Skip to main content
American Journal of Cancer Research logoLink to American Journal of Cancer Research
. 2023 Jan 15;13(1):307–325.

Synergistic activity of ABT-263 and ONC201/TIC10 against solid tumor cell lines is associated with suppression of anti-apoptotic Mcl-1, BAG3, pAkt, and upregulation of pro-apoptotic Noxa and Bax cleavage during apoptosis

Francesca R Di Cristofano 1,2,3,7, Mara W Fong 1,2,3,7, Kelsey E Huntington 1,2,3,4,7, Benedito A Carneiro 1,2,6,7, Lanlan Zhou 1,2,3,7, Wafik S El-Deiry 1,2,3,4,5,6,7
PMCID: PMC9906082  PMID: 36777502

Abstract

A major underlying cause of the resistance of solid tumor cells to cancer therapy is the evasion of cell death following anti-cancer drug treatment. We explored the combination of TRAIL-inducing compound ONC201/TIC10 and Bcl-xL/Bcl-2 inhibitor ABT-263 to target the extrinsic and intrinsic apoptotic pathways, respectively, in solid tumor cell lines (N = 13) derived from different tissues (colon, prostate, lung, breast, ovary, bladder). We found an IC50 range of 0.83-20.10 μM for ONC201 and 0.06-14.75 μM for ABT-263 among the 13 cancer cell lines. We show that combination of ONC201 and ABT-263 produces a strong synergistic effect leading to tumor cell death, and that the combination is not toxic to human fibroblast cells. In OVCAR-3 ovarian cancer cells, 2.5 μM ONC201 and 1.25 μM ABT-263 yielded 37% and 27% inhibition of viability, respectively, while the combination of the two agents yielded 92% inhibition of viability, resulting in a high synergy score of 52; conversely, the same combination in the HFF-1 human fibroblast cells yielded 2.45% inhibition of viability and a synergy score of 6.92 (synergy scores were calculated using SynergyFinder; scores greater than 10 are considered synergistic). We also found that the combination of these two agents resulted in synergistic caspase activation and PARP cleavage consistent with induction of apoptosis. Combination therapy-induced cell death correlated with decreased levels of Mcl-1, BAG3, pAkt, and upregulation of Noxa along with Bax cleavage during apoptosis at 48 hours, and ATF4, TRAIL, and DR5 induction at 24 hours. There was some heterogeneity in the cell lines with regard to these responses. Our data provide evidence for synergy from the combination of ONC201 and ABT-263 against human solid tumor cell lines associated with alterations in cell death and pro-survival mediators. The combination of ONC201 and ABT-263 merits further exploration in vivo and in clinical trials against a variety of solid malignancies.

Keywords: Apoptosis, targeted therapy, drug synergy, ONC201, ABT-263, Mcl-1, BAG3, Akt, Noxa

Introduction

Despite remarkable progress over the last two decades in the development of targeted cancer therapy, immunotherapy, and precision medicine, according to the ACS there will be over 600,000 deaths from cancer in the US in 2022. While it is a goal of the field of oncology to prevent or diagnose cancer early, the presentation of cancer at a late stage continues to be associated with a poor prognosis. A general feature of advanced cancer that contributes to this poor prognosis is resistance to therapy that may be intrinsic or acquired. As such, there is an urgent need for the development of novel therapeutic approaches that can address the complex nature of cancer, including tumor heterogeneity, evolution, and resistance to anti-cancer therapy.

A major underlying cause of the resistance of tumors cells to cancer therapy is the evasion of cell death following anti-cancer drug treatment [1]. Cell death resistance is a hallmark of cancer and is characterized by various phenotypes including upregulation of anti-apoptotic proteins, increased activation of pro-survival factors, and downregulation of expression or activity of pro-apoptotic mediators [2]. A great deal of progress has been made in the discovery and early development of cancer therapeutics that target cell death through the intrinsic or extrinsic cell death pathways.

ABT-263 is a small molecule that targets the intrinsic apoptotic pathway. It induces tumor cell death by directly binding to Bcl-xL and Bcl-2 and blocking their anti-apoptotic function. The ABT-263 targeting of Bcl-xL has been found to cause an on-target dose-dependent thrombocytopenia in vivo and in the clinic [3-5]. Consequently, ABT-199 was developed to specifically target Bcl-2 [6]. Although ABT-199’s selectivity for Bcl-2 results in greater patient tolerance and has led to its approval by the FDA for the treatment of chronic lymphocytic leukemia (CLL) [7] and acute myeloid leukemia (AML) [8], it is ineffective in cell lines that preferentially express Bcl-xL over Bcl-2 [9]. Thus, despite the limitations of ABT-263, tumor heterogeneity keeps it of interest to the field.

ONC201/TIC10 is a first-in-class small molecule that targets the extrinsic apoptotic pathway. It was originally discovered as a TRAIL-inducing compound from a cell-based screen of an NCI library [10,11]. Subsequent studies demonstrated that ONC201 not only upregulates the cytotoxic ligand TRAIL but also its pro-apoptotic receptor DR5, and showed activity mediated by upregulation of the integrated stress response [12]. ONC201 has been found to target cancer stem cells [13,14] and to upregulate an immune response involving natural killer cells which act in part through TRAIL production as part of the host innate immune response against cancer [15]. More recently, ONC201 and its analogues ONC206 and ONC212 have been found to bind to mitochondrial caseinolytic protease ClpP to inhibit oxidative phosphorylation as part of the mechanism of its anti-cancer effect [10,16]. This mitochondria-mediated cell death by ONC201 may involve the suppression of Mcl-1, an anti-apoptotic Bcl-2 family protein [17].

A major mechanism of resistance to ABT-263 involves the upregulation of anti-apoptotic Mcl-1 in ABT-263-treated solid tumors [18]. Moreover, overexpression of Bcl-2 has been found to limit the efficacy of ONC201 [17]. Given that ONC201 has been found to decrease Mcl-1 expression and that Bcl-2 is a target of ABT-263, the combination of ONC201 and ABT-263 should in theory have great synergistic potential. Synergistic combination therapies between ABT-263 and other anti-cancer molecules may allow for the use of lower concentrations of ABT-263 in the clinic, suppressing the dose-dependent thrombocytopenia and allowing for greater clinical success. The combination of ONC201 and ABT-263 has been tested in glioblastoma [19] and the combination of ONC201 and ABT-199 has been tested in hematological malignancies [17]. However, ABT-263 is not able to cross the blood-brain barrier [20], and the combination of ONC201 and ABT-263 has not been tested in other solid tumors. Thus, there remains a gap in the field to test this combination therapy in tissue types to which ONC201 and ABT-263 are bioavailable.

We investigated the activity of ONC201 and ABT-263 against solid tumor cell lines as single agents and in combination. We identified potent synergy between the two cancer therapeutic agents against a broad range of solid tumor cell lines and provided experimental evidence for alterations in cell death mediators as potential explanations for the observed preclinical synergy. Our results support further preclinical and clinical studies of this drug combination in cancer therapy.

Methods

Reagents

ABT-263 was purchased from MedKoo Biosciences (Morrisville, NC, USA) and was solubilized in DMSO at a storage concentration of 20 mM. ONC201 was supplied by Oncoceutics/Chimerix, Inc (Philadelphia, PA, USA) and was reconstituted in DMSO at a storage concentration of 20 mM. Both reagents were stored at -20°C.

Cell cultures and growth conditions

All cell lines but one were obtained from the American Type Culture Collection (ATCC). The IGROV-1 cell line was obtained from the Fox Chase Cancer Center Cell Culture Facility. All cell lines were confirmed to be mycoplasma-free using PCR testing methods. HT29 and HCT116 cells were cultured in McCoy’s 5A modified medium supplemented with 10% fetal bovine serum and 1% Penicillin/Streptomycin. SW480, MDA-MB-231, MDA-MB-436, H1048, and SKOV-3 cells were cultured in DMEM medium supplemented with 10% fetal bovine serum and 1% Penicillin/Streptomycin. HFF-1 cells were cultured in DMEM supplemented with 15% fetal bovine serum and 1% Penicillin/Streptomycin. PC3, 22RV1, OVCAR-3, and IGROV-1 cells were cultured in RPMI medium supplemented with 10% fetal bovine serum and 1% Penicillin/Streptomycin. UM-UC-3 and J82 cells were cultured in Eagle’s Minimum Essential Medium supplemented with 10% fetal bovine serum and 1% Penicillin/Streptomycin. All cell cultures were stored in an incubator at 37°C within a 95% humidified atmosphere containing 5% carbon.

Cell viability assays

Cells were plated in opaque-walled 96-well plates at a density of 3000 cells per well and incubated overnight in 100 μL culture medium. After 24 hrs, single-agent ONC201 and ABT-263 or combined ONC201 and ABT-263 were added and treatment was continued for 72 hrs. 20 μL of CellTiterGlo bioluminescence agent (Promega Corporation, Madison, WI) was then added into each well and the contents were mixed for 2 min on a plate shaker to induce cell lysis. Cell viability was determined using the CellTiterGlo assay. Single-drug dose response curves were created using GraphPad Prism. Synergy scores were calculated using SynergyFinder.

Colony formation assays

With variations between cell lines, cells were plated at a density of 200-500 cells per well of a 12-well plate and allowed to grow overnight. After 24 hrs, the cells were treated with 1.25 μM ONC201, 5 μM ABT-263, or the combination of the two reagents. After an additional six days of incubation, wells were washed with phosphate-buffered saline (PBS), fixed by methanol for 20 min, and stained by Coomassie Brilliant Blue for 10 min. Colonies were imaged and counted using ImageJ.

Western blot analysis

Cells were plated in triplicate at a density of 5 × 105-1 × 106 cells per well of a 6-well plate and allowed to grow over night. Cells were treated with single-agent ONC201 and ABT-263 or the combination. After 24 or 48 hrs of treatment, the cells were washed with PBS and lysed using protein lysis buffer. Proteins were quantified using the Pierce BCA Protein Assay (ThermoFisher Scientific, Waltham, MA). Cell lysates were electrophoresed through 4-12% SDS-PAGE then transferred to PVDF membranes. The PVDF membrane was blocked using 5% non-fat milk. The membranes were incubated overnight at 4°C with primary antibodies diluted in 5% non-fat milk according to the suggested datasheet dilutions. Invitrogen Goat anti-Rabbit IgG (H+L) Secondary Antibody and HRP #31460 and Goat anti-Mouse IgG (H+L) Secondary Antibody, HRP #31430 were diluted 1:10,000 in 5% non-fat milk. Each membrane was probed with a loading control once and stripped and reprobed for additional proteins no more than one time.

Immunofluorescence analysis

Cells were plated in triplicate at a density of 30 k cells per well of an 8-chamber slide and treated with single-agent ONC201 and ABT-263 or the combination. After 24 or 48 hrs, the cells were washed with PBS, fixed with 4% paraformaldehyde, permeabilized with 0.1% Triton X-100, and blocked with 2% BSA. The cells were incubated with ATF4 primary antibody diluted in 0.1% BSA overnight at 4°C, after which they were washed with PBS and incubated with Alexa Fluor 488 donkey anti-rabbit secondary antibodies for 45 min. The cells were washed with PBS and mounted using a mounting medium containing DAPI stain. The coverslips were sealed and the plates were imaged using a fluorescent microscope.

Statistical analysis

Cell viability assay and colony formation assay data are all represented as the mean ± SD. Single-drug dose response curves were generated by nonlinear regression analyses using GraphPad Prism. The treatment groups for the colony formation assay were compared using ordinary one-way ANOVA tests; P values < 0.05 were considered to be statistically significant. HSA model synergy scores for the two-drug viability assays were determined using SynergyFinder (Network Pharmacology for Precision Medicine in the Research Program of System Oncology, Faculty of Medicine at University of Helsinki, Helsinki, Finland); synergy scores < -10 indicate antagonism, between -10 and 10 indicate an additive effect, and > 10 indicate synergism. Single-drug viability assays were conducted in replicates of six; two-drug viability assays, colony formation assays, and western blot analyses were conducted in replicates of three.

Results

TRAIL pathway inducer ONC201/TIC10 and Bcl-xL/Bcl-2 inhibitor ABT-263 show cytotoxicity towards solid tumor cell lines (N = 13) derived from different tissues (colon, prostate, lung, breast, ovary, bladder)

To investigate the activity of cell death-targeting therapeutics against solid tumor cell lines, we treated a panel of human cancer cell lines (N = 13) with varying doses of TRAIL pathway inducer ONC201/TIC10 and Bcl-xL/Bcl-2 inhibitor ABT-263 (Figure 1 and Table 1). Dose response relationships are shown for ONC201 (Figure 1A) and ABT-263 (Figure 1B). Loss of viability was observed at a range of drug concentrations in the panel of cell lines. IC50 values along with tissue of origin and known genomic driver gene alterations are listed in Table 1.

Figure 1.

Figure 1

Concentration-dependent effect of ONC201 and ABT-263 on cell viability. 13 solid tumor cell lines were treated with DMSO or varying concentrations of either ONC201 (A) and ABT-263 (B) for 72 hrs. Cell viability was then determined using the CellTiter-Glo Luminescent Cell Viability Assay.

Table 1.

Tissues of origin, mutational profiles, IC50 concentrations of selected solid tumor cell lines

Cell Line Tissue Mutational Profile ABT-263 IC50 ONC201 IC50

KRAS BRAF PIK3CA PTEN TP53
HT29 Colon wt V600E P449T wt R273H 6.19 μM 5.07 μM
HCT116 Colon ascendens G13D wt H1047R wt wt 4.24 μM 2.54 μM
SW480 Colon G12V wt wt wt R273H, P309S 0.43 μM 3.14 μM
PC3 Prostate wt wt wt Null Null 4.39 μM 20.10 μM
22RV1 Prostate wt L597R Q546R wt Q331R 6.91 μM 0.83 μM
H1048 Lung wt wt H1047R wt R273C 0.06 μM 1.89 μM
MDA-MB-231 Breast G13D G464V wt wt R280K 0.43 μM 2.96 μM
MDA-MB-436 Breast wt wt wt wt E204fs 14.75 μM 3.94 μM
SKOV-3 Ovary wt wt H1047R wt Null 13.04 μM 2.28 μM
IGROV-1 Ovary wt wt R38C Y155C, T319* Y126C, S90Lfs*59 1.95 μM 5.55 μM
OVCAR-3 Ovary wt wt wt wt R248Q 1.20 μM 1.98 μM
UM-UC-3 Urinary tract; bladder G12C wt wt Null F113C 5.46 μM 5.86 μM
J82 Urinary tract; bladder wt wt P124L Null E271K, K320N 2.64 μM 2.45 μM

(*) denotes a mutation that results in the creation of a translation stop codon.

Following treatment with ONC201, the most sensitive cell lines within the 13-cell line panel were 22RV1 prostate cancer cells (ONC201 IC50 = 0.83 μM), H1048 small cell lung cancer (ONC201 IC50 = 1.89 μM), and OVCAR-3 high grade serous ovarian carcinoma cells (ONC201 IC50 = 1.98 μM). The cell lines most sensitive to ABT-263 were H1048 small cell lung cancer (ABT-263 IC50 = 0.06 μM), SW480 colon cancer cells (ABT-263 IC50 = 0.43 μM), and MDA-MB-231 triple negative breast cancer cells (ABT-263 IC50 = 0.43 μM).

Combination of ONC201 and ABT-263 produces strong synergy with loss of cell viability in solid tumor cell lines

Having observed single agent cytotoxicity with monotherapy, we explored whether the combination of ONC201 and ABT-263 produced synergistic effects. We performed cell viability assays with the solid tumor cell lines using increasing concentrations of both drugs in combination. The results shown in Figure 2 and Table 2 reveal combinatorial interactions of the drugs suggesting strong synergy, with scores greater than 10 found in all cell lines across the 13-cell line panel.

Figure 2.

Figure 2

ONC201 and ABT-263 synergize to reduce cell viability in solid tumor cell lines. HT29 cells (A), HCT116 cells (B), MDA-MB-231 cells (C), OVCAR-3 cells (D), IGROV-1 cells (E), and SKOV-3 cells (F) were treated for 72 hrs. with the indicated concentrations of ONC201 and ABT-263. Viability is visualized (top) on a rainbow scale using the CellTiter-Glo reagent as described in the methods, and inhibition is indicated numerically (middle). HSA synergy scores are shown (bottom) for the combination of ONC201 and ABT-263 at various non-constant concentration ratios. Scores < -10 are considered antagonistic, scores between -10 and 10 are considered additive, and scores > 10 are considered synergistic. Combination of ONC201 and ABT-263 resulted in high synergy scores across all cell lines.

Table 2.

Percent inhibition and top synergy scores for cells treated with ONC201 and ABT-263

Cell Line ONC201 ABT-263 Combination



% inhibition [] % inhibition [] % inhibition Score
HT29 45.93±4.55 5 μM 33.32±2.08 5 μM 80.18±1.65 35.41
HCT116 20.13±9.66 1.25 μM 24.27±9.20 2.5 μM 58.00±1.74 30.16
SW480 46.71±3.49 2.5 μM 39.46±3.94 0.31 μM 69.92±1.15 23.15
PC3 47.18±3.38 2.5 μM 13.19±1.6 2.5 μM 71.23±2.06 24.47
22RV1 20.38±8.12 1.25 μM 31.03±4.72 5 μM 50.87±5.19 19.88
H1048 19.09±1.14 1.25 μM 28.90±2.75 0.02 μM 55.05±2.45 25.01
MDA-MB-231 61.65±9.19 10 μM 64.43±1.66 1.25 μM 93.59±1.96 27.14
MDA-MB-436 50.04±4.43 5 μM 29.84±8.44 10 μM 71.26±3.69 25.42
SKOV-3 40.83±4.69 5 μM 33.72±4.59 10 μM 89.43±1.38 48.29
IGROV-1 58.09±4.44 5 μM 37.29±8.29 5 μM 90.89±0.71 33.92
OVCAR-3 36.57±4.64 2.5 μM 26.52±13.4 1.25 μM 91.80±1.16 52.23
UM-UC-3 55.09±13.23 2.5 μM 30.20±23.94 5 μM 70.44±14.77 13.56
J82 59.47±10.19 10 μM 24.99±13.2 10 μM 86.12±5.21 23.87

The cell lines with the highest synergy scores were the OVCAR-3 ovarian cancer cells, the SKOV-3 ovarian cancer cells, and the HT29 colon cancer cells. The synergy score for the OVCAR-3 cell line was 52.23 at 2.5 μM ONC201 and 1.25 μM ABT263, with a two-drug percent inhibition of 91.80% as compared to the single-drug percent inhibition of 36.57% for ONC201 and 26.52% for ABT-263. The synergy score for the SKOV-3 cells was 48.29 at 5 μM ONC201 and 10 μM ABT-263, with a two-drug percent inhibition of 89.43% as compared to 40.83% and 33.72% inhibition with ONC201 and ABT-263, respectively. Finally, the synergy score for the HT29 cells was 35.41 at 5 μM ONC201 and 5 μM ABT-263, with a two-drug percent inhibition of 80.18% as compared to 45.93% and 33.32% inhibition with ONC201 and ABT-263, respectively.

ONC201 and ABT-263 are not toxic to human fibroblast cell lines

Having observed high levels of synergy across the panel of solid tumor cell lines, we sought to determine the sensitivity of normal fibroblast cells to ONC201 and ABT-263 as single agents and in combination. We performed cell viability assays using HFF-1 human foreskin fibroblast cells.

As seen in Figure 3A and 3B, in comparison to the solid tumor cell lines, the HFF-1 cells were relatively insensitive to ONC201 and ABT-263 as single agents: even at the highest concentration tested for each drug (10 μM), IC50 was not reached. As shown in Figure 3C, when the two drugs were combined, sensitivity remained low, with a maximum inhibition of 67% at the highest concentrations of ONC201 and ABT-263. Although scores indicating synergy were seen at some drug concentration combinations, the great majority of combinations yielded no synergy (scores between -10 and 10) or antagonism (scores less than -10). More specifically, dose combinations of ONC201 and ABT-263 that yielded high synergy scores in solid tumor cell lines yielded no synergy or antagonism in the fibroblast cell line: 2.5 μM ONC201 combined with 2.5 μM ABT-263 synergy scores of 23.7, 49.7, and 2.44 in the HCT116, OVCAR-3, and HFF-1 cells, respectively.

Figure 3.

Figure 3

Sensitivity to ONC201 and ABT-263 as single agents and in combination is low in human fibroblast (normal) cells. HFF-1 cells were treated with varying concentrations of ONC201 as a single agent (A), ABT-263 as a single agent (B), or ONC201 and ABT-263 in combination (C) for 72 hrs. Viability is visualized on a rainbow scale using the CellTiter-Glo reagent as described in the methods. HSA synergy scores are shown (C) for the combination of ONC201 and ABT-263 at various non-constant concentration ratios. Combination of ONC201 and ABT-263 resulted in negative to moderate synergy scores across all cell lines.

Combination of ONC201 and ABT-263 synergistically suppresses colony formation of solid tumor cell lines

Having observed synergistic cytotoxicity across the 13-cell line panel in short-term cell viability assays, we investigated whether similar phenotypes of sensitivity and synergy might be observed when using long-term colon formation assays.

Our examples shown in Figure 4 and Table 3 suggest that the synergistic effects of ONC201 plus ABT-263 extend to longer-term (7 days) colony formation assays, further supporting the observation that the two drugs have potent anti-cancer effects when combined in the treatment of solid tumor cells. The cell lines that showed the greatest reduction in colony forming ability for combined ONC201 and ABT-263 treatment as compared to single agent treatment were the 22RV1 prostate cancer cells and the HCT116 colon cancer cells, with ordinary one-way ANOVA tests showing P values of < 0.0001 between the ONC201 and ABT-263 combination group and all other treatment conditions (Figure 4).

Figure 4.

Figure 4

ONC201 and ABT-263 combined reduce colony forming ability of solid tumor cell lines. 22RV1 (A) and HCT116 (B) cells were treated with DMSO, 1.25 μM ONC201, 5 μM ABT-263, or 1.25 μM ONC201 + 5 μM ABT-263 24 hrs. after plating and allowed to grow for six days, after which colonies were stained using Coomassie Brilliant Blue. Images of the colonies are shown (left). Colonies were quantified and expressed as mean ± SD, and ordinary one-way ANOVA tests were used to compare the treatment groups (right). Combination of ONC201 and ABT-263 is shown to significantly reduce the colony forming ability of solid tumor cell lines.

Table 3.

Mean colony number ± SD and results of ordinary one-way ANOVA test for cells treated with ONC201 and ABT-263

Cell Line Control ONC201 ABT-263 Combo Combo to Control Combo to ONC201 Combo to ABT-263
HT29 149±1.528 131±7.550 109±22.81 46±1.528 P < 0.0001 P = 0.0001 P = 0.0009
HCT116 135±8.718 111±5.686 78±7.000 27±3.000 P < 0.0001 P < 0.0001 P < 0.0001
SW480 149±10.44 122±12.17 60±5.859 16±2.082 P < 0.0001 P < 0.0001 P = 0.0012
PC3 139±7.937 116±8.505 97±4.163 53±5.568 P < 0.0001 P < 0.0001 P = 0.0002

ONC201 and ABT-263 enhance caspase cleavage and apoptosis

Having found high levels of synergy across all cell lines, we sought to explore whether apoptosis was induced following treatment with ONC201 and ABT-263. We performed western blot analyses for several markers of apoptosis, including cleaved PARP, caspase 8, caspase 9, and cleaved caspase 3 in three colorectal and two ovarian cancer cell lines. As shown in Figures 5 and 6, caspase activation and PARP cleavage consistent with induction of apoptosis was found following treatment of solid tumor cell lines by ONC201 and ABT-263 both as single agents and in combination. In most instances, when comparing the combined agent groups to the single agent and control groups, enhanced PARP, caspase 8, caspase 9, and caspase 3 cleavage were found. These findings are consistent with the high levels of synergism found across the 13-cell line panel.

Figure 5.

Figure 5

Synergy between ONC201 and ABT-263 is associated with PARP cleavage and caspase activation, decreased BAG3 and Mcl-1, Noxa induction, and BAX cleavage. The expression of various pro- and anti-apoptotic proteins was detected by western blot analysis for the HT29, IGROV-1, and SW480 cell lines. Panels are organized with apoptosis markers in the left column, intrinsic pathway proteins in the middle column, and extrinsic pathway proteins in the right column. A. HT29 cells were treated with DMSO, 5 μM ONC201, 5 μM ABT-263, or the combination for 48 hrs. B. IGROV-1 cells were treated with DMSO, 5 μM ONC201, 1.25 μM ABT-263, or the combination for 48 hrs. C. SW480 cells were treated with DMSO, 5 μM ONC201, 0.625 μM ABT-263, or the combination for 48 hrs. Elevated levels of PARP cleavage, caspase 3/8/9 activation, NOXA expression, and BAX cleavage, as well as reduced levels of Mcl-1, BAG3, and pAkt were observed in comparison to the control and single drug groups across the three solid tumor cell lines. For each cell line, all membranes were produced from a single set of lysates. Each membrane was probed with a loading control once and stripped and reprobed for additional proteins no more than one time. Some loading controls appear twice as they were used for different protein categories. For example, for the HT29 cells, one membrane was first probed for Noxa and Actin and then stripped and reprobed for ERK and pERK, and thus the loading control image is repeated in the middle and right columns. Similarly: for the HT29 cells, one actin was used as a loading control for cPARP, CP8, CP9, cCP3, Mcl-1, BAG3, BAX, BID, PUMA; for the IGROV-1 cells, one actin was used as a loading control for cPARP, CP8, CP9, cCP3, Mcl-1, BAG3, BAX, BID, PUMA and another was used for Noxa, ERK, pERK; for the SW480 cells, one actin was used as a loading control for Noxa, ERK, pERK.

Figure 6.

Figure 6

Combined treatment with ONC201 and ABT-263 is associated with changes in anti-apoptotic and pro-apoptotic protein expression at 48 hrs. The expression of various pro- and anti-apoptotic proteins was detected by western blot analysis for the HCT116 and SKOV-3 cell lines. Panels are organized with apoptosis markers in the left column, intrinsic pathway proteins in the middle column, and extrinsic pathway proteins in the right column. (A) HCT116 cells were treated with DMSO, 2.5 μM ONC201, 2.5 μM ABT-263, or the combination for 48 hrs. (B) SKOV-3 cells were treated with DMSO, 2.5 μM ONC201, 2.5 μM ABT-263, or the combination for 48 hrs. Elevated levels of PARP cleavage, caspase activation, and BAX cleavage were observed in both cell lines. BAG3 reduction and NOXA induction were observed in the HCT116 cells (A). Mcl-1 and pAkt reduction was observed in SKOV-3 cells (B). For each cell line, all membranes were produced from a single set of lysates. Each membrane was probed with a loading control once and stripped and reprobed for additional proteins no more than one time. Some loading controls appear twice as they were used for different protein categories. For example, for the HCT-116 cells, one membrane was first probed for Bcl-XL, Noxa, BIM, and Actin and then stripped and reprobed for ERK and pERK, and thus the loading control image is repeated in the middle and right columns. Similarly: for the HCT-116 cells, one actin was used as a loading control for cPARP, CP8, CP9, cCP3, Mcl-1, BAG3, BAX, BID, PUMA; for the SKOV-3 cells, one actin was used as a loading control for cPARP, CP8, CP9, cCP3, Mcl-1, BAX while another was used for BAG3, Noxa, TRAIL, DR5, Akt, pAkt.

Decreased Mcl-1, BAG3, pAkt, and upregulation of Noxa and Bax cleavage during apoptosis of cancer cell lines following 48-hour treatment with ONC201 and ABT-263

We further investigated the molecular basis for the synergism found between ONC201 and ABT-263. We hypothesized that strong synergy between the two drugs that target the apoptotic pathways might be associated with alterations in relevant anti-apoptotic and pro-apoptotic proteins. To test this hypothesis, we performed western blot analyses across five solid tumor cell lines (Figures 5 and 6).

Elevated levels of Mcl-1 are a well-known mechanism of resistance to ABT-263. ONC201 has previously been found to reduce expression of Mcl-1, providing a basis for the combination of the two agents. Indeed, ABT-263 was found to maintain or increase Mcl-1 expression in all tested cell lines, whereas ONC201 was shown to reduce Mcl-1 expression in the HT29, IGROV-1, HCT116, and SKOV-3 cell lines. Combined treatment of ONC201 and ABT-263 at 48 hours was shown in maintain this decrease (HT29, IGROV-1, SKOV-3) or even further reduce (SW480) Mcl-1 expression in comparison to single agent ONC201 treatment. To further explore the mechanism by which Mcl-1 expression is reduced, we decided to look at the expression of BAG3, a protein known to stabilize Mcl-1, following 48-hour treatment. In the HT29, SW480, and HCT116 cell lines, ONC201 and ABT-263 as single agents were found to maintain or decrease BAG3 expression. When ONC201 and ABT-263 were combined in these three cell lines, an enhanced reduction in BAG3 expression was found as compared to the single agent groups.

pAkt is known to inhibit FOXO3a, preventing its translocation to the nucleus and the transcriptional activation of the TRAIL gene. ONC201 is known to cause downstream inhibition of pAkt; as such, we explored the expression of pAkt. In the HT29, IGROV-1, SW480, and SKOV-3 cells, 48-hour combined treatment with ONC201 and ABT-263 was found to greatly reduce pAkt expression as compared to the control and single agent groups.

Noxa is a pro-apoptotic BH3-only protein. In the HT29, IGROV-1, SW480, and HCT116 cell lines, ABT-263 both alone and in combination with ONC201 was found to induce Noxa expression. Furthermore, in the HT29, IGROV-1, and SW480 cell lines, the combination of ONC201 and ABT-263 was found to induce Noxa expression at higher levels than with single agent ABT-263. Finally, enhanced Bax cleavage was found in HT29, IGROV-1, SW480, HCT116, and SKOV-3 cells treated with ONC201 in combination with ABT-263 as compared to the single agent groups.

Early increase in ATF4, TRAIL and DR5 expression following 24-hour treatment with ONC201 and ABT-263

The expression patterns for the panel of extrinsic pathway proteins at the 48-hour timepoint were rather unexpected, with paradoxical TRAIL suppression and inconsistent DR5 and ATF4 expression (Figures 5 and 6). However, it is known that when it comes to complex pathways that involve many molecular interactions, transcriptional level expression changes, and several cellular compartments, protein expression is timepoint sensitive. Moreover, at later time points, cells that experience early upregulation of pro-apoptotic proteins experience cell death and are consequently eliminated. As the extrinsic pathway is essential to ONC201 function, we decided to investigate the expression of these key proteins at various timepoints via western blot and immunostaining analysis. We selected two cell lines from the previous five-cell line panel: IGROV-1 and SW480. Results are shown in Figure 7.

Figure 7.

Figure 7

Combined treatment with ONC201 with ABT-263 is associated with induction of key extrinsic pathway proteins at 24 hrs, followed by suppression at 48 hrs. The expression of various extrinsic pathway proteins was detected by western blot and immunofluorescence analysis for the SW480 and IGROV-1 cell lines. A, C, D. IGROV-1 cells were treated with DMSO, 5 μM ONC201, 1.25 μM ABT-263, or the combination for 24 or 48 hrs. B, E, F. SW480 cells were treated with DMSO, 5 μM ONC201, 0.625 μM ABT-263, or the combination for 24 or 48 hrs. An induction of these proteins at 24 hrs was followed by their downregulation at 48 hrs. C, E. Quantification of mean anti-ATF4 fluorescence intensity relative to whole cell staining with DAPI at 24 and 48 hrs. D, F. Images showing ATF4 expression for cells treated with ONC201 and ABT-263 in combination for 24 or 48 hrs.

ONC201 induction of ATF4 is thought to be upstream of an increase in DR5, while its effects on Foxo3a increase TRAIL expression. Indeed, following 24-hour treatment with ONC201, an increase in ATF4, TRAIL, and DR5 expression was seen (Figure 7A and 7B). In the IGROV1 cells, combined treatment with ONC201 and ABT-263 appeared to enhance this induction, providing further evidence of synergy. Interestingly, at 48 hours, the expression of these three proteins appeared to drop in the ONC201 and ONC201+ABT-263-treated cells. This resulted in an expression pattern consistent with that seen in Figures 5 and 6.

We further examined ATF4 expression via immunostaining at 24 and 48 hours in the IGROV-1 and SW480 cells. As shown in Figure 7C and 7E, when looking at the ratio between ATF4 fluorescence and total cell fluorescence, there was a drop at 48 hours that was specific to the combined agent group, consistent with the above findings. These results are supported visually in Figure 7D and 7F.

Discussion

The treatment of solid tumors remains a great challenge in the field of cancer therapeutics. While surgery is a first-line treatment for most solid tumors, late-stage diagnosis often eliminates it as a viable option. On the other hand, later-stage treatments such as chemotherapeutics or targeted therapies are often limited by tumor cell resistance. One mechanism behind this resistance is tumor cell evasion of programmed cell death through the upregulation of anti-apoptotic molecules and the downregulation of pro-apoptotic molecules. With this, targeting apoptotic pathways has become a focus of the field.

The dual induction of the extrinsic and intrinsic apoptotic pathways has long been recognized as a potentially powerful therapeutic approach. The discovery of ONC201/TIC10 [21] has facilitated the development of such a therapy. Unlike previous inducers of the extrinsic apoptotic pathway, which have been associated with unimpressive patient outcomes [22,23], issues related to pharmacokinetics [24], and inherent or acquired resistance [25], ONC201 induces TRAIL at the transcriptional level and has shown enhanced tumor control and greater promise in the clinic [10,11]. Consequently, ONC201 has been combined with BH3-mimetics in glioblastoma [19] and leukemia [17]. However, BH3-mimetics do not cross the blood brain barrier, and solid tumors are more genetically complex than hematological malignancies.

In this study, we tested the combination of ONC201 and ABT-263 against a panel of 13 solid tumor cell lines derived from different tissues, including colon, prostate, lung, breast, ovary, and bladder. This presents the first time ONC201 and ABT-263 have been tested in solid tissues to which they are both bioavailable. We showed that this dual targeting of the extrinsic and intrinsic apoptotic pathways produced a potent anti-tumor effect and high levels of synergy across all tested cell lines. We demonstrated preclinical pro-apoptotic anti-cancer phenotypes by short term cell viability assays, long-term colony assays, and western blot analysis of apoptotic cell markers such as PARP cleavage and caspase 3/8/9 activation.

Our investigation of the underlying mechanisms of synergy revealed drug effects on relevant pro- and anti-apoptotic mediators as well as cell survival signaling. We showed novel ABT-263-mediated NOXA induction and Bax cleavage, both of which were enhanced by the addition of ONC201. Combination therapy-induced cell death was additionally associated with decreased levels of anti-apoptotic Mcl-1, BAG3, and pAkt at 48 hours and earlier induction of pro-apoptotic ATF4, TRAIL, and DR5 at 24 hours. These effects were over and above individual drug effects that may be important for each of the drugs when used alone or in other therapeutic combinations.

ABT-263 selectively inhibits the anti-apoptotic proteins Bcl-2 and Bcl-xL, leaving Mcl-1 free to block pro-apoptotic proteins. It is well-known that innate resistance to treatment with ABT-263 is often caused by the intrinsically enhanced expression of Mcl-1, and that ABT-263 itself paradoxically induces the expression of Mcl-1, leading to acquired resistance [26,27]. Indeed, combination therapies between ABT-263 and Mcl-1 suppressors are being explored to circumvent this resistance mechanism [28-31]. Our results show that the IGROV-1 and SW480 cell lines express elevated levels of Mcl-1 in the ABT-263-treated cells as compared to the control group, suggesting that even within 72 hours of treatment, mechanisms of acquired resistance are induced. In accordance with previous findings, we show that ONC201 leads to decreased expression of Mcl-1. Moreover, we show for the first time that Noxa was induced by ABT-263 both as a single agent and in combination with ONC201. Noxa is a pro-apoptotic protein that directly inhibits Mcl-1 [32,33], and it has been shown that base Noxa expression is a critical determinant of sensitivity to ABT-263 and its analogs [34,35]. Taken together, these data suggest that ONC201 suppression of Mcl-1 along with ABT-263-mediated inhibition of Mcl-1 via Noxa induction are key components of the mechanism of synergy of ONC201 and ABT-263 combination. Furthermore, we found that BAG3, a chaperone protein known to stabilize Mcl-1 [36-38], was downregulated in an ONC201-dependent manner. BAG3 has previously been suggested as a target for anti-cancer therapy [39], and initial work has been done to develop an anti-BAG3 antibody for the treatment of pancreatic cancer [40]. Our findings support the growing relevance of BAG3 as a significant player in cancer progression.

Interestingly, Bax cleavage was among the most consistent findings in the western blot analysis, with all five tested cell lines showing a lower molecular weight band corresponding to cleaved Bax for the combination treatment groups. Bax is a critical component of the intrinsic mitochondrial apoptotic pathway, as its activation initiates mitochondrial outer membrane permeabilization (MOMP) and the release of key intermembrane space proteins that lead to caspase activation and cell death. A previous study [41] has shown that during stress-induced apoptosis, Bax is cleaved to p18 Bax. p18 Bax is more potent than full-length Bax in its ability to disrupt mitochondrial integrity, and thus its formation leads to accelerated and enhanced apoptosis.

It must be acknowledged that the expression level of some apoptotic proteins was heterogeneous. Further exploration revealed that the variability of extrinsic pathway proteins was likely due to timing-while at 24 hours ONC201 both alone and in combination led to a potent induction of ATF4, TRAIL, and DR5 expression, there was a dramatic decrease in the levels of these proteins at 48 hours. Nevertheless, the question of whether heterogeneity between patients could hinder the success of the ONC201 and ABT-263 combination remains important. Thus, the biological relevance of these findings remains unclear and should be subject to further exploration.

A limitation of ABT-263 therapies is that the drug has been found to induce thrombocytopenia in clinical trials. It is believed that the targeting of Bcl-xL on circulating platelet cells is the cause of this toxicity [42]. Consequently, ABT-199 was developed as a specific inhibitor of Bcl-2. Although ABT-199 is effective in Bcl-2-addicted cells, many cells either do not express Bcl-2 or are addicted to other pro-apoptotic proteins, such as Bcl-xL [9,43]. Thus, despite its limitations in the clinic, ABT-263 continues to be of interest in pre-clinical studies. Because toxicity to ABT-263 is dose-dependent and caused by effects on non-cancerous cells, the future success of ABT-263 in the clinic is dependent on two factors: first, synergistic combinations between ABT-263 and other compounds that allow for the use of lower concentrations of ABT-263; second, specificity of this synergism to cancerous cells. Here we show that the combination of ONC201 and ABT-263 induces high levels of synergy and cell death at relatively low concentrations of each drug in all tested solid tumor cell lines. Moreover, we show that synergy of this significance is not reproduced in HFF-1 fibroblast cell line, with the majority of ONC201 and ABT-263 concentration combinations yielding no synergy or antagonistic effects. Although this points to specificity and the potential evasion of toxicity in the clinic, further exploration of this drug combination in vivo is needed.

Overall, our data provide evidence for synergy from the combination of ONC201 and ABT-263 against human solid tumor cell lines associated with alterations in cell death and pro-survival mediators. The combination of ONC201 and ABT-263 merits further exploration in vivo and in clinical trials against a variety of solid malignancies.

Acknowledgements

W.S.E-D. is an American Cancer Society Research Professor and is supported by the Mencoff Family University Professorship at Brown University. This work was supported by an NIH grant (CA173453) to W.S.E-D. The contents of this manuscript are solely the responsibility of the authors and do not necessarily represent the official views of the National Cancer Institute, the National Institutes of Health, or the American Cancer Society.

Disclosure of conflict of interest

W.S.E-D. is a co-founder of Oncoceutics, Inc., a subsidiary of Chimerix. Dr. El-Deiry has disclosed his relationship with Oncoceutics/Chimerix and potential conflict of interest to his academic institution/employer and is fully compliant with NIH and institutional policy that is managing this potential conflict of interest.

References

  • 1.Carneiro BA, El-Deiry WS. Targeting apoptosis in cancer therapy. Nat Rev Clin Oncol. 2020;17:395–417. doi: 10.1038/s41571-020-0341-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144:646–674. doi: 10.1016/j.cell.2011.02.013. [DOI] [PubMed] [Google Scholar]
  • 3.Tse C, Shoemaker AR, Adickes J, Anderson MG, Chen J, Jin S, Johnson EF, Marsh KC, Mitten MJ, Nimmer P, Roberts L, Tahir SK, Xiao Y, Yang X, Zhang H, Fesik S, Rosenberg SH, Elmore SW. ABT-263: a potent and orally bioavailable Bcl-2 family inhibitor. Cancer Res. 2008;68:3421–3428. doi: 10.1158/0008-5472.CAN-07-5836. [DOI] [PubMed] [Google Scholar]
  • 4.Gandhi L, Camidge DR, Ribeiro de Oliveira M, Bonomi P, Gandara D, Khaira D, Hann CL, McKeegan EM, Litvinovich E, Hemken PM, Dive C, Enschede SH, Nolan C, Chiu YL, Busman T, Xiong H, Krivoshik AP, Humerickhouse R, Shapiro GI, Rudin CM. Phase I study of Navitoclax (ABT-263), a novel Bcl-2 family inhibitor, in patients with small-cell lung cancer and other solid tumors. J. Clin. Oncol. 2011;29:909–916. doi: 10.1200/JCO.2010.31.6208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Rudin CM, Hann CL, Garon EB, Ribeiro de Oliveira M, Bonomi PD, Camidge DR, Chu Q, Giaccone G, Khaira D, Ramalingam SS, Ranson MR, Dive C, McKeegan EM, Chyla BJ, Dowell BL, Chakravartty A, Nolan CE, Rudersdorf N, Busman TA, Mabry MH, Krivoshik AP, Humerickhouse RA, Shapiro GI, Gandhi L. Phase II study of single-agent navitoclax (ABT-263) and biomarker correlates in patients with relapsed small cell lung cancer. Clin Cancer Res. 2012;18:3163–3169. doi: 10.1158/1078-0432.CCR-11-3090. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Souers AJ, Leverson JD, Boghaert ER, Ackler SL, Catron ND, Chen J, Dayton BD, Ding H, Enschede SH, Fairbrother WJ, Huang DC, Hymowitz SG, Jin S, Khaw SL, Kovar PJ, Lam LT, Lee J, Maecker HL, Marsh KC, Mason KD, Mitten MJ, Nimmer PM, Oleksijew A, Park CH, Park CM, Phillips DC, Roberts AW, Sampath D, Seymour JF, Smith ML, Sullivan GM, Tahir SK, Tse C, Wendt MD, Xiao Y, Xue JC, Zhang H, Humerickhouse RA, Rosenberg SH, Elmore SW. ABT-199, a potent and selective BCL-2 inhibitor, achieves antitumor activity while sparing platelets. Nat Med. 2013;19:202–208. doi: 10.1038/nm.3048. [DOI] [PubMed] [Google Scholar]
  • 7.Roberts AW, Davids MS, Pagel JM, Kahl BS, Puvvada SD, Gerecitano JF, Kipps TJ, Anderson MA, Brown JR, Gressick L, Wong S, Dunbar M, Zhu M, Desai MB, Cerri E, Heitner Enschede S, Humerickhouse RA, Wierda WG, Seymour JF. Targeting BCL2 with venetoclax in relapsed chronic lymphocytic leukemia. N Engl J Med. 2016;374:311–322. doi: 10.1056/NEJMoa1513257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Konopleva M, Pollyea DA, Potluri J, Chyla B, Hogdal L, Busman T, McKeegan E, Salem AH, Zhu M, Ricker JL, Blum W, DiNardo CD, Kadia T, Dunbar M, Kirby R, Falotico N, Leverson J, Humerickhouse R, Mabry M, Stone R, Kantarjian H, Letai A. Efficacy and biological correlates of response in a phase II study of venetoclax monotherapy in patients with acute myelogenous leukemia. Cancer Discov. 2016;6:1106–1117. doi: 10.1158/2159-8290.CD-16-0313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Wang Q, Wan J, Zhang W, Hao S. MCL-1 or BCL-xL-dependent resistance to the BCL-2 antagonist (ABT-199) can be overcome by specific inhibitor as single agents and in combination with ABT-199 in acute myeloid leukemia cells. Leuk Lymphoma. 2019;60:2170–2180. doi: 10.1080/10428194.2018.1563694. [DOI] [PubMed] [Google Scholar]
  • 10.Prabhu VV, Morrow S, Rahman Kawakibi A, Zhou L, Ralff M, Ray J, Jhaveri A, Ferrarini I, Lee Y, Parker C, Zhang Y, Borsuk R, Chang WI, Honeyman JN, Tavora F, Carneiro B, Raufi A, Huntington K, Carlsen L, Louie A, Safran H, Seyhan AA, Tarapore RS, Schalop L, Stogniew M, Allen JE, Oster W, El-Deiry WS. ONC201 and imipridones: anti-cancer compounds with clinical efficacy. Neoplasia. 2020;22:725–744. doi: 10.1016/j.neo.2020.09.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Allen JE, Krigsfeld G, Mayes PA, Patel L, Dicker DT, Patel AS, Dolloff NG, Messaris E, Scata KA, Wang W, Zhou JY, Wu GS, El-Deiry WS. Dual inactivation of Akt and ERK by TIC10 signals Foxo3a nuclear translocation, TRAIL gene induction, and potent antitumor effects. Sci Transl Med. 2013;5:171ra17. doi: 10.1126/scitranslmed.3004828. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Kline CL, Van den Heuvel AP, Allen JE, Prabhu VV, Dicker DT, El-Deiry WS. ONC201 kills solid tumor cells by triggering an integrated stress response dependent on ATF4 activation by specific eIF2alpha kinases. Sci Signal. 2016;9:ra18. doi: 10.1126/scisignal.aac4374. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Prabhu VV, Allen JE, Dicker DT, El-Deiry WS. Small-molecule ONC201/TIC10 targets chemotherapy-resistant colorectal cancer stem-like cells in an Akt/Foxo3a/TRAIL-dependent manner. Cancer Res. 2015;75:1423–1432. doi: 10.1158/0008-5472.CAN-13-3451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Prabhu VV, Lulla AR, Madhukar NS, Ralff MD, Zhao D, Kline CLB, Van den Heuvel APJ, Lev A, Garnett MJ, McDermott U, Benes CH, Batchelor TT, Chi AS, Elemento O, Allen JE, El-Deiry WS. Cancer stem cell-related gene expression as a potential biomarker of response for first-in-class imipridone ONC201 in solid tumors. PLoS One. 2017;12:e0180541. doi: 10.1371/journal.pone.0180541. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Wagner J, Kline CL, Zhou L, Campbell KS, MacFarlane AW, Olszanski AJ, Cai KQ, Hensley HH, Ross EA, Ralff MD, Zloza A, Chesson CB, Newman JH, Kaufman H, Bertino J, Stein M, El-Deiry WS. Dose intensification of TRAIL-inducing ONC201 inhibits metastasis and promotes intratumoral NK cell recruitment. J Clin Invest. 2018;128:2325–2338. doi: 10.1172/JCI96711. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Ferrarini I, Louie A, Zhou L, El-Deiry WS. ONC212 is a novel mitocan acting synergistically with glycolysis inhibition in pancreatic cancer. Mol Cancer Ther. 2021;20:1572–1583. doi: 10.1158/1535-7163.MCT-20-0962. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Ishizawa J, Kojima K, Chachad D, Ruvolo P, Ruvolo V, Jacamo RO, Borthakur G, Mu H, Zeng Z, Tabe Y, Allen JE, Wang Z, Ma W, Lee HC, Orlowski R, Sarbassov dos D, Lorenzi PL, Huang X, Neelapu SS, McDonnell T, Miranda RN, Wang M, Kantarjian H, Konopleva M, Davis RE, Andreeff M. ATF4 induction through an atypical integrated stress response to ONC201 triggers p53-independent apoptosis in hematological malignancies. Sci Signal. 2016;9:ra17. doi: 10.1126/scisignal.aac4380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Anstee NS, Bilardi RA, Ng AP, Xu Z, Robati M, Vandenberg CJ, Cory S. Impact of elevated anti-apoptotic MCL-1 and BCL-2 on the development and treatment of MLL-AF9 AML in mice. Cell Death Differ. 2019;26:1316–1331. doi: 10.1038/s41418-018-0209-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Karpel-Massler G, Ba M, Shu C, Halatsch ME, Westhoff MA, Bruce JN, Canoll P, Siegelin MD. TIC10/ONC201 synergizes with Bcl-2/Bcl-xL inhibition in glioblastoma by suppression of Mcl-1 and its binding partners in vitro and in vivo. Oncotarget. 2015;6:36456–36471. doi: 10.18632/oncotarget.5505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Yamaguchi R, Perkins G. Finding a Panacea among combination cancer therapies. Cancer Res. 2012;72:18–23. doi: 10.1158/0008-5472.CAN-11-3091. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Allen JE, Kline CL, Prabhu VV, Wagner J, Ishizawa J, Madhukar N, Lev A, Baumeister M, Zhou L, Lulla A, Stogniew M, Schalop L, Benes C, Kaufman HL, Pottorf RS, Nallaganchu BR, Olson GL, Al-Mulla F, Duvic M, Wu GS, Dicker DT, Talekar MK, Lim B, Elemento O, Oster W, Bertino J, Flaherty K, Wang ML, Borthakur G, Andreeff M, Stein M, El-Deiry WS. Discovery and clinical introduction of first-in-class imipridone ONC201. Oncotarget. 2016;7:74380–74392. doi: 10.18632/oncotarget.11814. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Soria JC, Mark Z, Zatloukal P, Szima B, Albert I, Juhasz E, Pujol JL, Kozielski J, Baker N, Smethurst D, Hei YJ, Ashkenazi A, Stern H, Amler L, Pan Y, Blackhall F. Randomized phase II study of dulanermin in combination with paclitaxel, carboplatin, and bevacizumab in advanced non-small-cell lung cancer. J. Clin. Oncol. 2011;29:4442–4451. doi: 10.1200/JCO.2011.37.2623. [DOI] [PubMed] [Google Scholar]
  • 23.Hotte SJ, Hirte HW, Chen EX, Siu LL, Le LH, Corey A, Iacobucci A, MacLean M, Lo L, Fox NL, Oza AM. A phase 1 study of mapatumumab (fully human monoclonal antibody to TRAIL-R1) in patients with advanced solid malignancies. Clin Cancer Res. 2008;14:3450–3455. doi: 10.1158/1078-0432.CCR-07-1416. [DOI] [PubMed] [Google Scholar]
  • 24.Yuan X, Gajan A, Chu Q, Xiong H, Wu K, Wu GS. Developing TRAIL/TRAIL death receptor-based cancer therapies. Cancer Metastasis Rev. 2018;37:733–748. doi: 10.1007/s10555-018-9728-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Azijli K, Weyhenmeyer B, Peters GJ, de Jong S, Kruyt FA. Non-canonical kinase signaling by the death ligand TRAIL in cancer cells: discord in the death receptor family. Cell Death Differ. 2013;20:858–868. doi: 10.1038/cdd.2013.28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Goodwin CM, Rossanese OW, Olejniczak ET, Fesik SW. Myeloid cell leukemia-1 is an important apoptotic survival factor in triple-negative breast cancer. Cell Death Differ. 2015;22:2098–2106. doi: 10.1038/cdd.2015.73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Konopleva M, Contractor R, Tsao T, Samudio I, Ruvolo PP, Kitada S, Deng X, Zhai D, Shi YX, Sneed T, Verhaegen M, Soengas M, Ruvolo VR, McQueen T, Schober WD, Watt JC, Jiffar T, Ling X, Marini FC, Harris D, Dietrich M, Estrov Z, McCubrey J, May WS, Reed JC, Andreeff M. Mechanisms of apoptosis sensitivity and resistance to the BH3 mimetic ABT-737 in acute myeloid leukemia. Cancer Cell. 2006;10:375–388. doi: 10.1016/j.ccr.2006.10.006. [DOI] [PubMed] [Google Scholar]
  • 28.Lee A, Jin HO, Masudul Haque M, Kim HY, Jung H, Park JH, Kim I, Song JY, Yoon HK, Kim HK, Han J, Park IC, Kim KS, Park SG. Synergism of a novel MCL1 downregulator, acriflavine, with navitoclax (ABT263) in triplenegative breast cancer, lung adenocarcinoma and glioblastoma multiforme. Int J Oncol. 2022;60:2. doi: 10.3892/ijo.2021.5292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Dastur A, Choi A, Costa C, Yin X, Williams A, McClanaghan J, Greenberg M, Roderick J, Patel NU, Boisvert J, McDermott U, Garnett MJ, Almenara J, Grant S, Rizzo K, Engelman JA, Kelliher M, Faber AC, Benes CH. NOTCH1 represses MCL-1 levels in GSI-resistant T-ALL, making them susceptible to ABT-263. Clin Cancer Res. 2019;25:312–324. doi: 10.1158/1078-0432.CCR-18-0867. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Pan R, Ruvolo VR, Wei J, Konopleva M, Reed JC, Pellecchia M, Andreeff M, Ruvolo PP. Inhibition of Mcl-1 with the pan-Bcl-2 family inhibitor (-)BI97D6 overcomes ABT-737 resistance in acute myeloid leukemia. Blood. 2015;126:363–372. doi: 10.1182/blood-2014-10-604975. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Kour S, Rana S, Contreras JI, King HM, Robb CM, Sonawane YA, Bendjennat M, Crawford AJ, Barger CJ, Kizhake S, Luo X, Hollingsworth MA, Natarajan A. CDK5 inhibitor downregulates Mcl-1 and sensitizes pancreatic cancer cell lines to navitoclax. Mol Pharmacol. 2019;96:419–429. doi: 10.1124/mol.119.116855. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Pang X, Zhang J, Lopez H, Wang Y, Li W, O’Neill KL, Evans JJ, George NM, Long J, Chen Y, Luo X. The carboxyl-terminal tail of Noxa protein regulates the stability of Noxa and Mcl-1. J Biol Chem. 2014;289:17802–17811. doi: 10.1074/jbc.M114.548172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Ouchi K, Kuwahara Y, Iehara T, Miyachi M, Katsumi Y, Tsuchiya K, Konishi E, Yanagisawa A, Hosoi H. A NOXA/MCL-1 imbalance underlies chemoresistance of malignant rhabdoid tumor cells. J Cell Physiol. 2016;231:1932–1940. doi: 10.1002/jcp.25293. [DOI] [PubMed] [Google Scholar]
  • 34.Nakajima W, Hicks MA, Tanaka N, Krystal GW, Harada H. Noxa determines localization and stability of MCL-1 and consequently ABT-737 sensitivity in small cell lung cancer. Cell Death Dis. 2014;5:e1052. doi: 10.1038/cddis.2014.6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Tromp JM, Geest CR, Breij EC, Elias JA, van Laar J, Luijks DM, Kater AP, Beaumont T, van Oers MH, Eldering E. Tipping the Noxa/Mcl-1 balance overcomes ABT-737 resistance in chronic lymphocytic leukemia. Clin Cancer Res. 2012;18:487–498. doi: 10.1158/1078-0432.CCR-11-1440. [DOI] [PubMed] [Google Scholar]
  • 36.Boiani M, Daniel C, Liu X, Hogarty MD, Marnett LJ. The stress protein BAG3 stabilizes Mcl-1 protein and promotes survival of cancer cells and resistance to antagonist ABT-737. J Biol Chem. 2013;288:6980–6990. doi: 10.1074/jbc.M112.414177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Habata S, Iwasaki M, Sugio A, Suzuki M, Tamate M, Satohisa S, Tanaka R, Saito T. BAG3-mediated Mcl-1 stabilization contributes to drug resistance via interaction with USP9X in ovarian cancer. Int J Oncol. 2016;49:402–410. doi: 10.3892/ijo.2016.3494. [DOI] [PubMed] [Google Scholar]
  • 38.Mani J, Antonietti P, Rakel S, Blaheta R, Bartsch G, Haferkamp A, Kogel D. Knockdown of BAG3 sensitizes bladder cancer cells to treatment with the BH3 mimetic ABT-737. World J Urol. 2016;34:197–205. doi: 10.1007/s00345-015-1616-2. [DOI] [PubMed] [Google Scholar]
  • 39.Kirk JA, Cheung JY, Feldman AM. Therapeutic targeting of BAG3: considering its complexity in cancer and heart disease. J Clin Invest. 2021;131:e149415. doi: 10.1172/JCI149415. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Basile A, De Marco M, Festa M, Falco A, Iorio V, Guerriero L, Eletto D, Rea D, Arra C, Lamolinara A, Ballerini P, Damiani V, Rosati A, Sala G, Turco MC, Marzullo L, De Laurenzi V. Development of an anti-BAG3 humanized antibody for treatment of pancreatic cancer. Mol Oncol. 2019;13:1388–1399. doi: 10.1002/1878-0261.12492. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Cao X, Deng X, May WS. Cleavage of Bax to p18 Bax accelerates stress-induced apoptosis, and a cathepsin-like protease may rapidly degrade p18 Bax. Blood. 2003;102:2605–2614. doi: 10.1182/blood-2003-01-0211. [DOI] [PubMed] [Google Scholar]
  • 42.Mohamad Anuar NN, Nor Hisam NS, Liew SL, Ugusman A. Clinical review: navitoclax as a pro-apoptotic and anti-fibrotic agent. Front Pharmacol. 2020;11:564108. doi: 10.3389/fphar.2020.564108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Wang Y, Wang Y, Fan X, Song J, Wu H, Han J, Lu L, Weng X, Nie G. ABT-199-mediated inhibition of Bcl-2 as a potential therapeutic strategy for nasopharyngeal carcinoma. Biochem Biophys Res Commun. 2018;503:1214–1220. doi: 10.1016/j.bbrc.2018.07.027. [DOI] [PubMed] [Google Scholar]

Articles from American Journal of Cancer Research are provided here courtesy of e-Century Publishing Corporation

RESOURCES