spleen damage (Figure 6D). The reduction of leukemia burden induced by 753B was associated with a notable reduction in BCL-XL expression in leukemia cells harvested after 753B treatment (Figure 6E, F).
Discussion
Both BCL-XL and BCL-2 are validated therapeutic targets with overexpression noted in a wide range of hematologic malignancies.31-33 In addition to MCL-1, BCL-XL is also a key mediator of chemotherapy and venetoclax resistance.4,5,34 Thus, developing a tolerable and effective BCL-XL-targeted or BCL-2/BCL-XL co-targeted therapy is of high priority. Given the caution required in dealing with navitoclax-induced thrombocytopenia, drug discovery efforts have aimed at circumventing this limitation through optimizing navitoclax-based targeting. In this study, we validated the effect of a novel dual BCL-XL/BCL-2 PROTAC 753B, that was developed using navitoclax as a ligand in genetically diverse leukemia cells. We observed that 753B exhibited a potent anti-leukemia efficacy similar to that of navitoclax, making it a promising therapeutic candidate. We noted that 753B does not degrade BCL-2 as readily as BCL-XL . We speculate that one of the possible mechanisms for the discrepancy in degradation between BCL-XL and BCL-2 is at least partially to be attributed to the differential distribution and orientation of lysines on the protein surfaces, as we reported recently.10 In addition, the protonated state, flexibility, as well as the other residues around the lysine involved in the E2/POI interface would also be key factors contributing to the difference in lysine ubiquitination effectiveness between BCL-XL and BCL-2. These differences may render BCL-XL more susceptible to 753B-induced protein ubiquitination and degradation than BCL-2. The detailed mechanisms and optimization of the dual degradation will require future studies.
753B induced rapid dose-dependent BCL-XL and BCL-2 degradation in leukemia, which translated into inhibition of cell growth and apoptosis. This effect was observed in 11 BCL-2/BCL-XL co-dependent cell lines (2/2 MPN-AML and 9/17 AML cell lines including the high-risk EVI-1 rearranged cells) and in the majority of the primary AML samples harboring high-risk mutations (such as FLT-ITD, TP53 and NRAS), exhibiting a broader activity than venetoclax. Notably, 10 out of 16 samples were collected from patients, whose disease progressed after venetoclax-containing regimens, and all ten samples responded in vitro to navitoclax and 753B. These data indicate a potential utility of BCL-XL-targeting agents in overcoming resistance to venetoclax. 753B showed preliminary in vivo efficacy in AML-derived PDX harboring FLT3-ITD, DNMT3A, and IDH1 mutations. We noted the rebound in hCD45 cellularity after the last week of 753B treatment and assume that the rebound of leukemia could result from MCL-1 upregulation or due to a sub-optimal dose of 753B used in the in vivo study. Efficacy of 753B or optimized next generation degrader should be tested in additional AML PDX models with diverse genetic backgrounds in future studies. With further optimization of formulation and dosing, 753B has the potential to become the first-in-class platelet-sparing BCL-XL/BCL-2 targeting antitumor agent.
Chemotherapy works in part by inducing cellular senescence; immune cells then clear senescent cancer cells, leading to tumor stasis or regression. However, these senescent cells can escape from immune surveillance and re-enter the cell cycle after escaping from a resting state with accelerated potential for growth.18,35,36 These cells can acquire novel stem-cell and self-renewing features, promote disease relapse, and enhance aggressiveness, as evidenced in several mouse tumor models, including AML.27 Recent studies identified navitoclax and other BCL-XL/BCL-2 inhibitors (ABT-737 and A1331852) as potent senolytics.21,37,38 Because BCL-XL/BCL-2 are key anti-apoptotic proteins in many types of senescent cells, we hypothesized that 753B may eliminate senescent cells before escaping from senescence. In our study, we found that 753B largely reversed Ara-C-induced senescence markers and enhanced the efficacy of Ara-C by targeting senescent AML cells, suggesting an added benefit from targeting BCL-XL/BCL-2 with 753B in combination with chemotherapy for AML treatment. This finding provides a rationale for sequential treatment of chemotherapy followed by 753B, which could reduce the toxicity of concomitant administration, yet avoid relapse by eliminating chemotherapy-induced senescent tumor cells.
Our mechanistic studies suggest that the sensitivity of AML cell lines to 753B inversely correlates with high MCL-1 expression. MCL-1 expression commonly accounts for resistance to other BH3 mimetics, including venetoclax.39,40 Our data indicate that 753B has a strong synergistic effect on inducing cell death in MOLM-14 cells when combined with a small molecule inhibitor targeting MCL-1 (Figure 5F). However, the synergy of 753B and MCL-1 inhibitors should be tested in additional cells lines or xenograft models in future studies. Notably, co-administration of MCL-1 inhibitor (VU661013) and BCL-2 inhibitor has been reported to have a reasonable safety profile in AML xenografts models.41 Besides, co-targeting BCL-XL and MCL-1 with DT2216 and AZD8055 showed a synergistic effect on inhibiting small-cell lung cancer growth without causing on-target toxicities in mice.42 On the contrary, the simultaneous use of MCL-1 and BCL-XL inhibitors caused acute lethality in mice in lung squamous cell carcinomas and pancreatic cancer in vivo models.43,44 Since 753B is a BCL-2/BCL-XL co-targeted compound, the safety of directly combining an MCL-1 inhibitor with 753B will require careful in vivo safety studies in future experiments.
Recognition of the important mechanistic role of BCL-XL in leukemias sparked renewed interest in therapeutically inhibiting BCL-XL. In a recently reported phase I study (clinicaltrials. gov identifier: NCT03181126), co-targeting BCL-XL and BCL-2 by combination of reduced doses of navitoclax and venetoclax showed promising efficacy in patients with relapsed or refractory ALL who progressed after multiple lines of therapy.45 The use of navitoclax as an “add-on” approach to the JAK2 inhibitor ruxolitinib in patients with myelofibrosis produced objective responses in a phase II clinical study (clinicaltrials. gov identifier: NCT03222609), although careful dose titration of navitoclax was required due to an overall decrease in platelet counts.46 AZD0466 is a nanoparticle-formulated novel dual BCL-XL/BCL-2 inhibitor that is currently in a phase I clinical trial for hematologic malignancies (clinicaltrials. gov identifier: NCT04214093) and it is pharmacologically optimized to minimize thrombocytopenia.47 In contrast, 753B is a first-in-class dual BCL-XL/BCL-2 degrader that avoids platelet toxicity due to selective expression of VHL E3 ligase. Because 753B is derived from navitoclax, 753B is expected to be a safe strategy to target both BCL-2 and BCL-XL in AML, without resultant thrombocytopenia.
Taken together, our study demonstrates that the singleagent 753B has an anti-tumor activity in a subset of hematologic cell lines, primary patient-derived AML blasts, and a xenograft model via BCL-2 family protein degradation. We demonstrated that the senolytic properties of 753B may enhance the efficacy of chemotherapy by targeting BCL-XL-expressing senescent cells. Importantly, the first-in-human dose escalation study of BCL-XL PROTAC DT2216 has opened for patient accrual (clinicaltrials. gov identifier: NCT04886622). We anticipate that the clinical use of PROTAC based on our informative pre-clinical findings will lead to safe and effective co-targeting of BCL-2 and BCL-XL in AML.
Footnotes
- Received August 22, 2022
- Accepted April 7, 2023
Correspondence
Disclosures
GZ and DZ are co-founders of and have equity in Dialectic Therapeutics, which develops BCL-XL PROTAC to treat cancer. The other authors have no conflict of interests to disclose.
Contributions
YJ conceived, designed and performed most of the biological and biochemical experiments, analyzed and interpreted data, and wrote the manuscript. LH, CR, ZW, CW, LY, SC and HM performed and analyzed some of the biological experiments. WZ, MA, ND, NJ, NP, KB and SM supervised some of the biological studies and revised the manuscript. PZ synthesized 753B, and prepared the formulation of vehicle and 753B for animal study. GZ conceived, designed and supervised the synthesis of BCL-XL/2 PROTAC, and revised the manuscript. DZ conceived, designed and supervised the synthesis of BCL-XL/2 PROTAC, revised the manuscript, and guided senescence studies. QZ and MK conceived, designed, and supervised the study, analyzed and interpreted data, and wrote the manuscript. All authors discussed the results and commented on the manuscript.
Data-sharing statement
Original data and protocols are available to other investigators upon request by contacting the corresponding author or last author.
Funding
This study was supported by US National Institutes of Health (NIH) grants R01 CA241191 (to GZ, MK and DZ), the Cancer Foundation Finland, and the Academy of Finland (to SM).
References
- Adams JM, Cory S. The Bcl-2 apoptotic switch in cancer development and therapy. Oncogene. 2007; 26(9):1324-1337. https://doi.org/10.1038/sj.onc.1210220PubMedPubMed CentralGoogle Scholar
- Konopleva M, Contractor R, Tsao T. Mechanisms of apoptosis sensitivity and resistance to the BH3 mimetic ABT-737 in acute myeloid leukemia. Cancer Cell. 2006; 10(5):375-388. https://doi.org/10.1016/j.ccr.2006.10.006PubMedGoogle Scholar
- Pan R, Hogdal LJ, Benito JM. Selective BCL-2 inhibition by ABT-199 causes on-target cell death in acute myeloid leukemia. Cancer Discov. 2014; 4(3):362-375. https://doi.org/10.1158/2159-8290.CD-13-0609PubMedPubMed CentralGoogle Scholar
- Konopleva M, Pollyea DA, Potluri J. Efficacy and biological correlates of response in a phase II study of venetoclax monotherapy in patients with acute myelogenous leukemia. Cancer Discov. 2016; 6(10):1106-1117. https://doi.org/10.1158/2159-8290.CD-16-0313PubMedPubMed CentralGoogle Scholar
- Lin KH, Winter PS, Xie A. Targeting MCL-1/BCL-XL forestalls the acquisition of resistance to ABT-199 in acute myeloid leukemia. Sci Rep. 2016; 6:27696. https://doi.org/10.1038/srep27696PubMedPubMed CentralGoogle Scholar
- Zhang Q, Riley-Gillis B, Han L. Activation of RAS/MAPK pathway confers MCL-1 mediated acquired resistance to BCL-2 inhibitor venetoclax in acute myeloid leukemia. Signal Transduct Target Ther. 2022; 7(1):51. https://doi.org/10.1038/s41392-022-00958-4PubMedPubMed CentralGoogle Scholar
- Oppermann S, Ylanko J, Shi Y. High-content screening identifies kinase inhibitors that overcome venetoclax resistance in activated CLL cells. Blood. 2016; 128(7):934-947. https://doi.org/10.1182/blood-2015-12-687814PubMedPubMed CentralGoogle Scholar
- Schoenwaelder SM, Jarman KE, Gardiner EE. Bcl-xL-inhibitory BH3 mimetics can induce a transient thrombocytopathy that undermines the hemostatic function of platelets. Blood. 2011; 118(6):1663-1674. https://doi.org/10.1182/blood-2011-04-347849PubMedGoogle Scholar
- Khan S, Zhang X, Lv D. A selective BCL-XL PROTAC degrader achieves safe and potent antitumor activity. Nat Med. 2019; 25(12):1938-1947. https://doi.org/10.1038/s41591-019-0668-zPubMedPubMed CentralGoogle Scholar
- Lv D, Pal P, Liu X. Development of a BCL-xL and BCL-2 dual degrader with improved anti-leukemic activity. Nat Commun. 2021; 12(1):6896. https://doi.org/10.1038/s41467-021-27210-xPubMedPubMed CentralGoogle Scholar
- Duy C, Li M, Teater M. Chemotherapy induces senescence-like resilient cells capable of initiating AML recurrence. Cancer Discov. 2021; 11(6):1542-1561. https://doi.org/10.1158/2159-8290.CD-20-1375PubMedPubMed CentralGoogle Scholar
- Milanovic M, Fan DNY, Belenki D. Senescence-associated reprogramming promotes cancer stemness. Nature. 2018; 553(7686):96-100. https://doi.org/10.1038/nature25167PubMedGoogle Scholar
- Yosef R, Pilpel N, Tokarsky-Amiel R. Directed elimination of senescent cells by inhibition of BCL-W and BCL-XL. Nat Commun. 2016; 7:11190. https://doi.org/10.1038/ncomms11190PubMedPubMed CentralGoogle Scholar
- He Y, Zhang X, Chang J. Using proteolysis-targeting chimera technology to reduce navitoclax platelet toxicity and improve its senolytic activity. Nat Commun. 2020; 11(1):1996. https://doi.org/10.1038/s41467-020-15838-0PubMedPubMed CentralGoogle Scholar
- Han L, Zhang Q, Dail M. Concomitant targeting of BCL2 with venetoclax and MAPK signaling with cobimetinib in acute myeloid leukemia models. Haematologica. 2020; 105(3):697-707. https://doi.org/10.3324/haematol.2018.205534PubMedPubMed CentralGoogle Scholar
- Debacq-Chainiaux F, Erusalimsky JD, Campisi J, Toussaint O. Protocols to detect senescence-associated beta-galactosidase (SA-betagal) activity, a biomarker of senescent cells in culture and in vivo. Nat Protoc. 2009; 4(12):1798-1806. https://doi.org/10.1038/nprot.2009.191PubMedGoogle Scholar
- Matsuo H, Kajihara M, Tomizawa D. EVI1 overexpression is a poor prognostic factor in pediatric patients with mixed lineage leukemia-AF9 rearranged acute myeloid leukemia. Haematologica. 2014; 99(11):e225-227. https://doi.org/10.3324/haematol.2014.107128PubMedPubMed CentralGoogle Scholar
- De Blander H, Morel AP, Senaratne AP, Ouzounova M, Puisieux A. Cellular plasticity: a route to senescence exit and tumorigenesis. Cancers (Basel). 2021; 13(18):4561. https://doi.org/10.3390/cancers13184561PubMedPubMed CentralGoogle Scholar
- Demaria M, O'Leary MN, Chang J. Cellular senescence promotes adverse effects of chemotherapy and cancer relapse. Cancer Discov. 2017; 7(2):165-176. https://doi.org/10.1158/2159-8290.CD-16-0241PubMedPubMed CentralGoogle Scholar
- Munoz DP, Yannone SM, Daemen A. Targetable mechanisms driving immunoevasion of persistent senescent cells link chemotherapy-resistant cancer to aging. JCI Insight. 2019; 5(14):e124716. https://doi.org/10.1172/jci.insight.124716PubMedPubMed CentralGoogle Scholar
- Zhu Y, Tchkonia T, Fuhrmann-Stroissnigg H. Identification of a novel senolytic agent, navitoclax, targeting the Bcl-2 family of anti-apoptotic factors. Aging Cell. 2016; 15(3):428-435. https://doi.org/10.1111/acel.12445PubMedPubMed CentralGoogle Scholar
- Dimri GP, Lee X, Basile G. A biomarker that identifies senescent human cells in culture and in aging skin in vivo. Proc Natl Acad Sci U S A. 1995; 92(20):9363-9367. https://doi.org/10.1073/pnas.92.20.9363PubMedPubMed CentralGoogle Scholar
- Schmitt CA, Fridman JS, Yang M. A senescence program controlled by p53 and p16INK4a contributes to the outcome of cancer therapy. Cell. 2002; 109(3):335-346. https://doi.org/10.1016/S0092-8674(02)00734-1PubMedGoogle Scholar
- Suzuki K, Mori I, Nakayama Y, Miyakoda M, Kodama S, Watanabe M. Radiation-induced senescence-like growth arrest requires TP53 function but not telomere shortening. Radiat Res. 2001; 155(1 Pt 2):248-253. https://doi.org/10.1667/0033-7587(2001)155[0248:RISLGA]2.0.CO;2Google Scholar
- Serrano M, Lin AW, McCurrach ME, Beach D, Lowe SW. Oncogenic ras provokes premature cell senescence associated with accumulation of p53 and p16INK4a. Cell. 1997; 88(5):593-602. https://doi.org/10.1016/S0092-8674(00)81902-9PubMedGoogle Scholar
- Elashiry M, Elsayed R, Cutler CW. Exogenous and endogenous dendritic cell-derived exosomes: lessons learned for immunotherapy and disease pathogenesis. Cells. 2021; 11(1):115. https://doi.org/10.3390/cells11010115PubMedPubMed CentralGoogle Scholar
- Zhang L, Pitcher LE, Prahalad V, Niedernhofer LJ, Robbins PD. Targeting cellular senescence with senotherapeutics: senolytics and senomorphics. FEBS J. 2023; 290(5):1362-1383. https://doi.org/10.1111/febs.16350PubMedGoogle Scholar
- Delbridge AR, Grabow S, Strasser A, Vaux DL. Thirty years of BCL-2: translating cell death discoveries into novel cancer therapies. Nat Rev Cancer. 2016; 16(2):99-109. https://doi.org/10.1038/nrc.2015.17PubMedGoogle Scholar
- Delbridge AR, Strasser A. The BCL-2 protein family, BH3-mimetics and cancer therapy. Cell Death Differ. 2015; 22(7):1071-1080. https://doi.org/10.1038/cdd.2015.50PubMedPubMed CentralGoogle Scholar
- Konopleva M, Milella M, Ruvolo P. MEK inhibition enhances ABT-737-induced leukemia cell apoptosis via prevention of ERK-activated MCL-1 induction and modulation of MCL-1/BIM complex. Leukemia. 2012; 26(4):778-787. https://doi.org/10.1038/leu.2011.287PubMedPubMed CentralGoogle Scholar
- Espana L, Fernandez Y, Rubio N, Torregrosa A, Blanco J, Sierra A. Overexpression of Bcl-xL in human breast cancer cells enhances organ-selective lymph node metastasis. Breast Cancer Res Treat. 2004; 87(1):33-44. https://doi.org/10.1023/B:BREA.0000041579.51902.89PubMedGoogle Scholar
- Amundson SA, Myers TG, Scudiero D, Kitada S, Reed JC, Fornace AJ Jr. An informatics approach identifying markers of chemosensitivity in human cancer cell lines. Cancer Res. 2000; 60(21):6101-6110. Google Scholar
- Olopade OI, Adeyanju MO, Safa AR. Overexpression of BCL-x protein in primary breast cancer is associated with high tumor grade and nodal metastases. Cancer J Sci Am. 1997; 3(4):230-237. Google Scholar
- Bose P, Gandhi V, Konopleva M. Pathways and mechanisms of venetoclax resistance. Leuk Lymphoma. 2017; 58(9):1-17. https://doi.org/10.1080/10428194.2017.1283032PubMedPubMed CentralGoogle Scholar
- Sagiv A, Krizhanovsky V. Immunosurveillance of senescent cells: the bright side of the senescence program. Biogerontology. 2013; 14(6):617-628. https://doi.org/10.1007/s10522-013-9473-0PubMedGoogle Scholar
- Coppe JP, Desprez PY, Krtolica A, Campisi J. The senescence-associated secretory phenotype: the dark side of tumor suppression. Annu Rev Pathol. 2010; 5:99-118. https://doi.org/10.1146/annurev-pathol-121808-102144PubMedPubMed CentralGoogle Scholar
- Gasek NS, Kuchel GA, Kirkland JL, Xu M. Strategies for targeting senescent cells in human disease. Nat Aging. 2021; 1(10):870-879. https://doi.org/10.1038/s43587-021-00121-8PubMedPubMed CentralGoogle Scholar
- Zhu Y, Doornebal EJ, Pirtskhalava T. New agents that target senescent cells: the flavone, fisetin, and the BCL-XL inhibitors, A1331852 and A1155463. Aging (Albany NY). 2017; 9(3):955-963. https://doi.org/10.18632/aging.101202PubMedPubMed CentralGoogle Scholar
- Carter BZ, Mak PY, Tao W. Targeting MCL-1 dysregulates cell metabolism and leukemia-stroma interactions and resensitizes acute myeloid leukemia to BCL-2 inhibition. Haematologica. 2022; 107(1):58-76. https://doi.org/10.3324/haematol.2020.260331PubMedPubMed CentralGoogle Scholar
- Cory S, Adams JM. Killing cancer cells by flipping the Bcl-2/Bax switch. Cancer Cell. 2005; 8(1):5-6. https://doi.org/10.1016/j.ccr.2005.06.012PubMedGoogle Scholar
- Ramsey HE, Fischer MA, Lee T. A novel MCL1 inhibitor combined with venetoclax rescues venetoclax-resistant acute myelogenous leukemia. Cancer Discov. 2018; 8(12):1566-1581. https://doi.org/10.1158/2159-8290.CD-18-0140PubMedPubMed CentralGoogle Scholar
- Khan S, Kellish P, Connis N. Co-targeting BCL-X(L) and MCL-1 with DT2216 and AZD8055 synergistically inhibit smallcell lung cancer growth without causing on-target toxicities in mice. Cell Death Discov. 2023; 9(1):1. https://doi.org/10.1038/s41420-022-01296-8PubMedPubMed CentralGoogle Scholar
- Weeden CE, Ah-Cann C, Holik AZ. Dual inhibition of BCL-XL and MCL-1 is required to induce tumour regression in lung squamous cell carcinomas sensitive to FGFR inhibition. Oncogene. 2018; 37(32):4475-4488. https://doi.org/10.1038/s41388-018-0268-2PubMedGoogle Scholar
- Thummuri D, Khan S, Underwood PW. Overcoming gemcitabine resistance in pancreatic cancer using the BCL-XL-Specific Degrader DT2216. Mol Cancer Ther. 2022; 21(1):184-192. https://doi.org/10.1158/1535-7163.MCT-21-0474PubMedPubMed CentralGoogle Scholar
- Pullarkat VA, Lacayo NJ, Jabbour E. Venetoclax and navitoclax in combination with chemotherapy in patients with relapsed or refractory acute lymphoblastic leukemia and lymphoblastic lymphoma. Cancer Discov. 2021; 11(6):1440-1453. https://doi.org/10.1158/2159-8290.CD-20-1465PubMedPubMed CentralGoogle Scholar
- Harrison CN, Garcia JS, Somervaille TCP. Addition of navitoclax to ongoing ruxolitinib therapy for patients with myelofibrosis with progression or suboptimal response: phase II safety and efficacy. J Clin Oncol. 2022; 40(15):1671-1680. https://doi.org/10.1200/JCO.21.02188PubMedPubMed CentralGoogle Scholar
- Balachander SB, Criscione SW, Byth KF. AZD4320, A dual inhibitor of Bcl-2 and Bcl-xL, induces tumor regression in hematologic cancer models without dose-limiting thrombocytopenia. Clin Cancer Res. 2020; 26(24):6535-6549. https://doi.org/10.1158/1078-0432.CCR-20-0863PubMedGoogle Scholar
Data Supplements
Figures & Tables
Article Information
This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.