Purpose:

Development of B-cell lymphoma 2 (BCL-2)–specific inhibitors poses unique challenges in drug design because of BCL-2 homology domain 3 (BH3) shared homology between BCL-2 family members and the shallow surface of their protein–protein interactions. We report herein discovery and extensive preclinical investigation of lisaftoclax (APG-2575).

Experimental Design:

Computational modeling was used to design “lead” compounds. Biochemical binding, mitochondrial BH3 profiling, and cell-based viability or apoptosis assays were used to determine the selectivity and potency of BCL-2 inhibitor lisaftoclax. The antitumor effects of lisaftoclax were also evaluated in several xenograft models.

Results:

Lisaftoclax selectively binds BCL-2 (Ki < 0.1 nmol/L), disrupts BCL-2:BIM complexes, and compromises mitochondrial outer membrane potential, culminating in BAX/BAK-dependent, caspase-mediated apoptosis. Lisaftoclax exerted strong antitumor activity in hematologic cancer cell lines and tumor cells from patients with chronic lymphocytic leukemia, multiple myeloma, or Waldenström macroglobulinemia. After lisaftoclax treatment, prodeath proteins BCL-2‒like protein 11 (BIM) and Noxa increased, and BIM translocated from cytosol to mitochondria. Consistent with these apoptotic activities, lisaftoclax entered malignant cells rapidly, reached plateau in 2 hours, and significantly downregulated mitochondrial respiratory function and ATP production. Furthermore, lisaftoclax inhibited tumor growth in xenograft models, correlating with caspase activation, poly (ADP-ribose) polymerase 1 cleavage, and pharmacokinetics of the compound. Lisaftoclax combined with rituximab or bendamustine/rituximab enhanced antitumor activity in vivo.

Conclusions:

These findings demonstrate that lisaftoclax is a novel, orally bioavailable BH3 mimetic BCL-2–selective inhibitor with considerable potential for the treatment of certain hematologic malignancies.

Translational Relevance

B-cell lymphoma 2 (BCL-2) is a key antideath protein that is often highly expressed in hematologic malignancies and plays important roles in dysfunctional apoptosis signaling and tumorigenesis. Thus, BCL-2 is one of the most well-known, but challenging, targets for pharmacologic intervention. Our study establishes lisaftoclax (APG-2575) as a new BCL-2 homology domain 3 mimetic BCL–2–selective inhibitor that enters tumor cells rapidly and disrupts BCL-2:BIM complexes. Lisaftoclax induces BAX/BAK-dependent apoptosis and antitumor activity, as a single agent or combined with standard-of-care therapeutics, in multiple preclinical models, including chronic lymphocytic leukemia, multiple myeloma, Waldenström macroglobulinemia, and malignancies relapsed from ibrutinib therapy. These findings provide rationales for ongoing clinical trials investigating lisaftoclax as monotherapy or in combination with a CD20 antibody in hematologic malignancies (NCT03537482; NCT04260217).

B-cell lymphoma 2 (BCL-2) proteins control programmed cell death (apoptosis), and dysregulation of these proteins can promote tumor cell survival. The BCL-2 family comprises antiapoptotic proteins [e.g., BCL-2, myeloid cell leukemia 1 (MCL-1), BCL‒extra-large (BCL-xL)] that bind to and sequester proapoptotic family members, such as BCL-2‒like protein 11 (BIM), BCL-2 homology domain 3 (BH3)-interacting domain death agonist (BID), BCL-2–associated X and K proteins (BAX/BAK), Noxa, and p53 upregulated modulator of apoptosis (1–3). Small-molecule inhibitors of BCL-2 proteins enhance cancer cell apoptosis across many hematologic malignancies, such as chronic lymphocytic leukemia (CLL), diffuse large B-cell lymphoma (DLBCL), acute myeloid leukemia (AML), myelodysplastic syndrome, Waldenström macroglobulinemia (WMG), and multiple myeloma (4–9).

Structural insights into interactions between proapoptotic and antiapoptotic proteins culminated in the discovery and clinical development of the first BH3 mimetic: navitoclax (ABT-263; ref. 10). Despite clinical efficacy among patients with advanced cancer, many patients who received navitoclax experienced high-grade thrombocytopenia driven by on-target BCL-xL–mediated platelet inhibition (11, 12).

Subsequently, BH3-mimetic venetoclax (ABT-199; refs. 13, 14) was developed to bind more selectively to BCL-2 and became the first FDA-approved BCL-2–selective small-molecule inhibitor, initially in patients with CLL (and subsequently AML). However, many individuals who receive venetoclax experience hematologic toxicities and tumor lysis syndrome (15–17), which warrants a 5-week dose ramp-up (according to US product labeling) that may pose both logistical challenges for some patients as well as a burden on the healthcare system.

Therefore, we designed lisaftoclax (APG-2575) as a novel, potent, oral BCL-2–specific inhibitor. We herein describe preclinical characterization of lisaftoclax across a range of hematologic malignancies, focusing on the promising antitumor activity and potential underlying mechanisms of action for this new BCL-2 inhibitor.

Study approval

In NOD/SCID NOD.CB17-Prkdcscid/J or NOD/SCID/γ (NSG) NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ mice, a patient-derived xenograft (PDX) tumor model was developed using a specimen of fresh primary tumor tissue collected posthumously (under an approved protocol by the Mayo Clinic Institutional Review Board) from a patient with aggressive WMG. The PDX mouse model was established as previously reported (18–20) and per Mayo Clinic Institutional Animal Care and Use Committee approval and institutional guidelines.

Computational modeling/code availability

BCL-2 in the crystal structure between BCL-2 and a navitoclax analog (PDB ID 4LVT; ref. 13) was extracted to construct the binding modes of lisaftoclax with BCL-2. Ionization states of the amino acids in BCL-2 were calculated and hydrogen atoms added using the protonate 3D module in the Molecular Operating Environment (MOE) program (21) by setting the pH to physiologic 7.0. The structure of lisaftoclax was also prepared using MOE and docking simulation performed using the Genetic Optimisation for Ligand Docking program (version 5.6.2; refs. 22, 23). We performed the template docking protocol by imposing distance constraints (1.5–3.5 Å and a force constant of 5.0 kcal/mol/Å2) between 7 atoms on navitoclax and atoms on BCL-2 (D137, W141, R143, G191, V89, L94, and S102). The center of the binding site was set at G142 in BCL-2, and the radius of the binding site was 13 Å. Default parameters and the GoldScore fitness function were used in the docking calculations. The top-ranked pose of lisaftoclax was selected as the predicted binding model.

Fluorescence polarization competitive binding assay

Recombinant human BCL-2, BCL-xL, and MCL-1 protein were prepared as previously described (24). Specifically, an isoform 2 construct of human BCL-2 with an N-terminal 6xHis tag (a polyhistidine tag) was used. Human BCL-xL constructs with internal deletions of amino acids 45–85 and a C-terminal truncation of amino acid residues 212–233 were cloned into a pHis-TEV vector (a modified pET vector). An MCL-1 fragment encoding amino acid residues 171–327 was also cloned into a pHis-TEV vector.

Fluorescein-labeled BIM (amino acids 81–106), BAK (amino acids 72–87), and BID (amino acids 79–99) peptides, named as Flu-BIM, Flu-BAK, and Flu-BID, respectively, were used as fluorescent probes in fluorescence polarization (FP) assays for BCL-2, BCL-xL, and MCL-1, respectively. For each reaction, 1 nmol/L of Flu-BIM and 2 nmol/L of Flu-BAK or Flu-BID (plus increasing concentrations of BCL-2, BCL-xL, or MCL-1) were added to a final volume of 125 μL in assay buffer (100 mmol/L potassium phosphate, pH 7.5; 100 μg/mL bovine γ-globulin; 0.02% sodium azide; Invitrogen, with 0.01% Triton X-100; 4% DMSO). Reactants were mixed and incubated at room temperature for 1 hour, with gentle agitation to assure equilibrium.

All FP measurements were performed using Microfluor 1 96-well, black, round-bottom plates (Thermo Fisher, M33089) on an Infinite M1000 plate reader (Tecan US, Inc; Morrisville, NC). Polarization values in millipolarization units were measured at an excitation wavelength of 485 nm and an emission wavelength of 530 nm. Equilibrium dissociation constants (Kd) were then calculated using Prism 5.0 software (GraphPad Software, San Diego, CA) by fitting sigmoidal dose-dependent FP increases as a function of protein concentration. By monitoring total FP values of mixtures composed of fluorescent probes at fixed concentrations and proteins with increasing concentrations to full saturation, we determined mean (SD) Kd values of Flu-BIM to BCL-2 (0.55 ± 0.15 nmol/L), Flu-BAK to BCL-xL (4.4 ± 0.8 nmol/L), and Flu-BID to MCL-1 (6.9 ± 0.9 nmol/L).

Inhibition constants (Ki) for binding of lisaftoclax and venetoclax to BCL-2, BCL-xL, and MCL-1 were determined from competitive binding experiments in which serial dilutions of the BCL-2 inhibitors were added to 96-well plates containing fixed concentrations of fluorescent probes and proteins in each well. Mixtures of 5 μL of the tested inhibitors in DMSO and 120 μL of preincubated protein/probe complexes in the assay buffer were added to assay plates and incubated at room temperature for 2 hours with gentle agitation. Final concentrations of the protein and probe (respectively) were 1.5 nmol/L and 1 nmol/L for the BCL-2 assay, 10 nmol/L and 2 nmol/L for the BCL-xL assay, and 20 nmol/L and 2 nmol/L for the MCL-1 assay. Negative controls containing only protein/probe complexes (equivalent to 0% inhibition) and positive controls containing only free probe (equivalent to 100% inhibition) were included in each assay plate.

FP values were measured as described above. Half-maximal IC50 values were determined by nonlinear regression fitting of the competition curves. Ki values of competitive inhibitors were calculated using a previously published equation (25) based on measured IC50 values, Kd values of probes to proteins, and concentrations of proteins and probes in the competitive assays. Ki values were also calculated using a previously described equation (26).

Cellular uptake assay, liquid chromatography/tandem mass spectrometry conditions, and sample quantitation

To assess the kinetics of BCL-2 complex disruption by lisaftoclax, we performed cellular uptake assays on RPMI8226 and KMS-11 cells. Cancer cell lines and culture conditions are summarized in Supplementary Table S1. RPMI8226 and KMS-11 cells were cultured to confluence on a 24-well plate, then incubated with fresh culture medium containing 1 μmol/L lisaftoclax or venetoclax at 37°C.

At predefined time points, culture media were removed and cells washed 3 times with ice-cold PBS. Cell lysis buffer was then added, followed by centrifugation to collect the supernatant as a cell lysate. A total of 50 μL of cell lysate was added with 1,000 μL of acetonitrile containing tolbutamide 5 ng/mL [internal standard (IS)]. The mixture was vortexed for 2 minutes and centrifuged at 13,000 rpm for 10 minutes, and the supernatant transferred to vial inserts and injected into the liquid chromatography/tandem mass spectrometry (LC/MS-MS) system for analysis. Total protein concentration in the cell lysate was quantitated using a Pierce BCA Protein Assay Kit (Thermo Fisher, 23227) according to the manufacturer instructions. Cellular concentrations of lisaftoclax and venetoclax were normalized to protein concentration and expressed as pmol drug/mg protein.

The LC/MS-MS analysis was conducted using an ExionLC HPLC system (AB Sciex LLC, Framingham, MA) coupled with an atmospheric pressure ionization 5500 LC/MS-MS system (AB Sciex). HPLC separation was achieved using an Agilent C-18 column (5 μmol/L, 2.1 mm × 50 mm) (Agilent Technologies, Santa Clara, CA). Mobile phases contained acetonitrile, water, and formic acid (A, 5:95:0.1 by volume; B, 95:5:0.1 by volume), and the flow rate was 0.5 mL/min. The MS system was operated at electrospray ionization-positive ion mode and ion detection performed in the multiple-reaction-monitoring mode. Mass transition of m/z 882.3→638.3, 868.3→636.3, and 271.1→172.0 were monitored for lisaftoclax, venetoclax, and IS, respectively. The ion spray voltage and ionization temperature were set at 5,500 V and 500°C, respectively. Instrument parameters, including curtain gas, Gas 1, and Gas 2 (auxiliary gas), were set at 40, 55, and 55 psi, respectively. Compound parameters, including declustering potential and collision energy, were set at 58 and 41 V, 99 and 38 V, and 80 and 18 V for lisaftoclax, venetoclax, and IS, respectively. Data acquisition and processing were performed using Analyst software version 1.6.3 (AB Sciex).

A calibration curve was generated for sample quantitation. Samples were prepared by mixing a series of working solutions of lisaftoclax or venetoclax with blank cell lysate matrix containing IS, followed by the same processing procedure as described above. The calibration curve was obtained by plotting the peak area ratio of lisaftoclax or venetoclax to IS against the corresponding standard concentrations. Linear regression range was used, and the range of linearity was 1 to 3,000 nmol/L for each agent. Quality-control samples at low (3 nmol/L), medium (240 nmol/L), and high (2,400 nmol/L) concentrations were prepared independently in the same way, with acceptable accuracy (85%–115%).

In vivo PDX model of aggressive WMG

Six- to 8-week-old male mice (founder group) were maintained and pretreated with ibuprofen (4.5 mL/water pouch). We implanted ∼5 mm of tumor tissue subcutaneously, along with 200 μL of Matrigel (Corning; Glendale, AZ), and monitored tumors daily. When xenograft tumors reached a volume of ∼500 to 1,500 mm3, mice were humanely euthanized and tissue fragments transplanted to new mice in order to expand xenograft tissue. Tumor tissue was also maintained in liquid nitrogen in a freezing medium containing 10% FBS, 5% DMSO, and 200 mmol/L trehalose in RPMI1640 medium. Male mice were chosen to sex match the index patient from whom the tumor was derived.

To test the effect of lisaftoclax, tumors extracted from the founder group were implanted subcutaneously into 60 mice (∼5 mm tumor tissue/mouse) and randomized to 4 groups of 8 each on Day 5 for treatments with (i) vehicle (60% phosphatidylcholine concentrate with propylene glycol [Phosal 50], 30% polyethylene glycol 400, and 10% ethanol); (ii) lisaftoclax (100 mg/kg); (iii) ABT-199 (100 mg/kg); and (iv) the Bruton tyrosine kinase (BTK) inhibitor ibrutinib (25 mg/kg; Selleck Chemicals; Houston, TX). From Day 5, all mice received daily treatment, and tumor volumes were measured by caliper every third day using the following formula:

where length represents the largest tumor diameter, and width represents the perpendicular tumor diameter. Percent treatment/control (T/C %) or percent regression (regression %) values were calculated using the above formula. Survival curves were recorded and compared within different treatment groups. Mice were observed for survival and euthanized when tumor burden reached 10% of body weight.

Statistical analysis

Values are expressed as mean ± SEM for normally distributed data and median ± range for non-normally distributed data. Data were analyzed for significance using Prism 6.0, according to paired or unpaired t tests, Mann–Whitney U tests, and/or one-way analysis of variance, with a two-tailed P = 0.0500. Overall survival was determined by Kaplan–Meier analysis. Other detailed protocols are reported in the Supplementary Materials and Methods.

Data and materials availability

All data associated with this study are present in the paper or Supplementary Materials. The authors agree to make additional data supporting the results or analyses presented in their paper available from the corresponding author (Y. Zhai) upon reasonable request.

Lisaftoclax is a novel and selective BCL-2 inhibitor

Studies were conducted to identify new compounds with high affinity of binding to BCL-2 and excellent pharmacokinetic profiles in vivo. These investigations resulted in a series of novel analogs, ultimately showing lisaftoclax to be a highly potent, selective, orally active BCL-2 inhibitor (Fig. 1A).

Figure 1.

Identification of lisaftoclax (APG-2575) as a potent and selective BCL-2 inhibitor. A, Biochemical structures of lisaftoclax and venetoclax (ABT-199). B, Computational modeling of lisaftoclax binding to BCL-2, as predicted on the basis of PDBID:4LVT (navitoclax [ABT-263]). Left, superposition of the lisaftoclax/BCL-2 complex model onto cocrystal structure of venetoclax/BCL-2. The interacting amino acids of BCL-2 with lisaftoclax are highlighted in red. Sites 1 and 2 are displayed to show key differences between lisaftoclax and venetoclax. Right, two alternative structures of venetoclax were resolved from the crystal structure. C, BCL-2–dependent cell lines (RS4;11, HL-60, MV-4–11) were exposed to lisaftoclax or reference compounds for 4 days, and cell proliferation was measured via WST-based CCK-8 proliferation assays. D, BCL-xL dependent cell line SKLU-1 was treated with BCL-2 selective inhibitors (lisaftoclax, venetoclax), a BCL-xL selective inhibitor (WEHI-539), or a BCL-2/BCL-xL inhibitor (navitoclax) for 4 days, and cell proliferation was measured as in C. E, MCL-1-dependent cell line NCI-H23 was treated with BCL-2 selective inhibitors (lisaftoclax, venetoclax) or an MCL-1 inhibitor A1210477 for 4 days, and cell proliferation was measured as in C. CCK-8, Cell Counting Kit-8; WST, water-soluble tetrazolium.

Figure 1.

Identification of lisaftoclax (APG-2575) as a potent and selective BCL-2 inhibitor. A, Biochemical structures of lisaftoclax and venetoclax (ABT-199). B, Computational modeling of lisaftoclax binding to BCL-2, as predicted on the basis of PDBID:4LVT (navitoclax [ABT-263]). Left, superposition of the lisaftoclax/BCL-2 complex model onto cocrystal structure of venetoclax/BCL-2. The interacting amino acids of BCL-2 with lisaftoclax are highlighted in red. Sites 1 and 2 are displayed to show key differences between lisaftoclax and venetoclax. Right, two alternative structures of venetoclax were resolved from the crystal structure. C, BCL-2–dependent cell lines (RS4;11, HL-60, MV-4–11) were exposed to lisaftoclax or reference compounds for 4 days, and cell proliferation was measured via WST-based CCK-8 proliferation assays. D, BCL-xL dependent cell line SKLU-1 was treated with BCL-2 selective inhibitors (lisaftoclax, venetoclax), a BCL-xL selective inhibitor (WEHI-539), or a BCL-2/BCL-xL inhibitor (navitoclax) for 4 days, and cell proliferation was measured as in C. E, MCL-1-dependent cell line NCI-H23 was treated with BCL-2 selective inhibitors (lisaftoclax, venetoclax) or an MCL-1 inhibitor A1210477 for 4 days, and cell proliferation was measured as in C. CCK-8, Cell Counting Kit-8; WST, water-soluble tetrazolium.

Close modal

There are two key structural differences between lisaftoclax (APG-2575) and the reference BCL-2 inhibitor venetoclax (ABT-199; Fig. 1B). At site 1, the dimethyl group was replaced by a cyclobutyl ring, and at site 2 the tetrahydro-2H-pyran group was replaced by 1,4-dioxan. Structure–activity relationship studies established that modifications at sites 1 and 2 were feasible, allowing lisaftoclax to still possess potent binding affinity for BCL-2, exhibit cellular activity in BCL-2–dependent cell lines, and demonstrate an acceptable pharmacokinetic profile in animals. In competitive FP binding assays, lisaftoclax had a very similar profile of binding to BCL-2, BCL-xL, and MCL-1 as compared with venetoclax, with a Ki < 0.1 nmol/L for BCL-2, a weaker affinity of binding to BCL-xL, and a much weaker affinity of binding to MCL-1 (Supplementary Fig. S1).

In vitro cellular viability activity of lisaftoclax was examined in BCL-2–dependent cell lines RS4;11, HL-60, and MV-4–11. IC50 values of lisaftoclax in these cells were 3.6 nmol/L (RS4;11), 2.4 nmol/L (HL-60), and 1.9 nmol/L (MV4;11; each comparable with or lower than venetoclax; Fig. 1C). Despite its nanomolar binding affinity to BCL-xL, lisaftoclax showed minimal effects on cell viability in the SKLU-1 cell line, which depended on BCL-xL antiapoptotic protein for survival, as demonstrated by the cell line's sensitivity to BCL-xL–specific inhibitor WEHI-539 (27) and BCL-2/BCL-xL inhibitor ABT-263 (Fig. 1D, bottom; ref. 11). Consistent with its weak affinity of binding to MCL-1, lisaftoclax also had no effect on the viability of NCI-H23 cells, which were MCL-1–dependent, and cell viability was inhibited by MCL-1 inhibitor A1210477 (Fig. 1E, bottom; ref. 28).

CLL-patient–derived primary samples were more sensitive to lisaftoclax (vs. venetoclax), with IC50 values that were 2-fold lower for lisaftoclax after 4 hours of exposure to each BCL-2 inhibitor (Fig. 2A, left; P = 0.05; Supplementary Table S2A). Additional CLL samples also showed more potent inhibition of cellular viability by lisaftoclax (P = 0.0022 vs. venetoclax; Fig. 2A, right; Supplementary Table S2B). In cellular proliferation assays, the median IC50 of lisaftoclax in multiple myeloma cell lines was 3.6 μmol/L (vs. 4.4 μmol/L for venetoclax), and the median IC50 in WMG cell lines was also lower (i.e., lisaftoclax more potent; 3.0 vs. 7.0 μmol/L; P = 0.0027). In primary human multiple myeloma cells, the median IC50 was ≥2 times lower for lisaftoclax (1.33 μmol/L) compared with venetoclax (3.01 μmol/L; P = 0.0030; Fig. 2B; Supplementary Table S3). In an extended panel of 30 hematologic cancer cell lines, lisaftoclax alone showed activity that was generally equivalent to venetoclax (Fig. 2C; Supplementary Table S4).

Figure 2.

Lisaftoclax (APG-2575) demonstrates potent antiproliferative activity across a variety of hematologic malignancies. A, Two sets of primary samples (CD19+/CD5+ B cells, n = 4 and n = 10, respectively) derived from patients with CLL, were treated with lisaftoclax or venetoclax (ABT-199) for 4 hours and harvested for cell viability assay using a CellTiter-Glo (CTG) kit. *, P = 0.05; **, P = 0.0022 by unpaired t test. B, Patient-derived multiple myeloma/WMG cell lines treated in vitro or ex vivo with 1 μmol/L of either lisaftoclax or venetoclax for 3 days, with viability measured using the CTG kit. C, An extensive panel of hematologic cancer cell lines representing leukemia, lymphoma, and multiple myeloma were exposed to lisaftoclax or venetoclax for 3 days, and cell proliferation was measured using WST-based CCK-8 assays. Mean (± SD) IC50 values from 2 independent experiments (each with triplicate wells) are represented. CCK-8, Cell Counting Kit-8; WST, water-soluble tetrazolium.

Figure 2.

Lisaftoclax (APG-2575) demonstrates potent antiproliferative activity across a variety of hematologic malignancies. A, Two sets of primary samples (CD19+/CD5+ B cells, n = 4 and n = 10, respectively) derived from patients with CLL, were treated with lisaftoclax or venetoclax (ABT-199) for 4 hours and harvested for cell viability assay using a CellTiter-Glo (CTG) kit. *, P = 0.05; **, P = 0.0022 by unpaired t test. B, Patient-derived multiple myeloma/WMG cell lines treated in vitro or ex vivo with 1 μmol/L of either lisaftoclax or venetoclax for 3 days, with viability measured using the CTG kit. C, An extensive panel of hematologic cancer cell lines representing leukemia, lymphoma, and multiple myeloma were exposed to lisaftoclax or venetoclax for 3 days, and cell proliferation was measured using WST-based CCK-8 assays. Mean (± SD) IC50 values from 2 independent experiments (each with triplicate wells) are represented. CCK-8, Cell Counting Kit-8; WST, water-soluble tetrazolium.

Close modal

Lisaftoclax disrupts BCL-2 complexes in a time- and dose-dependent manner and is a true BH3 mimetic

To elucidate the mechanism of action of lisaftoclax, we implemented an advanced electrochemiluminescence assay (ECLA) that quantitates protein–protein complexes formed between proapoptotic and antiapoptotic BCL-2 family members. When treated with either lisaftoclax or venetoclax, RS4;11 [acute lymphoblastic leukemia (ALL)] and Toledo (DLBCL) cell lines, demonstrated a decrease in BCL-2:BIM but not BCL-xL:BIM complex in cell lysates (Fig. 3A). The latter complex was in fact increased by lisaftoclax and venetoclax, probably due to the binding of BIM liberated from BCL-2. Coimmunoprecipitation (Co-IP) assays using SU-DHL-4 (another DLBCL cell line) or Toledo cells confirmed the dose-dependent and selective disruption of BCL-2:BIM complex by lisaftoclax and venetoclax (Fig. 3B). Such rapid, time-dependent disruption of BCL-2:BIM complex by lisaftoclax in RS4;11 cells (by 2–6 hours) coincided with increased apoptosis in these cells (Fig. 3C).

Figure 3.

Lisaftoclax (APG-2575) disrupts BCL-2 complexes in a time- and dose-dependent manner and is a true BH3 mimetic. A, Advanced ECLA was carried out to detect BCL-2:BIM, or BCL-xL:BIM complex in RS4;11 (left) and Toledo (right) cells exposed to lisaftoclax or venetoclax (ABT-199) for 24 hours. B, SU-DHL-4 and Toledo cells were exposed to the specified lisaftoclax and venetoclax concentrations for 24 hours and harvested for immunoprecipitation with BCL-2 antibody and western blotting to detect both BCL-2 and BIM. C, RS4;11 cells were exposed to 0.1 μmol/L of either lisaftoclax or venetoclax as indicated, and time courses show assessment of BCL-2:BIM complex by ECLA (left) and apoptotic Annexin V positive cells detected by flow cytometry analysis (right). D, multiple myeloma cell line KMS-11 and WMG cell line BCWM.1 were exposed to 1 μmol/L of either lisaftoclax or venetoclax for the specified time course and harvested for analysis of BCL-2: BIM protein complex using the MSD ECLA system. **, P < 0.0040 for lisaftoclax by paired t test versus vehicle. E, BH3 profiling as cytochrome c loss (left) and apoptosis test (right) were carried out in four cell lines (O-BCL-2, O-BCL-xL, O-MCL-1, O-BFL1) engineered with overexpression of antiapoptotic BCL-2 family proteins and one additional line carrying BAX and BAK deletions (DKO). Cells were harvested 24 hours after exposure to lisaftoclax.

Figure 3.

Lisaftoclax (APG-2575) disrupts BCL-2 complexes in a time- and dose-dependent manner and is a true BH3 mimetic. A, Advanced ECLA was carried out to detect BCL-2:BIM, or BCL-xL:BIM complex in RS4;11 (left) and Toledo (right) cells exposed to lisaftoclax or venetoclax (ABT-199) for 24 hours. B, SU-DHL-4 and Toledo cells were exposed to the specified lisaftoclax and venetoclax concentrations for 24 hours and harvested for immunoprecipitation with BCL-2 antibody and western blotting to detect both BCL-2 and BIM. C, RS4;11 cells were exposed to 0.1 μmol/L of either lisaftoclax or venetoclax as indicated, and time courses show assessment of BCL-2:BIM complex by ECLA (left) and apoptotic Annexin V positive cells detected by flow cytometry analysis (right). D, multiple myeloma cell line KMS-11 and WMG cell line BCWM.1 were exposed to 1 μmol/L of either lisaftoclax or venetoclax for the specified time course and harvested for analysis of BCL-2: BIM protein complex using the MSD ECLA system. **, P < 0.0040 for lisaftoclax by paired t test versus vehicle. E, BH3 profiling as cytochrome c loss (left) and apoptosis test (right) were carried out in four cell lines (O-BCL-2, O-BCL-xL, O-MCL-1, O-BFL1) engineered with overexpression of antiapoptotic BCL-2 family proteins and one additional line carrying BAX and BAK deletions (DKO). Cells were harvested 24 hours after exposure to lisaftoclax.

Close modal

We then investigated the ability of lisaftoclax to disrupt BCL-2:BIM complex formation in multiple myeloma and WMG models using ECLA. Similar to findings in RS4;11 and Toledo cells, we observed rapid and time-dependent disruption of BCL-2:BIM complexes in KMS-11 and BCWM.1 (WMG) cell lines. Lisaftoclax reduced BCL-2:BIM complexes by more than 50% within 12 hours [P = 0.0020 (KMS-11) and P = 0.0022 (BCWM.1)], with less than 10% complex remaining by 24 hours (P = 0.0032 and P = 0.0015, respectively vs. vehicle; Fig. 3D).

Further assessment of lisaftoclax was conducted by direct mitochondrial exposure using four cell lines with enforced expression of human BCL-2 (O-BCL-2), BCL-xL (O-BCL-xL), MCL-1 (O-MCL-1), or BFL-1/A1 (O-BFL-1/A1), as well as one additional line with homozygous deletions of both BAX and BAK [O-double knockout (DKO); ref. 29]. Lisaftoclax induced cytochrome c release from O-BCL-2 mitochondria at a concentration that was 2 orders of magnitude lower than in O-BCL-xL mitochondria (Fig. 3E). Lisaftoclax had no effect on cytochrome c release in O-MCL-1 or O-BFL-1/A1 mitochondria and did not trigger cytochrome c release in O-DKO (BAX/BAK DKO) mitochondria. Cellular toxicity assays demonstrated that lisaftoclax had greater activity in killing O-BCL-2 cells (vs. O-BCL-xL, O-MCL-1, and O-BFL1), but this activity was abolished in O-DKO and other cells (Fig. 3E).

Lisaftoclax enters cells more rapidly than venetoclax and is more effective in its apoptosis-inducing activity in multiple myeloma and WMG models

In RPMI8226 (multiple myeloma) cells, cellular uptake essays showed that lisaftoclax entered tumor cells rapidly, with intracellular concentrations reaching a plateau in 2 hours that was sustained for the 6-hour incubation (Fig. 4A). Given that lisaftoclax entered RPMI8226 cells more rapidly, 1.44- to 1.61-fold higher intracellular concentrations were reached at each time point as compared with venetoclax (P < 0.0500). A similar pattern was observed with KMS-11 (multiple myeloma) cells (Fig. 4A), with intracellular lisaftoclax concentrations that were 1.46- to 1.92-fold higher (P < 0.0500 vs. venetoclax).

Figure 4.

Lisaftoclax (APG-2575) demonstrates more potent apoptosis-inducing activity and more severely impairs mitochondrial function compared with venetoclax (ABT-199) in multiple myeloma and WMG models. A, Cellular uptake assays for RPMI8226 and KMS-11 cells exposed to 1 μmol/L of lisaftoclax and venetoclax. Data are expressed as mean ± SD. *, P < 0.0500; **, P < 0.0100; ***, P < 0.0010 by paired t tests for lisaftoclax versus venetoclax at the same conditions (n = 3 independent experiments). B, Caspase-3/7 activation measured by flow cytometry analysis in multiple myeloma or WMG cells after treatment with 1 μmol/L of either lisaftoclax or venetoclax for 24 hours. Results are means from multiple myeloma cell lines (KMS-11, KMS-12BM, KMS-12PE) and WMG cell lines (BCWM.1, MWCL.1, RPCI-WM.1). *, P = 0.0022 and P = 0.0023, respectively, by paired t tests comparing lisaftoclax with venetoclax. C, MOMP was measured in primary samples derived from patients (pt.) with multiple myeloma (n = 3) or WMG (n = 3) in triplicate, using MitoSpy Red CMXRos mitochondrial probe dye (BioLegend), after 24 hours treatment. *, P < 0.0500 by paired t tests comparing lisaftoclax with vehicle for multiple myeloma and either lisaftoclax or venetoclax compared with vehicle for WMG. D, Primary samples derived from pt. with multiple myeloma or WMG were exposed ex vivo to 1 μmol/L specified agent for 3 days, and apoptosis was measured by flow cytometry after Annexin V–PI staining. Lisaftoclax compared with vehicle with **, P ≤ 0.0010 by paired t test; *, P = 0.0015 and *, P = 0.0043 by paired t test comparing lisaftoclax with venetoclax in multiple myeloma and WMG primary samples, respectively. E, KMS-11 and BCWM.1 cells were exposed to lisaftoclax or venetoclax (1 μmol/L) before being harvested for BIM and Noxa protein analysis. ****, P < 0.0001 by paired t test for BIM (pre- vs. post-lisaftoclax); ***, P = 0.0004 (KMS-11 cell), and **, P = 0.0027 (BCWM.1 cells) by paired t test for Noxa (pre- vs. post-lisaftoclax). F, KMS-11 cells were exposed to vehicle, lisaftoclax, or venetoclax (1 μmol/L each) for 4 hours and stained with MitoTracker Red and DAPI, then probed with an anti-BIM antibody and detected by Alexa Fluor 488 goat antirabbit IgG. Data are representative of 3 individual experiments. Magnifications: 10x (bottom) and 40x (top). G, OCR was measured in KMS-11 cells exposed to vehicle, lisaftoclax or venetoclax (1 μmol/L each) for 24 hours. OCR is displayed as values normalized to total protein content. H, Maximal and nonmitochondrial respiration, spare respiratory capacity, as well as mitochondrial ATP production rate, were calculated at the end of the experiment in G. *, P < 0.0500 by paired t test comparing lisaftoclax with vehicle for spare mitochondrial respiratory capacity (P = 0.0150) and nonmitochondrial respiration (P = 0.0035); **, P = 0.0035 by paired t test comparing mitochondrial ATP production with lisaftoclax versus vehicle. DAPI, 4′,6-diamidino-2-phenylindole.

Figure 4.

Lisaftoclax (APG-2575) demonstrates more potent apoptosis-inducing activity and more severely impairs mitochondrial function compared with venetoclax (ABT-199) in multiple myeloma and WMG models. A, Cellular uptake assays for RPMI8226 and KMS-11 cells exposed to 1 μmol/L of lisaftoclax and venetoclax. Data are expressed as mean ± SD. *, P < 0.0500; **, P < 0.0100; ***, P < 0.0010 by paired t tests for lisaftoclax versus venetoclax at the same conditions (n = 3 independent experiments). B, Caspase-3/7 activation measured by flow cytometry analysis in multiple myeloma or WMG cells after treatment with 1 μmol/L of either lisaftoclax or venetoclax for 24 hours. Results are means from multiple myeloma cell lines (KMS-11, KMS-12BM, KMS-12PE) and WMG cell lines (BCWM.1, MWCL.1, RPCI-WM.1). *, P = 0.0022 and P = 0.0023, respectively, by paired t tests comparing lisaftoclax with venetoclax. C, MOMP was measured in primary samples derived from patients (pt.) with multiple myeloma (n = 3) or WMG (n = 3) in triplicate, using MitoSpy Red CMXRos mitochondrial probe dye (BioLegend), after 24 hours treatment. *, P < 0.0500 by paired t tests comparing lisaftoclax with vehicle for multiple myeloma and either lisaftoclax or venetoclax compared with vehicle for WMG. D, Primary samples derived from pt. with multiple myeloma or WMG were exposed ex vivo to 1 μmol/L specified agent for 3 days, and apoptosis was measured by flow cytometry after Annexin V–PI staining. Lisaftoclax compared with vehicle with **, P ≤ 0.0010 by paired t test; *, P = 0.0015 and *, P = 0.0043 by paired t test comparing lisaftoclax with venetoclax in multiple myeloma and WMG primary samples, respectively. E, KMS-11 and BCWM.1 cells were exposed to lisaftoclax or venetoclax (1 μmol/L) before being harvested for BIM and Noxa protein analysis. ****, P < 0.0001 by paired t test for BIM (pre- vs. post-lisaftoclax); ***, P = 0.0004 (KMS-11 cell), and **, P = 0.0027 (BCWM.1 cells) by paired t test for Noxa (pre- vs. post-lisaftoclax). F, KMS-11 cells were exposed to vehicle, lisaftoclax, or venetoclax (1 μmol/L each) for 4 hours and stained with MitoTracker Red and DAPI, then probed with an anti-BIM antibody and detected by Alexa Fluor 488 goat antirabbit IgG. Data are representative of 3 individual experiments. Magnifications: 10x (bottom) and 40x (top). G, OCR was measured in KMS-11 cells exposed to vehicle, lisaftoclax or venetoclax (1 μmol/L each) for 24 hours. OCR is displayed as values normalized to total protein content. H, Maximal and nonmitochondrial respiration, spare respiratory capacity, as well as mitochondrial ATP production rate, were calculated at the end of the experiment in G. *, P < 0.0500 by paired t test comparing lisaftoclax with vehicle for spare mitochondrial respiratory capacity (P = 0.0150) and nonmitochondrial respiration (P = 0.0035); **, P = 0.0035 by paired t test comparing mitochondrial ATP production with lisaftoclax versus vehicle. DAPI, 4′,6-diamidino-2-phenylindole.

Close modal

Consistent with the rapid cellular uptake of lisaftoclax, release of cytochrome c into the cytoplasm triggered by lisaftoclax was stronger compared with venetoclax, as indicated by immunoblot analysis (Supplementary Fig. S2A). Significantly higher downstream caspase-3/7 (proapoptotic) activity was further demonstrated with lisaftoclax in multiple myeloma (P = 0.0022) and WMG cell lines (P = 0.00230; each vs. venetoclax; Fig. 4B). Median lisaftoclax-induced apoptosis was also significantly higher (vs. venetoclax) in multiple myeloma (72.92% vs. 37.04%; P = 0.0022) and WMG (60.20% vs. 50.00%; P = 0.0411) cell lines (Supplementary Fig. S2B).

Characterization of lisaftoclax was further performed in primary tumor cells derived from patients with multiple myeloma or WMG. Mitochondrial outer membrane permeabilization (MOMP), a key biomarker for initiation of apoptosis, was significantly increased in cells exposed to lisaftoclax, represented by a decrease in mitochondrial depolarization (Fig. 4C). Compared with venetoclax, apoptosis resulting from lisaftoclax treatment was increased and significantly higher in primary tumor cells derived from patients with multiple myeloma (61.68% vs. 53.28%; P = 0.0015) or WMG (73.84% vs. 58.00%; P = 0.0043; Fig. 4D).

Western blot analysis of BCL-2 family proteins revealed that levels of proapoptotic proteins BIM and Noxa were rapidly increased by lisaftoclax (Fig. 4E; Supplementary Fig. S2C and S2D). In a time-course analysis of KMS-11 and BCWM.1 cells, BIM significantly increased within 2 hours after lisaftoclax treatment onset (P < 0.0001 vs. vehicle for each) and decreased by 6 hours. Treatment with lisaftoclax also significantly increased Noxa expression within 1 to 6 hours (KMS-11 P = 0.0004; BCWM.1 P = 0.0027 vs. vehicle). MCL-1 levels increased modestly in KMS-11 but not in BCWM.1 cells (Supplementary Fig. S2E and S2F). As a measure of the role of BIM in initiating apoptosis, more BIM was observed to be accumulated in mitochondria of KMS-11 cells after exposure to lisaftoclax (vs. vehicle or venetoclax; Fig. 4F).

Intrigued by the ability of lisaftoclax to trigger the mitochondrial apoptotic signaling cascade, we assessed its potential impact on mitochondrial bioenergetics by measuring oxygen consumption rate (OCR). Maximal respiration, which was achieved when cells were treated with the mitochondrial uncoupler carbonyl cyanide-4-phenylhydrazone (FCCP), was significantly lower in KMS-11 cells exposed to lisaftoclax (P = 0.0027 vs. venetoclax; P = 0.0023 vs. vehicle; Fig. 4G). Nonmitochondrial respiration (P = 0.0035) and spare respiratory capacity (P = 0.0150) were also significantly decreased in cells exposed to lisaftoclax (vs. vehicle). These changes resulted in a significant reduction in mitochondrial ATP production within lisaftoclax-treated cells (P = 0.0035 vs. vehicle; Fig. 4H), supporting the ability of lisaftoclax to induce damage to other mitochondrial functions when initiating apoptosis.

Lisaftoclax suppresses in vivo growth of hematologic cancers and increases survival in cell line–derived xenograft and PDX models

In a BCL-2–dependent RS4;11 xenograft murine model of ALL (Fig. 5A), tumor growth inhibition was noted after oral administration of lisaftoclax 6.25 mg/kg once daily by Day 15 (T/C 60%), peaking at a 100-mg/kg dose (T/C 4%), without apparent changes in body weight (Fig. 5B). A Toledo xenograft model of DLBCL further confirmed that tumor growth inhibition was proportional to lisaftoclax dose, with the greatest inhibition at 100 mg/kg (Fig. 5C).

Figure 5.

Lisaftoclax (APG-2575) suppresses in vivo growth of hematologic cancers and increases survival in cell line xenografts and a PDX model. A, Mean (± SEM) tumor volumes in an RS4;11 ALL xenograft model exposed to different oral once-daily doses of lisaftoclax or venetoclax (ABT-199; 25 mg/kg) for 14 days (measured on indicated days); n = 6 mice per treatment group. B, Mean (± SD) murine body weight were presented for Days 0, 5, 8, 12, and 15 from the same study shown in A. C, Mean (± SEM) tumor volumes in a DLBCL xenograft model (Toledo) exposed to different doses of lisaftoclax and venetoclax (100 mg/kg) (measured on indicated days). D, Mean (± SEM) tumor volumes in a DLBCL xenograft model (OCI-LY8) exposed to lisaftoclax, alone, or combined with bendamustine and rituximab (B + R), and lisaftoclax plus B + R (BR); n = 5 to 6 mice per treatment group. E, NSG mice were implanted with KMS-11 (multiple myeloma) cells transduced with a luciferase reporter construct and randomized to receive vehicle, or lisaftoclax or venetoclax 100 mg/kg daily for 15 days (n = 10 per group). Tumor burden was measured by bioluminescent imaging (total flux). *, P < 0.0500 (lisaftoclax vs. vehicle). F, Tumor tissues from mice in (E) were stained for cleaved caspase-3 (CC3), and CC3 was quantitated using the Aperio image software system (Digital-CC3, D-CC3). *, P < 0.0500 (lisaftoclax or venetoclax vs. vehicle). G and H, PDX model derived from a patient with ibrutinib-resistant WMG was established in NSG mice and treated once daily with lisaftoclax (100 mg/kg), venetoclax (100 mg/kg), or ibrutinib (25 mg/kg) for 29 days. Tumor volumes were measured and are presented as mean (± SEM) in G. ***, P = 0.0002 (lisaftoclax vs. venetoclax); ***, P = 0.0003 (lisaftoclax vs. ibrutinib). A survival curve for each treatment group in G is shown in H. **, P = 0.002 (lisaftoclax vs. venetoclax). NSG, NOD/SCID gamma (mice).

Figure 5.

Lisaftoclax (APG-2575) suppresses in vivo growth of hematologic cancers and increases survival in cell line xenografts and a PDX model. A, Mean (± SEM) tumor volumes in an RS4;11 ALL xenograft model exposed to different oral once-daily doses of lisaftoclax or venetoclax (ABT-199; 25 mg/kg) for 14 days (measured on indicated days); n = 6 mice per treatment group. B, Mean (± SD) murine body weight were presented for Days 0, 5, 8, 12, and 15 from the same study shown in A. C, Mean (± SEM) tumor volumes in a DLBCL xenograft model (Toledo) exposed to different doses of lisaftoclax and venetoclax (100 mg/kg) (measured on indicated days). D, Mean (± SEM) tumor volumes in a DLBCL xenograft model (OCI-LY8) exposed to lisaftoclax, alone, or combined with bendamustine and rituximab (B + R), and lisaftoclax plus B + R (BR); n = 5 to 6 mice per treatment group. E, NSG mice were implanted with KMS-11 (multiple myeloma) cells transduced with a luciferase reporter construct and randomized to receive vehicle, or lisaftoclax or venetoclax 100 mg/kg daily for 15 days (n = 10 per group). Tumor burden was measured by bioluminescent imaging (total flux). *, P < 0.0500 (lisaftoclax vs. vehicle). F, Tumor tissues from mice in (E) were stained for cleaved caspase-3 (CC3), and CC3 was quantitated using the Aperio image software system (Digital-CC3, D-CC3). *, P < 0.0500 (lisaftoclax or venetoclax vs. vehicle). G and H, PDX model derived from a patient with ibrutinib-resistant WMG was established in NSG mice and treated once daily with lisaftoclax (100 mg/kg), venetoclax (100 mg/kg), or ibrutinib (25 mg/kg) for 29 days. Tumor volumes were measured and are presented as mean (± SEM) in G. ***, P = 0.0002 (lisaftoclax vs. venetoclax); ***, P = 0.0003 (lisaftoclax vs. ibrutinib). A survival curve for each treatment group in G is shown in H. **, P = 0.002 (lisaftoclax vs. venetoclax). NSG, NOD/SCID gamma (mice).

Close modal

A combination of lisaftoclax with anti-CD20 antibody rituximab in OCI-LY8 (DLBCL) tumor-bearing mice achieved better responses than either single agent, delaying tumor regrowth (Supplementary Fig. S3A). Also, in the OCI-LY8 model, a ternary combination of lisaftoclax, bendamustine, and rituximab (BR) yielded sustained tumor regression and longer duration of response [6/6 mice, 100% complete response (CR)], based on the modified Response Evaluation Criteria in Solid Tumors for animal studies (30), when compared with antitumor effects with lisaftoclax alone or BR dual combination (Fig. 5D). Further, in vivo target engagement of lisaftoclax on BCL-2 was tested in a xenograft model of AML (OCI-AML-3). Tumor tissues collected from lisaftoclax-treated tumor-bearing mice on Day 12 demonstrated a significant reduction of the BCL-2:BIM complex compared with vehicle control (Supplementary Fig. S3B), even in this insensitive model, because of high MCL-1 protein expression (31).

Using a 100-mg/kg dose, we next investigated the effect of lisaftoclax in a multiple myeloma xenograft model (KMS-11). Tumor burden was significantly reduced in the lisaftoclax-treated group by Day 20, as measured by bioluminescent signal intensity from Luc-labeled tumor cells (total flux; P = 0.0010 vs. vehicle), though there was no statistical difference between lisaftoclax- and venetoclax-treated mice (Fig. 5E). Lisaftoclax was well tolerated, with no significant decrease in body weight. Kaplan–Meier analysis demonstrated significantly longer survival in mice treated with lisaftoclax (median survival 25 vs. 21 days with vehicle control; P < 0.0500; Supplementary Fig. S3C). IHC analysis of staining intensity from resected lisaftoclax-treated tumors showed decreased Ki67 but significantly increased cleaved caspase-3 (P = 0.0220 vs. vehicle; Fig. 5F; Supplementary Fig. S3D).

In a WMG xenograft model (BCWM.1), a T/C ratio of 25.12% (with no apparent toxicities or weight loss) was evident after lisaftoclax treatment by Day 15. Median survival was longer in mice receiving lisaftoclax (29 days) compared with vehicle (21 days; P = 0.0190). Increased IHC staining intensity of cleaved caspase-3 (P = 0.0025 vs. vehicle) was observed in tumor tissues, confirming enhanced apoptosis in BCWM.1 tumor cells from lisaftoclax-treated mice (Supplementary Fig. S3E–S3G).

In addition to human cancer cell line-derived xenografts, a PDX model was established from a patient with ibrutinib-resistant WMG (Fig. 5G and H). Lisaftoclax was administered for 29 days to closely mimic one cycle of patient treatment. Mice exposed to lisaftoclax showed significantly lower tumor burden (T/C 17.56%) compared with venetoclax (P = 0.0002) or the BTK inhibitor ibrutinib (P = 0.0003; Fig. 5G). Correspondingly, mice treated with lisaftoclax showed significantly greater survival (63 days) compared with venetoclax (36 days; P = 0.002) or the other groups (vehicle 34 days; ibrutinib 38 days; Fig. 5H).

Pharmacodynamics and pharmacokinetics of lisaftoclax in RS4;11 xenograft model

In an RS4;11 xenograft model, mice received one oral dose of lisaftoclax or venetoclax 6.25 mg/kg before tumor samples were collected for pharmacodynamic analysis. Cleaved caspase-3 was induced rapidly after 4 hours and peaked at 24 hours of lisaftoclax treatment. The increase in cleaved caspase-3 was accompanied by induction of poly (ADP-ribose) polymerase 1 (PARP-1) cleavage, indicating comparable apoptosis-inducing activity triggered by lisaftoclax or venetoclax (Fig. 6A).

Figure 6.

Pharmacodynamics and pharmacokinetics of lisaftoclax (APG-2575) in an RS4;11 xenograft model. A, Mice in an RS4;11 xenograft model received a single oral dose of lisaftoclax or venetoclax (ABT-199) 6.25 mg/kg. Tumor samples were collected at prespecified times for Western blot analysis of caspase-3 activation and PARP-1 cleavage triggered by lisaftoclax (left) and venetoclax (right). B, Mice in an RS4;11 xenograft model were treated once with different doses of lisaftoclax or venetoclax. Plasma and tumor samples were collected at various time points for pharmacokinetic analysis shown by time versus concentration graphs.

Figure 6.

Pharmacodynamics and pharmacokinetics of lisaftoclax (APG-2575) in an RS4;11 xenograft model. A, Mice in an RS4;11 xenograft model received a single oral dose of lisaftoclax or venetoclax (ABT-199) 6.25 mg/kg. Tumor samples were collected at prespecified times for Western blot analysis of caspase-3 activation and PARP-1 cleavage triggered by lisaftoclax (left) and venetoclax (right). B, Mice in an RS4;11 xenograft model were treated once with different doses of lisaftoclax or venetoclax. Plasma and tumor samples were collected at various time points for pharmacokinetic analysis shown by time versus concentration graphs.

Close modal

Pharmacokinetic analysis was carried out in similar models of RS4;11-bearing mice receiving one oral dose of lisaftoclax 6.25 mg/kg, 25 mg/kg, or 100 mg/kg, and venetoclax 25 mg/kg as a control. Dose-dependent increases in lisaftoclax were observed in both plasma and tumors. Mice dosed with lisaftoclax 100 mg/kg had a plasma Cmax of 7,803.3 ng/mL and AUC0-last of 123,401.2 hr•ng/mL (Supplementary Table S5). Lisaftoclax and venetoclax 25 mg/kg showed similar elimination half-life (t1/2) values in plasma: 5.2 to 6.0 and 6.3 hours, respectively. In tumors, the t1/2 ranged from 13.2 to 48.1 hours with the three lisaftoclax doses, as compared with 18.1 hours with venetoclax 25 mg/kg (Fig. 6B; Supplementary Table S5).

Effect of lisaftoclax on hematology and hematopoiesis

To evaluate the effect of lisaftoclax on functional hematopoiesis, bone marrow samples were collected from normal inbred BALB/c (H2d) female mice and seeded in a 96-well plate for treatments with DMSO (vehicle control) and increasing concentrations of lisaftoclax and venetoclax. After 24 and 72 hours of treatment, viability analysis showed that IC50 values for lisaftoclax were approximately 20 μmol/L and 10 μmol/L, respectively. On the basis of IC50 values, cells were similarly sensitive to lisaftoclax and venetoclax at both 48- and 72-hour time points (Supplementary Fig. S4).

Further assessments were conducted in male and female CD-1 mice that were orally administered lisaftoclax at 100, 300, and 1,000 mg/kg daily for 28 days. Mild-to-moderate hematologic effects related to lisaftoclax were observed only in mice administered 1,000 mg/kg/day, with fewer findings noted in animals receiving 100 or 300 mg/kg/day (Supplementary Table S6A). The changes included mildly lower red cell mass in animals administered 1,000 mg/kg/day; mildly lower mean corpuscular hemoglobin (MCH) and MCH concentration in males administered ≥300 mg/kg/day and females administered ≥100 mg/kg/day; and a mildly-to-moderately lower lymphocyte count, resulting in a lower white blood cell count, in animals administered ≥ 100 mg/kg/day.

Similar to the hematology test results, only a few minor, potentially lisaftoclax-related findings were noted in the bone marrow smears of male mice administered 1,000 mg/kg/day. The findings included a mildly higher total erythroid count, which was due to a higher metarubricyte count, and a mildly lower lymphocyte count (Supplementary Table S6B).

It has been challenging to develop BCL-2 inhibitors for cancer therapy because of the inherent difficulty in disrupting intracellular, protein–protein interactions of the BCL-2 family proteins with high precision. Certain inhibitors have been reported to antagonize BCL-2 function in preclinical models, including those that decrease BCL-2 protein level; however, they do not possess features of BH3 mimetics. Currently, there is only one BCL-2 inhibitor (a BH3 mimetic) approved by the FDA. BH3 profiling has recently emerged as a functional platform to determine if a BCL-2 inhibitor is a bona fide BH3 mimetic (29). This methodology enabled us to measure BCL-2 family protein–protein interactions, which determine mitochondrial priming and the threshold of cells committed to apoptosis (6, 32, 33). On the basis of this unique mechanism of action, it has been suggested that BH3-mimetic BCL-2 inhibitors need to rapidly reach a mitochondrial threshold to trigger apoptosis; their effects are thus driven by the rate of cell penetration and maximum concentration (Cmax; ref. 3).

On the basis of structure–activity relationship studies, we have discovered lisaftoclax, a new BCL-2 inhibitor with high binding affinity and selectivity for the BCL-2 protein. Relative to more sensitive approaches such as homogeneous time-resolved fluorescence, which combines fluorescence resonance energy transfer with time-resolved measurement, the competitive FP binding assay demonstrated only a five-fold difference in affinity between BCL-2 and BCL-xL for lisaftoclax or venetoclax, which may be due to a limitation of the assay (26). Consistent with mechanisms of action ascribed to true BH3 mimetics, we have used BH3 profiling and cell death assays to demonstrate that lisaftoclax triggers cytochrome c release and apoptosis in BCL-2–dependent cell lines in a BAX/BAK–dependent manner. Knockdown of BAX and BAK completely blocks cytochrome c release and apoptosis in tumor cells treated with lisaftoclax. This mode of action coincides with disruption of BCL-2:BIM complexes. Once liberated from BCL-2, BIM can trigger BAX/BAK activation to increase MOMP, leading to apoptosis within 1 to 6 hours. Sample analysis using cancer cell lines and mouse xenograft models have shown that on-target engagement by lisaftoclax for the BCL-2:BIM complex underlies its antiproliferative and overall antitumor activities. A range of B-cell lymphoid cancers are sensitive to lisaftoclax, although there is some variation across the panel of cancer cells tested (as would be expected), with CLL and certain types of DLBCL (OCI-LY1), ALL (RS4;11), FL (DOHH2), and AML (MOLM-13) cells being the most sensitive.

Although lisaftoclax and venetoclax share a similar mechanism of action, the magnitudes of their anti-BCL-2 effects may differ. Treatment with lisaftoclax led to stronger caspase 3/7 activation and a greater extent of apoptosis-inducing activity in multiple myeloma and WMG cell lines and patient-derived primary samples. These features of lisaftoclax can likely be attributed to: (i) more rapid cellular uptake, with a higher accumulation in multiple myeloma cells (vs. venetoclax); (ii) more potent disruption of the BCL-2:BIM complex; (iii) stronger induction (by lisaftoclax) of prodeath BH3-only protein BIM and its translocation from the cytosol to the mitochondria; and/or (iv) more marked increases in levels of prodeath BH3-only protein Noxa (with lisaftoclax), which were not changed significantly by venetoclax. In addition to these cellular and molecular disparities between lisaftoclax and venetoclax, a PDX model derived from a patient with an aggressive form of WMG refractory to ibrutinib therapy illuminated a potentially clinically relevant distinction. Treatment with lisaftoclax markedly inhibited WMG tumor growth, leading to significantly prolonged survival in the model. In contrast, venetoclax displayed minimal antitumor activity. The biological mechanism underlying this difference is currently under investigation.

Lisaftoclax showed dose-dependent systemic and tumor tissue exposure in an RS4;11 xenograft model, with a slightly higher Cmax than that of venetoclax but with a similar plasma t1/2. The minimal effective dose of lisaftoclax was 6.25 mg/kg, which led to caspase-3 activation and PARP-1 cleavage as early as 4 hours and peaked at 24 hours after lisaftoclax treatment. While lisaftoclax demonstrated single-agent activity in an RS4;11 xenograft model, combination treatments revealed more pronounced tumor growth inhibition in a DLBCL xenograft model (OCI-LY8). When combined with rituximab, or rituximab and bendamustine, lisaftoclax exhibited a longer duration and deeper response (i.e., CR) compared with single or double agents, suggesting that combination therapies combining lisaftoclax with standard-of-care treatments may be more effective in certain types of cancers, such as lymphoma with heterogeneous dependencies on oncogenic or antiapoptotic proteins other than BCL-2.

The key mechanism of action of lisaftoclax is to disrupt the BCL-2 complex and antagonize its antideath function to resume apoptosis in cancer cells. Initiation of apoptosis requires activation of BAX/BAK with oligomerization for pore formation, leading to MOMP. Besides the canonical pathways central to the BCL-2 family of proteins, we also uncovered that lisaftoclax treatment reduced both mitochondrial and nonmitochondrial respiratory capacity, and decreased mitochondrial ATP production, as examined in multiple myeloma cells. Further, lisaftoclax induced more marked reductions in mitochondrial function compared with venetoclax in multiple myeloma and/or WMG cells. These findings suggest a wider cellular stress response triggered by damaged mitochondrion due to lisaftoclax and are aligned with a recent report showing that the electron transport chain complex was altered by venetoclax in T cells (34), in order to augment antileukemic activity via increased generation of reactive oxygen species.

Preliminary toxicity assessments of normal mouse hematology and hematopoiesis, both in vitro and in vivo, suggest that normal bone marrow tolerates lisaftoclax well. Low lymphocyte counts in peripheral blood and bone marrow samples after lisaftoclax treatment is likely related to the pharmacology of BCL-2 inhibitors, which would inhibit B cells and subsets of T cells (35). The high metarubricyte count observed in bone marrow smears of organisms treated with lisaftoclax may represent an early compensatory response to the lower red cell mass or a shift to mature erythroid precursors, but the exact mechanism was not determined. It is worth noting that hematology and bone marrow differential findings exhibited reversibility at the end of another 28-day recovery phase.

In conclusion, we have discovered a bona fide BH3 mimetic, BCL-2–selective inhibitor with potential biological and therapeutic relevance in several preclinical models of hematologic malignancies, including CLL, ALL, DLBCL, multiple myeloma, and WMG. Moreover, lisaftoclax treatment overcame BTK inhibitor resistance and inhibited tumor growth in a PDX model established from a patient with aggressive WMG that relapsed after ibrutinib. (For a summary of our working model, see Supplementary Fig. SS5.) Our findings show that lisaftoclax triggers mitochondria-mediated, BAX/BAK–dependent apoptosis in cell-based and xenograft models, with broad effects on mitochondrial functions.

Our findings should be considered in the context of potential study limitations. Although we have demonstrated that lisaftoclax confers stronger apoptosis-inducing and antitumor activity (vs. venetoclax) in multiple myeloma, WMG, and CLL models, activities of these BCL-2–selective inhibitors are equivalent in other models such as AML. Additional studies are warranted to further differentiate the agents in cancer-type or condition-specific models. Consistent with these encouraging preclinical results, several phase Ib and phase II clinical trials have been initiated and are currently ongoing to evaluate lisaftoclax in hematologic cancers as monotherapy or in combination with an anti-CD20 antibody or BTK inhibitor (NCT03537482; NCT04260217).

J. Deng was a full-time employee of and/or equity shareholder in Ascentage Pharma Group Inc. A. Paulus reports grants from Ascentage and personal fees from Ascentage during the conduct of the study; nonfinancial support and other support from Alpha2 Pharmaceuticals outside the submitted work. J. Chen reports grants from Ascentage during the conduct of the study, as well as a patent for BCL-2 inhibitors issued, licensed, and with royalties paid from Ascentage. L. Bai reports patents for US 8691184, US 9096625, and US 9403856 issued, licensed, and with royalties paid from Ascentage. D. McEachern reports a patent for WO2012103059A2 issued, licensed, and with royalties paid from Ascentage, a patent for WO/2006/050447 issued, a patent for WO/2006/099193 issued, and a patent for WO2012103059 issued, licensed, and with royalties paid from Ascentage. C.Y. Yang reports a patent for WO2012103059A2 issued, licensed, and with royalties paid from Ascentage, a patent for WO/2006/050447 issued, a patent for WO/2006/099193 issued, and a patent for US20120189539 issued, licensed, and with royalties paid from Ascentage. J. Ryan reports personal fees from Zentalis Pharmaceuticals outside the submitted work, as well as a patent for BH3 Profiling issued, licensed, and with royalties paid from Zentalis Pharmaceuticals. A. Letai reports personal fees from Zentalis Pharmaceuticals, Dialectic Therapeutitics, and Flash Therapeutics, as well as personal fees from Anji Onco outside the submitted work; in addition, A. Letai has a patent for BH3 profiling owned by Dana-Farber pending, issued, licensed, and with royalties paid from Zentalis and Dialectic. S. Ailawadhi reports grants from Ascentage during the conduct of the study; grants from GSK, AbbVie, Xencor, Janssen, Cellectar, Amgen, Pharmacyclics, and BMS; personal fees from Sanofi and Takeda; and personal fees from Beigene outside the submitted work. D. Yang reports personal fees from Ascentage during the conduct of the study; has patents issued for US8557812, US 163805, US20090092684A1, US7432304B2, US7354928B2, US20060084647A1, and US20040214902A1; is a board member within the funding institution; and owns stock/equity in Ascentage (HK: 6855). S. Wang reports grants, personal fees, and other support from Ascentage during the conduct of the study; grants, personal fees, and other support from Oncopia Therapeutics; grants and other support from Roivant Sciences; grants and personal fees from Proteovant Therapeutics; other support from Debiopharm; and personal fees from American Chemical Society outside the submitted work. In addition, S. Wang has a patent 10,829,488 issued, licensed, and with royalties paid from Ascentage; and is a member of the Board of Directors of Ascentage and a paid consultant of Ascentage. A. Chanan-Khan reports other support from Ascentage during the conduct of the study and outside the submitted work. In addition, A. Chanan-Khan has a patent for AT101 issued and licensed to Alpha2; and serves as the Chairman and CEO of Alpha2, which has a codevelopment agreement with Ascentage for their molecule AT-101. No disclosures were reported by the other authors.

J. Deng: Conceptualization and study design, data acquisition and interpretation, writing–original draft, review, editing, and approval; and investigation (led preclinical research, including mechanism of action, target engagement and apoptosis assay). A. Paulus: Conceptualization and study design, data acquisition and interpretation, writing–original draft, review, editing, and approval; and investigation (led the preclinical study of multiple myeloma [MM] and Waldenström macroglobulinemia [WM]). D.D. Fang: Conceptualization and study design, data acquisition and interpretation, writing–review, editing, and approval; and investigation (led preclinical research, including xenograft efficacy and pharmacokinetic and pharmacodynamic studies). A. Manna: Performed animal experiments and ECLA; data acquisition and interpretation; writing–review, editing, and approval; and investigation (MM and WM preclinical study). G. Wang: Data acquisition and interpretation, writing–review, editing, and approval; and investigation (led preclinical research, including in vitro and in vivo pharmacology studies). H. Wang: Data acquisition and interpretation, writing–review, editing, and approval; and investigation (preclinical research of pharmacokinetics study). S. Zhu: Data acquisition, interpretation, writing–review, editing, and approval; and investigation (preclinical research of cellular uptake in MM). J. Chen: Data acquisition and interpretation, writing–review, editing, and approval; and investigation (discovery chemistry, FP binding assay). P. Min: Data acquisition and interpretation, writing–review, editing, and approval; and investigation (led preclinical research, including in vitro and in vivo pharmacology studies). Y. Yin: Data acquisition, interpretation, writing–review, editing, and approval; and investigation (preclinical research on mechanism of action and target engagement). N. Dutta: Data acquisition and interpretation, writing–review, editing, and approval; and investigation (led the MM and WM preclinical study and performed the coimmunoprecipitation and drug synergy testing). N. Halder: Data acquisition and interpretation, writing–review, editing, and approval; and investigation (performed protein kinetic studies). G. Ciccio: Data acquisition and interpretation, writing–review, editing, and approval; and investigation (assistance in establishing the patient-derived xenograft [PDX] model). J.A. Copland III: Data acquisition and interpretation, writing–review, editing, and approval; and investigation (provided critical scientific oversight in establishing the patient xenograft model). J. Miller: Data acquisition and interpretation, writing–review, editing, and approval; and investigation (assistance in establishing the PDX model). B. Han: Data acquisition for seahorse experiments, functional hematopoiesis in lisaftoclax-treated MM and CLL patient analysis; writing–review, editing, and approval. L. Bai: Data acquisition and interpretation, writing–review, editing, and approval; and investigation (discovery chemistry, FP binding assay, and cell viability assays). L. Liu: Data acquisition and interpretation, writing–review, editing, and approval; and investigation (discovery chemistry, FP binding assay, and cell viability assays). M. Wang: Data acquisition and interpretation, writing–review, editing, and approval; and investigation (discovery chemistry, FP binding assay, and cell viability assays). D. McEachern: Data acquisition and interpretation, writing–review, editing, and approval; and investigation (discovery chemistry, FP binding assay, and cell viability assays). S. Przybranowski: Data acquisition and interpretation, writing–review, editing, and approval; and investigation (discovery chemistry, FP binding assay, and cell viability assays). C.Y. Yang: Data acquisition and interpretation, writing–review, editing, and approval; and investigation (discovery chemistry, FP binding assay, and cell viability assays). J.A. Stuckey: Data acquisition and interpretation, writing–review, editing, and approval; and investigation (discovery chemistry, FP binding assay, and cell viability assays). D. Wu: Data acquisition and interpretation, writing–review, editing, and approval; and investigation (provided the first set of chronic lymphocytic leukemia [CLL] ex vivo studies). C. Li: Data acquisition and interpretation, writing–review, editing, and approval; and investigation (provided the first set of chronic lymphocytic leukemia [CLL] ex vivo studies). J. Ryan: Data acquisition and interpretation, writing–review, editing, and approval; and investigation (provided unique BH3 profiling in a panel of cell lines demonstrating lisaftoclax as a true BH3 mimetic). A. Letai: Data acquisition and interpretation, writing–review, editing, and approval; and investigation (provided unique BH3 profiling in a panel of cell lines demonstrating lisaftoclax as a true BH3 mimetic). S. Ailawadhi: Data acquisition and interpretation, writing–review, editing, and approval; and investigation (led the MM and WM patient sample study study). D. Yang: Conceptualization and study design, data acquisition and interpretation, writing–review, editing, and approval. S. Wang: Conceptualization and study design, data acquisition and interpretation, writing–review, editing, and approval; and discovery investigation (performed discovery chemistry, fluorescence polarization [FP] binding assay, and cell viability assays). A. Chanan-Khan: Conceptualization and study design, data acquisition and interpretation, writing–review, editing, and approval; and investigation (led the MM and WM patient sample study study). Y. Zhai: Conceptualization and study design, data acquisition and interpretation, writing–review, editing, and approval.

We thank colleagues of Ascentage CMC and Analytic Center for providing lisaftoclax. The experiments and analyses carried out in the Mayo Clinic were supported in part by the Daniel Foundation of Alabama (to A. Chanan-Khan), the Predolin Foundation (to A. Chanan-Khan), the Mayo Clinic Cancer Center (CA015083, to A. Chanan-Khan), and the Mayo Clinic Multiple Myeloma SPORE (P50 CA186781–01A1, to A. Paulus). We thank Dr. Laura Lewis-Tuffin for her technical and scientific support for use of the Attune NxT Flow Cytometer and BD FACS Aria II cell sorter in the Mayo Florida Cellular Imaging and Flow Cytometry Shared Resource. Assistance in manuscript research and preparation was provided by Mr. Gutkin and Drs. Tao, Pathak, Ogba, and Fletcher (all with Ascentage). Preparation of this study report was informed by Animal Research: Reporting In Vivo Experiments (ARRIVE) guidelines for preclinical research (Kilkenny C, Browne WJ, Cuthill IC, Emerson M, Altman DG. Improving bioscience research reporting: the ARRIVE guidelines for reporting animal research. PLoS Biol 2010;8:e1000412). This study and its report were supported by Ascentage Pharma Group Corp Ltd.

The publication costs of this article were defrayed in part by the payment of publication fees. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

Note: Supplementary data for this article are available at Clinical Cancer Research Online (http://clincancerres.aacrjournals.org/).

1.
Bhola
PD
,
Letai
A
.
Mitochondria–judges and executioners of cell death sentences
.
Mol Cell
2016
;
61
:
695
704
.
2.
Roufayel
R
.
Regulation of stressed-induced cell death by the BCL-2 family of apoptotic proteins
.
Mol Membr Biol
2016
;
33
:
89
99
.
3.
Kalkavan
H
,
Green
DR
.
MOMP, cell suicide as a BCL-2 family business
.
Cell Death Differ
2018
;
25
:
46
55
.
4.
Delbridge
AR
,
Strasser
A
.
The BCL-2 protein family, BH3-mimetics, and cancer therapy
.
Cell Death Differ
2015
;
22
:
1071
80
.
5.
Del Gaizo Moore
V
,
Brown
JR
,
Certo
M
,
Love
TM
,
Novina
CD
,
Letai
A
.
Chronic lymphocytic leukemia requires BCL-2 to sequester prodeath BIM, explaining sensitivity to BCL-2 antagonist ABT-737
.
J Clin Invest
2007
;
117
:
112
21
.
6.
Deng
J
,
Carlson
N
,
Takeyama
K
,
Dal Cin
P
,
Shipp
M
,
Letai
A
.
BH3 profiling identifies three distinct classes of apoptotic blocks to predict response to ABT-737 and conventional chemotherapeutic agents
.
Cancer Cell
2007
;
12
:
171
85
.
7.
Pan
R
,
Hogdal
LJ
,
Benito
JM
,
Bucci
D
,
Han
L
,
Borthakur
G
, et al
.
Selective BCL-2 inhibition by ABT-199 causes on-target cell death in acute myeloid leukemia
.
Cancer Discov
2014
;
4
:
362
75
.
8.
Paulus
A
,
Akhtar
S
,
Yousaf
H
,
Manna
A
,
Paulus
SM
,
Bashir
Y
, et al
.
Waldenstrom macroglobulinemia cells devoid of BTKC481S or CXCR4WHIM-like mutations acquire resistance to ibrutinib through upregulation of Bcl-2 and AKT resulting in vulnerability towards venetoclax or MK2206 treatment
.
Blood Cancer J
2017
;
7
:
e565
.
9.
Touzeau
C
,
Dousset
C
,
Le Gouill
S
,
Sampath
D
,
Leverson
JD
,
Souers
AJ
, et al
.
The BCL-2–specific BH3 mimetic ABT-199: a promising targeted therapy for t(11;14) multiple myeloma
.
Leukemia
2014
;
28
:
210
2
.
10.
Oltersdorf
T
,
Elmore
SW
,
Shoemaker
AR
,
Armstrong
RC
,
Augeri
DJ
,
Belli
BA
, et al
.
An inhibitor of Bcl-2 family proteins induces regression of solid tumors
.
Nature
2005
;
435
;
677
81
.
11.
Tse
C
,
Shoemaker
AR
,
Adickes
J
,
Anderson
MG
,
Chen
J
,
Jin
S
, et al
.
ABT-263: a potent and orally bioavailable Bcl-2 family inhibitor
.
Cancer Res
2008
;
68
:
3421
28
.
12.
Wilson
WH
,
O'Connor
OA
,
Czuczman
MS
,
LaCasce
A
,
Gerecitano
JF
,
Leonard
JP
, et al
.
Navitoclax, a targeted high-affinity inhibitor of BCL-2, in lymphoid malignancies: a phase I dose-escalation study of safety, pharmacokinetics, pharmacodynamics, and antitumor activity
.
Lancet Oncol
2010
;
11
:
1149
59
.
13.
Souers
AJ
,
Leverson
JD
,
Boghaert
ER
,
Ackler
SL
,
Catron
ND
,
Chen
J
, et al
.
ABT-199, a potent and selective BCL-2 inhibitor, achieves antitumor activity while sparing platelets
.
Nat Med
2013
;
19
:
202
8
.
14.
Roberts
AW
,
Davids
MS
,
Pagel
JM
,
Kahl
BS
,
Puvvada
SD
,
Gerecitano
JF
, et al
.
Targeting BCL-2 with venetoclax in relapsed chronic lymphocytic leukemia
.
N Engl J Med
2016
;
374
:
311
22
.
15.
Roeker
LE
,
Fox
CP
,
Eyre
TA
,
Brander
DM
,
Allan
JN
,
Schuster
SJ
, et al
.
Tumor lysis, adverse events, and dose adjustments in 297 venetoclax-treated CLL patients in routine clinical practice
.
Clin Cancer Res
2019
;
25
:
4264
70
.
16.
Seymour
JF
,
Ma
S
,
Brander
DM
,
Choi
MY
,
Barrientos
J
,
Davids
MS
, et al
.
Venetoclax plus rituximab in relapsed or refractory chronic lymphocytic leukemia: a phase Ib study
.
Lancet Oncol
2017
;
18
:
230
40
.
17.
Davids
MS
,
von Keudell
G
,
Portell
CA
,
Cohen
JB
,
Fisher
DC
,
Foss
F
, et al
.
Revised dose ramp-up to mitigate the risk of tumor lysis syndrome when initiating venetoclax in patients with mantle cell lymphoma
.
J Clin Oncol
2018
;
JCO1800359
.
18.
Paulus
A
,
Akhtar
S
,
Caulfield
TR
,
Samuel
K
,
Yousaf
H
,
Bashir
Y
, et al
.
Coinhibition of the deubiquitinating enzymes, USP14 and UCHL5, with VLX1570 is lethal to ibrutinib- or bortezomib-resistant Waldenstrom macroglobulinemia tumor cells
.
Blood Cancer J
2016
;
6
:
e492
.
19.
Paulus
A
,
Manna
A
,
Akhtar
S
,
Paulus
SM
,
Sharma
M
,
Coignet
MV
, et al
.
Targeting CD38 with daratumumab is lethal to Waldenström macroglobulinaemia cells
.
Br J Haematol
2018
;
183
:
196
211
.
20.
Manna
A
,
Aulakh
S
,
Jani
P
,
Ahmed
S
,
Akhtar
S
,
Coignet
M
, et al
.
Targeting CD38 enhances the antileukemic activity of ibrutinib in chronic lymphocytic leukemia
.
Clin Cancer Res
2019
;
25
:
3974
85
.
21.
Chemical Computing Group
.
Molecular Operating Environment (MOE)
.
Available from
: https://www.chemcomp.com/.
22.
Jones
G
,
Willett
P
,
Glen
RC
,
Leach
AR
,
Taylor
R
.
Development and validation of a genetic algorithm for flexible docking
.
J Mol Biol
1997
;
267
:
727
48
.
23.
Verdonk
ML
,
Cole
JC
,
Hartshorn
MJ
,
Murray
CW
,
Taylor
RD
.
Improved protein-ligand docking using GOLD
.
Proteins
2003
;
52
:
609
23
.
24.
Wang
G
,
Nikolovska-Coleska
Z
,
Yang
C-Y
,
Wang
R
,
Tang
G
,
Guo
J
, et al
.
Structure-based design of potent small-molecule inhibitors of anti-apoptotic Bcl-2 proteins
.
J Med Chem
2006
;
49
:
6139
42
.
25.
Nikolovska-Coleska
Z
,
Wang
R
,
Fang
X
,
Pan
H
,
Tomita
Y
,
Li
P
, et al
.
Development and optimization of a binding assay for the XIAP BIR3 domain using fluorescence polarization
.
Anal Biochem
2004
;
332
:
261
73
.
26.
Huang
X
.
Fluorescence polarization competition assay: the range of resolvable inhibitor potency is limited by the affinity of the fluorescent ligand
.
J Biomol Screen
2003
;
8
:
34
8
.
27.
Lessene
G
,
Czabotar
PE
,
Sleebs
BE
,
Zobel
K
,
Lowes
KM
,
Adams
JM
, et al
.
Structure-guided design of a selective BCL-X(L) inhibitor
.
Nat Chem Biol
2013
;
9
:
390
7
.
28.
Wang
Q
,
Hao
S
.
A-1210477, a selective MCL-1 inhibitor, overcomes ABT-737 resistance in AML
.
Oncol Lett
2019
;
18
:
5481
9
.
29.
Koss
B
,
Ryan
J
,
Budhraja
A
,
Szarama
K
,
Yang
X
,
Bathina
M
, et al
.
Defining specificity and on-target activity of BH3-mimetics using engineered B-ALL cell lines
.
Oncotarget
2016
;
7
:
11500
11
.
30.
Fang
DD
,
Tang
Q
,
Kong
Y
,
Wang
Q
,
Gu
J
,
Fang
X
, et al
.
MDM2 inhibitor APG-115 synergizes with PD-1 blockade through enhancing antitumor immunity in the tumor microenvironment
.
J Immunother Cancer
2019
;
7
:
327
.
31.
Pan
R
,
Ruvolo
V
,
Mu
H
,
Leverson
JD
,
Nichols
G
,
Reed
JC
, et al
.
Synthetic lethality of combined Bcl-2 inhibition and p53 activation in AML: mechanisms and superior antileukemic efficacy
.
Cancer Cell
2017
;
32
:
748
60
.
32.
Certo
M
,
Del Gaizo Moore
V
,
Nishino
M
,
Wei
G
,
Korsmeyer
S
,
Armstrong
SA
, et al
.
Mitochondria primed by death signals determine cellular addiction to antiapoptotic BCL-2 family members
.
Cancer Cell
2006
;
9
:
351
65
.
33.
Ryan
J
,
Montero
J
,
Rocco
A
,
Letai
A
.
iBH3: simple, fixable BH3 profiling to determine apoptotic priming in primary tissue by flow cytometry
.
Biol Chem
2016
;
397
:
671
8
.
34.
Lee
JB
,
Khan
DH
,
Hurren
R
,
Xu
M
,
Na
Y
,
Kang
H
, et al
.
Venetoclax enhances T cell–mediated antileukemic activity by increasing ROS production
.
Blood
2021
;
138
:
234
45
.
35.
Kohlhapp
FJ
,
Hirabhai
D
,
Mathew
R
,
Duggan
R
,
Ellis
PA
,
Wang
R
, et al
.
Venetoclax increases intratumoral effector T cells and antitumor efficacy in combination with immune checkpoint blockade
.
Cancer Discov
2021
;
11
:
68
79
.

Supplementary data