inhibitionofbcl2familymembersincreasestheefficacy · cu chelator tetrathiomolybdate (ttm) is...

15
CANCER RESEARCH | METABOLISM AND CHEMICAL BIOLOGY Inhibition of BCL2 Family Members Increases the Efcacy of Copper Chelation in BRAF V600E -Driven Melanoma A C Ye-Jin Kim 1 , Tiffany Tsang 2 , Grace R. Anderson 3 , Jessica M. Posimo 1 , and Donita C. Brady 1,4 ABSTRACT The principal unmet need in BRAF V600E -positive melanoma is lack of an adequate therapeutic strategy capable of overcoming resistance to clinically approved targeted therapies against oncogenic BRAF and/or the downstream MEK1/2 kinases. We previously discovered that copper (Cu) is required for MEK1 and MEK2 activity through a direct CuMEK1/2 interaction. Repurposing the clinical Cu chelator tetrathiomolybdate (TTM) is supported by efcacy in BRAF V600E -driven melanoma models, due in part to inhibition of MEK1/2 kinase activity. However, the antineoplastic activity of Cu chelators is cytostatic. Here, we performed high-throughput small- molecule screens to identify bioactive compounds that synergize with TTM in BRAF V600E -driven melanoma cells. Genetic perturba- tion or pharmacologic inhibition of specic members of the BCL2 family of antiapoptotic proteins (BCL-W, BCL-XL, and MCL1) selectively reduced cell viability when combined with a Cu chelator and induced CASPASE-dependent cell death. Further, in BRAF V600E -positive melanoma cells evolved to be resistant to BRAF and/or MEK1/2 inhibitors, combined treatment with TTM and the clinically evaluated BCL2 inhibitor, ABT-263, restored tumor growth suppression and induced apoptosis. These ndings further support Cu chelation as a therapeutic strategy to target oncogene- dependent tumor cell growth and survival by enhancing Cu chelator efcacy with chemical inducers of apoptosis, especially in the context of refractory or relapsed BRAF V600E -driven melanoma. Signicance: This study unveils a novel collateral drug sensitivity elicited by combining copper chelators and BH3 mimetics for treatment of BRAF V600E mutation-positive melanoma. Introduction Melanoma is driven in 40% to 50% of cases by activating mutations in the BRAF serine/threonine kinases (1, 2). Over 90% of oncogenic BRAF mutations detected in melanoma are Val 600!Glu (V600E; refs. 3, 4). Activated BRAF V600E phosphorylates and activates MEK1/2, which subsequently phosphorylate and activate ERK1/2, resulting in hyperac- tivation of the evolutionarily conserved MAPK pathway to drive mel- anomagenesis (4, 5). Thus, patients with late-stage BRAF V600E -positive melanoma are typically treated with the FDA-approved combination of mutant-selective, ATP-competitive BRAF inhibitors (BRAFi, dabrafenib, and vemurafenib) and allosteric MEK1/2 inhibitors (MEK1/2i, trame- tinib, and cobimetinib; refs. 69). Although this standard of care is initially effective, patients with BRAF V600E -mutant melanoma have only modest improvements in median progression-free survival and eventu- ally develop resistance (4, 10, 11). The limited clinical durability of the combination has bolstered research aimed at additional combination strategies to forestall resistance development, targeting multiple signaling pathways capable of driving resistance, or exploring alternative pharma- cologic accessible nodes within the MAPK pathway (4, 10, 11). In search of identifying novel components of the canonical MAPK pathway, several groups have employed functional genomics approaches (12). Specically, a whole-genome RNAi screen revealed that the primary copper (Cu) transporter Ctr1 reduced ERK1/2 phosphorylation when knocked down in Drosophila S2 cells (13). We demonstrated that Cu directly binds to MEK1/2 and inuences the strength of the RAF-MEK-ERK cascade (14). Leveraging the depen- dence of BRAF mutationpositive cancers on MEK1/2 for tumori- genesis (15), we found that decreasing the levels of CTR1 or intro- ducing surface accessible mutations in MEK1 that disrupt Cu binding decreased BRAF V600E -driven signaling and tumor growth (16). Impor- tantly, the Cu-selective chelator tetrathiomolybdate (TTM), used as an investigational treatment of Wilson disease (17), diminished tumor- igenesis in models of BRAF V600E melanoma (18). Although TTM use has not been clinically explored in BRAF V600E -driven melanoma, TTM has been assessed in breast cancers as an antiangiogenic compound where patients have been treated safely for upwards of 65 months (19). Further, the combination of TTM, a well-tolerated and affordable drug, and vemurafenib led to a survival benet in a murine model of metastatic melanoma, but failed to yield tumor regression (18). In this study, we aimed to advance the therapeutic value of Cu chelation in BRAF V600E melanomas by identifying compounds that enhanced TTM efcacy. We performed high-throughput small- molecule screens with a panel of bioactive compounds to explore collateral drug sensitivities in combination with TTM. Here, we dem- onstrate that cotargeting select BCL2 proteins via BH3 mimetics syner- gizes with Cu chelators in both na ve and resistant forms of BRAF V600E melanoma cells. The ndings presented here highlight the potential of inducing apoptosis and melanoma tumor suppression when Cu chela- tors are combined with BCL2is. Materials and Methods Reagents A1210477 (ApexBio, B6011), ABT-199 (Selleck Chemicals, S8048), ABT-263 (M1637, AbMole), ABT-737 (Selleck Chemicals, S1002), 1 Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania. 2 Cell and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania. 3 Department of Pharmacology and Cancer Biology, Duke Uni- versity Medical Center, Durham, North Carolina. 4 Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania. Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/). Corresponding Author: Donita C. Brady, Department of Cancer Biology, Uni- versity of Pennsylvania, 421 Curie Blvd., 612 BRBII/II, Philadelphia, PA 19104. Phone: 215-573-9705; E-mail: [email protected] Cancer Res 2020;80:1387400 doi: 10.1158/0008-5472.CAN-19-1784 Ó2020 American Association for Cancer Research. AACRJournals.org | 1387 on March 1, 2021. © 2020 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from Published OnlineFirst January 31, 2020; DOI: 10.1158/0008-5472.CAN-19-1784

Upload: others

Post on 09-Oct-2020

3 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: InhibitionofBCL2FamilyMembersIncreasestheEfficacy · Cu chelator tetrathiomolybdate (TTM) is supported by efficacy in BRAFV600E-driven melanoma models, due in part to inhibition

CANCER RESEARCH | METABOLISM AND CHEMICAL BIOLOGY

Inhibition of BCL2 FamilyMembers Increases the Efficacyof Copper Chelation in BRAFV600E-Driven Melanoma A C

Ye-Jin Kim1, Tiffany Tsang2, Grace R. Anderson3, Jessica M. Posimo1, and Donita C. Brady1,4

ABSTRACT◥

The principal unmet need in BRAFV600E-positive melanoma islack of an adequate therapeutic strategy capable of overcomingresistance to clinically approved targeted therapies against oncogenicBRAF and/or the downstream MEK1/2 kinases. We previouslydiscovered that copper (Cu) is required forMEK1 andMEK2 activitythrough a direct Cu–MEK1/2 interaction. Repurposing the clinicalCu chelator tetrathiomolybdate (TTM) is supported by efficacy inBRAFV600E-driven melanoma models, due in part to inhibition ofMEK1/2 kinase activity. However, the antineoplastic activity of Cuchelators is cytostatic. Here, we performed high-throughput small-molecule screens to identify bioactive compounds that synergizewith TTM in BRAFV600E-driven melanoma cells. Genetic perturba-tion or pharmacologic inhibition of specific members of the BCL2family of antiapoptotic proteins (BCL-W, BCL-XL, and MCL1)

selectively reduced cell viability when combined with a Cuchelator and induced CASPASE-dependent cell death. Further, inBRAFV600E-positive melanoma cells evolved to be resistant to BRAFand/or MEK1/2 inhibitors, combined treatment with TTM andthe clinically evaluated BCL2 inhibitor, ABT-263, restored tumorgrowth suppression and induced apoptosis. These findings furthersupport Cu chelation as a therapeutic strategy to target oncogene-dependent tumor cell growth and survival by enhancing Cu chelatorefficacy with chemical inducers of apoptosis, especially in the contextof refractory or relapsed BRAFV600E-driven melanoma.

Significance: This study unveils a novel collateral drug sensitivityelicited by combining copper chelators and BH3 mimetics fortreatment of BRAFV600E mutation-positive melanoma.

IntroductionMelanoma is driven in 40% to 50% of cases by activatingmutations in

the BRAF serine/threonine kinases (1, 2). Over 90% of oncogenic BRAFmutations detected in melanoma are Val 600!Glu (V600E; refs. 3, 4).Activated BRAFV600E phosphorylates and activates MEK1/2, whichsubsequently phosphorylate and activate ERK1/2, resulting in hyperac-tivation of the evolutionarily conserved MAPK pathway to drive mel-anomagenesis (4, 5). Thus, patients with late-stage BRAFV600E-positivemelanoma are typically treated with the FDA-approved combination ofmutant-selective, ATP-competitive BRAF inhibitors (BRAFi, dabrafenib,and vemurafenib) and allosteric MEK1/2 inhibitors (MEK1/2i, trame-tinib, and cobimetinib; refs. 6–9). Although this standard of care isinitially effective, patients with BRAFV600E-mutant melanoma have onlymodest improvements in median progression-free survival and eventu-ally develop resistance (4, 10, 11). The limited clinical durability of thecombination has bolstered research aimed at additional combinationstrategies to forestall resistance development, targetingmultiple signalingpathways capable of driving resistance, or exploring alternative pharma-cologic accessible nodes within the MAPK pathway (4, 10, 11).

In search of identifying novel components of the canonical MAPKpathway, several groups have employed functional genomicsapproaches (12). Specifically, a whole-genome RNAi screen revealedthat the primary copper (Cu) transporter Ctr1 reduced ERK1/2phosphorylation when knocked down in Drosophila S2 cells (13). Wedemonstrated that Cu directly binds to MEK1/2 and influences thestrength of the RAF-MEK-ERK cascade (14). Leveraging the depen-dence of BRAF mutation–positive cancers on MEK1/2 for tumori-genesis (15), we found that decreasing the levels of CTR1 or intro-ducing surface accessible mutations in MEK1 that disrupt Cu bindingdecreased BRAFV600E-driven signaling and tumor growth (16). Impor-tantly, the Cu-selective chelator tetrathiomolybdate (TTM), used as aninvestigational treatment of Wilson disease (17), diminished tumor-igenesis in models of BRAFV600E melanoma (18). Although TTM usehas not been clinically explored in BRAFV600E-drivenmelanoma, TTMhas been assessed in breast cancers as an antiangiogenic compoundwhere patients have been treated safely for upwards of 65months (19).Further, the combination of TTM, a well-tolerated and affordabledrug, and vemurafenib led to a survival benefit in a murine model ofmetastatic melanoma, but failed to yield tumor regression (18).

In this study, we aimed to advance the therapeutic value of Cuchelation in BRAFV600E melanomas by identifying compounds thatenhanced TTM efficacy. We performed high-throughput small-molecule screens with a panel of bioactive compounds to explorecollateral drug sensitivities in combination with TTM. Here, we dem-onstrate that cotargeting select BCL2 proteins via BH3 mimetics syner-gizes with Cu chelators in both na€�ve and resistant forms of BRAFV600E

melanoma cells. The findings presented here highlight the potential ofinducing apoptosis and melanoma tumor suppression when Cu chela-tors are combined with BCL2is.

Materials and MethodsReagents

A1210477 (ApexBio, B6011), ABT-199 (Selleck Chemicals, S8048),ABT-263 (M1637, AbMole), ABT-737 (Selleck Chemicals, S1002),

1Department of Cancer Biology, Perelman School of Medicine, University ofPennsylvania, Philadelphia, Pennsylvania. 2Cell and Molecular Biology GraduateGroup, Perelman School of Medicine, University of Pennsylvania, Philadelphia,Pennsylvania. 3Department of Pharmacology and Cancer Biology, Duke Uni-versity Medical Center, Durham, North Carolina. 4Abramson Family CancerResearch Institute, Perelman School of Medicine, University of Pennsylvania,Philadelphia, Pennsylvania.

Note: Supplementary data for this article are available at Cancer ResearchOnline (http://cancerres.aacrjournals.org/).

Corresponding Author: Donita C. Brady, Department of Cancer Biology, Uni-versity of Pennsylvania, 421 Curie Blvd., 612 BRBII/II, Philadelphia, PA 19104.Phone: 215-573-9705; E-mail: [email protected]

Cancer Res 2020;80:1387–400

doi: 10.1158/0008-5472.CAN-19-1784

�2020 American Association for Cancer Research.

AACRJournals.org | 1387

on March 1, 2021. © 2020 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 31, 2020; DOI: 10.1158/0008-5472.CAN-19-1784

Page 2: InhibitionofBCL2FamilyMembersIncreasestheEfficacy · Cu chelator tetrathiomolybdate (TTM) is supported by efficacy in BRAFV600E-driven melanoma models, due in part to inhibition

Ammonium TTM (Sigma-Aldrich, 323446), trametinib (SelleckChemicals, S2673), WEHI-539 hydrochloride (ApexBio, A8634),vemurafenib (CT-P4032), and Z-DEVD-FMK (Selleck Chemicals,S7312) were purchased from indicated companies.

Cell lines293T/17 (ATCC, catalog #CRL-11268), A375 (ATCC, catalog

#CRL-1619), WM88 (Rockland, catalog #WM88-01-0001),WM3311 (Rockland, catalog #WM3311-01-0001), and WM3743(Rockland, catalog #WM3743-01-0001) cells were purchased fromthe indicated companies and maintained in DMEM (Gibco) sup-plemented with 10% v/v FBS (GE Lifesciences) and 1% penicillin–streptomycin (Gibco). 451Lu parental cells and resistant derivatives,451-Lu BRAFiR and 451-Lu MEKiR, were a kind gift from JessieVillanueva (Wistar Institute, Philadelphia, PA) and maintained inDMEM supplemented with 5 % FBS with 1 mmol/L vemurafenib or1 mmol/L trametinib (20, 21). WM983B parental cells and aresistant derivative, WM983B BRAFiR, were a kind gift from JessieVillanueva (Wistar Institute) and maintained in DMEM supple-mented with 5 % FBS with 1 mmol/L vemurafenib. Cell lines werenot authenticated. Mycoalert testing was done to test for myco-plasma contamination of all cell lines. Derived cell lines weregenerated by stable infection with lentiviruses derived from thepSMARTvector-inducible lentiviral shRNA plasmids (Dharmacon,see plasmids below) or retroviruses derived from the pBABEretroviral plasmid (see plasmids below) described below usingestablished methods.

PlasmidspSMARTvector-inducible lentiviral shRNA plasmids were pur-

chased from Dharmacon to express: nontargeted control, humanBCL2 target sequence #1 50-TGACGCTCTCCACACACAT, humanBCL2 target sequence #2 50-AAGAAGGCCACAATCCTCC, humanBCL-XL target sequence #1 50-CAAACTGCTGCTGTGGCCA,human BCL-XL target sequence #2 50-CTCCGATTCAGTCCCTTCT,human BCL-W target sequence #1 50-AGCGGGTCTCGAACT-CATC, human BCL-W target sequence #2 50-CTGCTGTGGATC-CAGTCAG, human MCL1 target sequence #1 50-CGAAGGAAG-TATCGAATTT, human MCL1 target sequence #2 50-AGAGTGTA-TACAGAACGAA. GFP and shRNA expression was induced byadding 0.5 mg/mL (for A375) or 1 mg/mL (for WM88) of doxycyclinehydrochloride (Dox, Fisher Scientific, AAJ67043AE) to DMEMsupplemented with 10% FBS and 1% penicillin–streptomycin, Gibco.pBABEpuro-HA-p61BRAFV600E, pBABEpuro-HA-MEK1C121S, andpBABEpuro-HA-PDGFRß were previously described (18).

Cell viability assayNote that 5 � 103 of the indicated cells per well were plated in

white 96-well plates (Fisher Scientific, 655098) and cultured for12 hours prior to treatment with the indicated drugs for 72 or96 hours. Cell viability was measured using CellTiter-Glo cellviability assay (Promega, G7571). Normalized %ATP values werecalculated by normalizing luminescence values for each drugtreatment condition to vehicle (DMSO)-treated wells. To generateIC50 curves, the nonlinear fit of Log(Drug) or Drug versusnormalized response (%ATP Normalized to DMSO) with a var-iable slope was calculated in Prism8 (GraphPad). Each drugtreatment condition was represented by at least three biologicalreplicates plated in technical triplicate. The Bliss Index to test forsynergy in drug combinations was performed as previouslydescribed (18, 22).

Flow cytometryNote that 2� 105 of the indicated cells were seeded in 6-well plates

(Genesee Scientific, 25-105) and treated as described, floating andattached cells were harvested, and stained with Annexin V-FITC andpropidium iodide (PI) usingAnnexin V-FITC apoptosis detection kit I(BD Biosciences, 556547) according to the manufacturer's protocols.Data were acquired using the Attune NxT flow cytometry (ThermoScientific) and analyzed with FlowJo 8.7 (Tree Star).

Immunoblot analysisIndicated cells or xenograft-derived tumor lysates were treated as

indicated and then washed with cold 1X PBS and lysed with cold RIPAbuffer containing 1X EDTA-free Halt protease and phosphataseinhibitor cocktail (Thermo Scientific, 78441). The protein concentra-tion was determined by BCA Protein Assay (Pierce) using BSA as astandard. Equal amounts of lysates were resolved by SDS-PAGE usingstandard techniques, and protein was detected with the followingprimary antibodies: rabbit anti-phospho (Thr202/Tyr204)-ERK1/2[1:1,000; Cell Signaling Technology (CST), 9101], mouse anti-ERK1/2 (1:1,000; CST, 9107), rabbit anti-phospho (Ser217/221)-MEK1/2 (1:1,000; CST, 9154), mouse anti-MEK1/2 (1:1,000; CST,4694), rabbit anti-PARP (1:1,000; CST, 9542), rabbit anticleaved PARP(1:1,000; CST, 9541), rabbit anti-CASPASE-3 (1:1,000; CST, 9662),mouse anti–b-actin (1:10,000; CST, 3700), rabbit anti-BCL2 (1:1,000;CST, 4223), rabbit anti–BCL-W (1:1,000; CST, 2724), rabbit anti–BCL-XL (1:1,000; CST, 2764), rabbit anti–MCL1 (1:1,000; CST,94296), rabbit anti-CYTOCHROME C (1:1,000; CST 11940), rabbitanti-HSP60 (1:1,000; CST 4870), mouse anti–a-TUBULIN (1:1,000;CST 3873), followed by detection with one of the horseradish perox-idase–conjugated secondary antibodies: goat anti-rabbit IgG (1:5,000;CST, 7074) or goat anti-mouse IgG (1:5,000; CST, 7076), usingSignalFire (CST) or SignalFire Elite ECL (CST) detection reagents.For detection of CYTOCHROME-C release, cells were treated asdescribed, washed with cold 1X PBS, and mitochondrial and cytosolicfractions were separated using mitochondria/cytosol fractionation kit(BioVision K256) according to the manufacturer's protocols.

High-throughput small-molecule library screenDrug screening was performed by the University of Pennsylvania

High-Throughput Screening Core. To determine the IC20 for theindicated cell lines, 103 cells were plated per well in white 384-wellplates, treated with eight concentrations of TTM (1:3 dilution,ranging from 0.0457 to 100 mmol/L), and cell viability was measured96 hours later with the ATPlite cell viability assay (Perkin Elmer) onan EnVision Plate Reader (Perkin Elmer). To generate IC50 curves,the logistic regression curve of Log(Drug) versus normalizedresponse (%ATP Normalized to DMSO) was fit in TIBCO Spotfiresoftware. The IC20 was calculated using the following formula:IC_F ¼ [(F/(100-F)^(1/H)]�IC50, where H ¼ the Hill slope. Forthe subsequent drug screens, indicated cell lines were treated with100 nmol/L of a compound from the SelleckChem BioactiveCompound Library with or without TTM at the IC20 for therespective cell line. DMSO and doxycycline were used as negativeand positive controls, respectively. Normalized percent inhibition(NPI) was calculated for all data points.

Mouse xenografts and drug treatmentsAll studies were approved by the University of Pennsylvania

Institutional Animal Care and Use Committee. Note that 5 � 106

A375,451-Lu-BRAFiR, or 451-Lu-MEKiR or 8 � 106 451-Lu cellsresuspended in 1X PBS were injected s.c. into flanks of SCID/beige

Kim et al.

Cancer Res; 80(7) April 1, 2020 CANCER RESEARCH1388

on March 1, 2021. © 2020 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 31, 2020; DOI: 10.1158/0008-5472.CAN-19-1784

Page 3: InhibitionofBCL2FamilyMembersIncreasestheEfficacy · Cu chelator tetrathiomolybdate (TTM) is supported by efficacy in BRAFV600E-driven melanoma models, due in part to inhibition

mice (Charles River Laboratory) as previously described (23). Whentumor volume reached approximately 100 mm3, mice were treateddaily via oral gavage: vehicle [1% methylcellulose (Sigma-Aldrich,M0512), 1% DMSO (Sigma-Aldrich)]; 80 mg/kg TTM in vehicle;25mg/kg vemurafenib in 10% ethanol, 30% polyethylene glycol (PEG)400 (EMD Millipore, 8.07485.1000), and 60% Phosal 50PG (Lipoid,368315); 0.5 mg/kg trametinib in PEG; or 25 mg/kg ABT-263 in PEG.

ImmunofluorescenceTumor tissue sections were stained with either a rabbit anti-Ki67

antibody (1:100; Novus Biologicals, NB100-89717) followed by detec-tion with a Alexa Fluor 647–conjugated anti-rabbit IgG (1:1,000;Thermo Scientific, A27040) or DeadEnd Colorimetric TUNEL System(Promega, G3250), following the manufacturer's protocol. Photo-graphs were taken on a Leica DMI6000B–inverted confocal micro-scope (40x) in a blinded fashion. Images were quantified using Image Jsoftware.

ResultsTTM-mediated reductions in BRAFV600E-driven cell viability andMAPK signaling fail to induce apoptosis

To examine whether Cu chelator efficacy could be improved inBRAFV600E-positive melanoma, we first treated BRAFV600E-mutantmelanoma cells in vitro with increasing concentrations of TTM andmeasured cell viability. TTM reduced the number of viable 451-Lu,A375, WM88, and WM983B cells in a dose-dependent manner(Fig. 1A–E). However, TTM treatment failed to increase the percent-age of apoptotic cells (Fig. 1F–I), as measured by Annexin V andPI positivity via flow cytometry (24), despite effectively decreasingERK1/2 phosphorylation (Fig. 1J–M; Supplementary Fig. S1A–S1D).In agreement, cleavage of CASPASE-3 and PARP (25, 26), well-knownmarkers of cells undergoing apoptosis, were only observed at con-centrations higher than the IC50 of TTM (Fig. 1J–M; SupplementaryFig. S1A–S1D). Similarly, treatment with either MAPKi (Supple-mentary Fig. S2A–S2J) was not sufficient to increase markers ofapoptosis (Supplementary Fig. S2K–S2R). Thus, in vitro Cu chelatorsblunt MAPK pathway activation and inhibit the viability ofBRAFV600E-driven melanoma cells but fail to induce apoptosis, akinto treatment with MAPKis, and support the rationale to identifyother therapeutic combinations to improve Cu chelation therapy.

BH3 mimetics synergize with TTM to reduce cell viability ofBRAFV600E-driven melanoma cells

In order to identify collateral drug sensitivities with Cu chelators,we investigated combination of a minimally inhibitory dose of TTMwith the Selleckchem bioactive compound library composed of aunique collection of 2123 bioactive chemical compounds, which areeither FDA-approved or have known bioactivity and safety profiles(Fig. 2A). A375 andWM88 cells were treated with a low dose of eachindividual compound within the Selleckchem bioactive compoundlibrary and either vehicle or the respective IC20 of TTM (Fig. 2A).A 20% reduction in A375 and WM88 cell viability was achievedfor the majority of the bioactive compounds, which reflected TTMefficacy alone, and indicated that most of the bioactive compoundsdisplayed minimal efficacy alone at 100 nmol/L (SupplementaryFig. S3A and S3B). Therefore, initial hits were restricted to com-pounds that when combined with TTM yielded the intended 20% orgreater inhibition of cell viability (Fig. 2B and C; SupplementaryFig. S3A and S3B). Further, synergistic combinations with TTMwere identified by calculating the Bliss Index (22), which considersthe expected additive effect of a drug combination compared with

the observed effect, for each initial compound hit (NPI TTM þCompound ≥20; Fig. 2D and E). Fifty and 233 bioactive compoundssynergistically inhibited cell viability of A375 or WM88 cells,respectively, with a Bliss Index ≥1.5 (Fig. 2F and G). Only fivebioactive compounds scored as hits (NPI TTM þ Compound≥20/Bliss Index ≥1.5) in both BRAFV600E-positive melanoma cells(Fig. 2H and I). These findings establish the utility of employinghigh-throughput small-molecule screens to identify synergisticdrug combinations for potential cancer treatment and more impor-tantly suggest that several bioactive compounds may be useful toenhance Cu chelation therapy efficacy in BRAFV600E-mutantmelanoma.

We were keenly interested in our finding that the pan BH3 mimeticABT-737 synergistically inhibited the viability of both A375 andWM88 cells to the greatest extent when combined with TTM(Fig. 2I). The B-cell lymphoma-2 (BCL2) family consists of two typesof proteins (27), antiapoptotic (e.g., BCL2, BCL-XL, BCL-W, andMCL1; refs. 28–30) and proapoptotic (e.g., BAK, BAX, and BIM),which cooperate through homo- or heterodimer formation to retainthe balance between cell survival and death (31–33). Drugs termed“BH3 mimetics” that bind the surface groove of certain antiapoptoticBCL2 proteins and thereby elicit apoptosis have beendeveloped (34–40). Among them, successful clinical trials of veneto-clax (ABT-199), a specific BCL2i, has led to its FDA approval forchronic lymphocytic leukemia and clinical evaluation for treatment ofother cancers (41–44). However, single-agent inhibition of BCL2 is notsufficient to induce cell death of melanomas due in part to differentialBCL2 protein expression and diversity (45–51). Similarly, ABT-199failed to significantly reduce BRAF-mutant melanoma cell viabilityalone or in combination with TTM in the high-throughput screen(Supplementary Fig. S3C). Inhibition of multiple antiapoptotic BCL2proteins is a major target for melanoma and other cancers (34, 46).

Collateral inhibition of oncogenic MAPK signaling and BCL2proteins with the pan-BH3 mimetic, ABT-737, reduces targetedtherapy resistance in BRAFV600E-driven melanoma (52–54). In sup-port of these findings, combining MAPKis and navitoclax (ABT-263),an orally bioavailable analog of ABT-737 (NCT01989585), is beingtested clinically in BRAFV600E-positive melanoma. Although not everyBH3 mimetic in the high-throughput screen increased the efficacy ofTTM,ABT-263 synergistically reducedA375 cell viability but was onlyadditive in the WM88 cell line (Supplementary Fig. S3C). Combina-tion of IC20 or IC50 doses of TTM and 100 nmol/L ABT-737 wassufficient to significantly reduce cell viability (Fig. 2J andK), but failedto increase the percentage of Annexin V–positive cells, cleaved-CASPASE-3, or cleaved-PARP (Supplementary Fig. S3D–S3G). Fur-ther, the addition of TTM to the combination of vemurafenib andABT-737 or trametinib and ABT-737 significantly reduced A375 andWM88 cell viability (Fig. 2L–O). Treatment with TTM, ABT-737, andeither MAPKi was either equivalent or superior to the combination ofvemurafenib, trametinib, and ABT-737 (Fig. 2L–O). Finally, TTMsignificantly increased the capacity of the combination of the MAPKisand ABT-737 to reduce A375 cell viability (Fig. 2L). These findingsindicate that Cu chelation therapy may be a viable alternative to eitherMAPKi when combined with a BH3 mimetic and, more importantly,could enhance the therapeutic benefit of the clinically investigatedcombination of the MAPKis and a BH3 mimetic.

Genetic loss of select antiapoptotic BCL2 family proteinsdecreases TTM IC50 in BRAFV600E-driven melanoma cells

We next explored what molecular target(s) of ABT-737 impart(s)sensitivity to Cu chelators. ABT-737 selectively targets the

TTM and BH3 Mimetics Synergize to Inhibit BRAFV600E Melanoma

AACRJournals.org Cancer Res; 80(7) April 1, 2020 1389

on March 1, 2021. © 2020 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 31, 2020; DOI: 10.1158/0008-5472.CAN-19-1784

Page 4: InhibitionofBCL2FamilyMembersIncreasestheEfficacy · Cu chelator tetrathiomolybdate (TTM) is supported by efficacy in BRAFV600E-driven melanoma models, due in part to inhibition

0

5

10

15

K A3750 10 5 2.5 1

P-ERK1/2

T-ERK1/2

Clv. CASPASE-3

CASPASE-3

Clv. PARP

PARP

b-Actin

b-Actin b-Actin

b-Actin

WM880 20 10 5 1

1917

116

89

P-ERK1/2

T-ERK1/2

Clv. CASPASE-3

CASPASE-3

Clv. PARP

PARP

L

451-Lu

0.0 0.5 1.0 1.5 2.00

50

100

150

% A

TP(N

orm

aliz

ed to

DM

SO

)

0.0 0.5 1.0 1.5 2.00

50

100

150

% A

TP(N

orm

aliz

ed to

DM

SO

)

BA

F G

H I

A375

0.0 0.5 1.0 1.5 2.00

50

100

150

% A

TP(N

orm

aliz

ed to

DM

SO

)

C WM88 D WM983B

0.0 0.5 1.0 1.5 2.00

50

100

150

% A

TP(N

orm

aliz

ed to

DM

SO

)

E

451-L

uA37

5W

M88

WM98

3B

Annexin-V FITC

VEH

Pro

pidi

um io

dide

451-Lu

Annexin-V FITC

VEH

Pro

pidi

um io

dide

WM88

Annexin-V FITC

VEH TTM (5 mmol/L)

Pro

pidi

um io

dide

A375

Annexin-V FITC

VEH

Pro

pidi

um io

dide

WM983B

J 451-Lu0 30 15 7.5 3.25

P-ERK1/2

T-ERK1/2

19

17Clv. CASPASE-3

CASPASE-3

Clv. PARP

116

89PARP

19

17

116

89

P-ERK1/2

T-ERK1/2

Clv. CASPASE-3

CASPASE-3

Clv. PARP

PARP

WM983BTTM (mmol/L):TTM (mmol/L):

TTM (mmol/L):

TTM (10 mmol/L)

TTM (mmol/L):

TTM (12.5 mmol/L)

TTM (12.5 mmol/L)

Log [TTM(mmol/L)] Log [TTM(mmol/L)] Log [TTM(mmol/L)] Log [TTM(mmol/L)]

TTM

IC50

(mm

ol/L

)

M

19

17

116

89

0 30 15 7.5 3.25

05

101520253035

Ann

exin

V–p

ositi

ve (%

)TT

MVEH

0

5

10

Ann

exin

V–p

ositi

ve (%

)

TTM

VEH

0.0

2.5

5.0

Ann

exin

V–p

ositi

ve (%

)

TTM

VEH

0

5

10

Ann

exin

V–p

ositi

ve (%

)

TTM

VEH

Clv. CAS-3/CAS-3 0.1 1.0 0.3 0.3 0.3

Clv. PARP/PARP 0.1 0.8 0.1 0.1 0.1

Clv. CAS-3/CAS-3 0.1 1.7 0.4 0.1 0.0

Clv. PARP/PARP 0.3 0.6 0.3 0.3 0.3

Fold 1.0 0.6 0.8 0.8 1.0 Fold 1.0 0.2 0.5 0.7 1.0

Fold 1.0 0.5 1.1 1.3 1.1

Clv. CAS-3/CAS-3 0.0 0.0 0.0 0.0 0.0

Clv. PARP/PARP 0.2 0.3 0.1 0.1 0.1

Fold 1.0 0.4 0.4 0.6 0.8

Clv. CAS-3/CAS-3 0.3 1.3 0.5 0.2 0.1

Clv. PARP/PARP 0.2 0.8 0.1 0.1 0.1

Figure 1.

TTM reduces MAPK pathway activation and viability without inducing apoptosis in BRAFV600E-positive melanoma cells. A–D, Relative CellTiter-Glo cell viability �SEM of indicated cells treated with indicated concentrations of TTM. n ¼ 3. E, Scatter dot plot of TTM IC50 in indicated cell lines � SEM. F–I, Representative flowcytometry graphs and scatter dot plots of flow cytometry analysis of Annexin V-FITC and PI� SEM from indicated cell lines treatedwith vehicle (VEH) or TTM. n¼ 3.J–M, Immunoblot detection of phosphorylated (P)-ERK1/2, total (T)-ERK1/2, Cleaved (Clv.) CASPASE-3, CASPASE-3, Clv. PARP, PARP, and b-actin from indicatedcells treated with vehicle or TTM. n ¼ 3. Quantification: P-ERK1/2/T-ERK1/2 normalized to vehicle control and fold change Clv. CASPASE-3/CASPASE-3 and Clv.PARP/PARP.

Kim et al.

Cancer Res; 80(7) April 1, 2020 CANCER RESEARCH1390

on March 1, 2021. © 2020 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 31, 2020; DOI: 10.1158/0008-5472.CAN-19-1784

Page 5: InhibitionofBCL2FamilyMembersIncreasestheEfficacy · Cu chelator tetrathiomolybdate (TTM) is supported by efficacy in BRAFV600E-driven melanoma models, due in part to inhibition

antiapoptotic proteins, BCL2, BCL-W, and BCL-XL, but not MCL1or A1 (37). Knockdown of BCL-XL, BCL-W, or MCL1 with twoindependent doxycycline-inducible shRNAs increased TTM effec-tiveness in both A375 and WM88 cells (Supplementary Fig. S4Aand S4B), as measured by a statistically significant shift in the IC50

of TTM in the presence of doxycycline (Fig. 3A–D). In contrast,depletion of BCL2 failed to decrease TTM IC50 (Fig. 3A–D). Theseresults suggest that lowering the antiapoptotic balance ofBRAFV600E-mutant melanoma cells by targeting specific antiapop-totic BCL2 proteins, BCL-W, BCL-XL, or MCL1, can enhance Cuchelator efficacy.

TTM enhances efficacy of select BH3 mimetics and inducesapoptosis in BRAFV600E-driven melanoma cells

To further examine the generalizability of combining Cu chela-tors with BH3 mimetics in BRAFV600E mutation–positive melano-ma, we tested whether treatment with TTM could lower the IC50 ofa panel of four BH3 mimetics, ABT-737 (37), ABT-199 (BCL2i;ref. 38), WEHI-539 (BCL-XLi; ref. 39), and A1210477 (MCL1i;ref. 40). ABT-737 IC50 was significantly decreased by approximately3-fold in A375 cells and approximately 10-fold in WM88 cellswhen cotreated with the highest concentration of TTM (Fig. 3E–H). However, the BRAF mutation–negative melanoma cell lines,WM3311 and WM3743, did not show a similar dose dependencewhen TTM and ABT-737 were coadministered (SupplementaryFig. S4C and S4D), suggesting that BRAFV600E-mutant melanomasare selectively sensitive to the combination. The efficacy of WEHI-539 and A1210477 was also substantially enhanced with TTM(Fig. 3E–H). TTM only slightly decreased the IC50 of ABT-199in either A375 or WM88 cells (Fig. 3E–H). Further, combination ofTTM with ABT-737, WEHI-539, or A1210477 synergisticallyinhibited A375 and WM88 cell viability, whereas combining TTMwith ABT-199 was additive by Bliss Indexes (Fig. 3I and J).Collectively, the above results suggest that the combined effectsof TTM and BH3 mimetics on BRAF-mutant melanoma cellviability are driven by targeting BCL-W, BCL-XL, and/or MCL1,but not BCL2.

Melanomas are known to harbor elevated levels of the antiapoptoticBCL2 proteins (55, 56), which underlies intrinsic resistance to BH3mimetics.Mechanistically, treatment ofBRAF-mutantmelanoma cellswith TTMwas sufficient to selectively reduce ERK1/2 phosphorylationand the expression of at least two BCL2 proteins (Fig. 4A and B;Supplementary Fig. S5A and S5B). The differential expression ofBCL-W, BCL-XL, and/or MCL1 correlated with significantly increas-ed cytosolic CYTOCHROME-C in A375 or WM88 cells treatedwith both TTM and ABT-737 (Fig. 4C and D; Supplementary S5Cand S5D), indicating that mitochondrial outer membrane permeabi-lization (MOMP) was triggered. In turn, cotreatment with TTM andABT-737 increased the population of cells undergoing apoptoticcell death by 2- or 3-fold (Fig. 4E and F). Finally, cleavedCASPASE-3 and PARP levels, which are initiated by MOMP-mediated CYTOCHROME-C release, were elevated following co-treatment with TTM and any of the three BH3 mimetics, ABT-737,WEHI-539, and A1210477 (Fig. 4G–J; Supplementary Fig. S5E–S5G).As expected, preincubation of A375 or WM88 cells with the irre-versible CASPASE-3i Z-DEVD-FMK blunted the induction ofmarkers of apoptosis in response to the combination of TTM andABT-737 (Fig. 4E–H; Supplementary Fig. S5E and S5F). Takentogether, these results revealed that Cu chelation can enhance theapoptotic activity of BH3mimetics in a CASPASE-dependent mannerin BRAFV600E-driven melanoma cells.

TTM treatment with ABT-737 sensitizes trametinib- orvemurafenib-resistant BRAFV600E-positive melanoma cells

To interrogate the utility of combining Cu chelation with BH3mimetics to restore apoptotic cell death in the setting of MAPKiacquired resistance, we employed BRAFi- or MEK1/2i-resistant var-iants of theBRAFV600E-mutant cell lines 451-Lu andWM983B (20, 21).Treatment with TTM, trametinib, or vemurafenib reduced the IC50 ofABT-737 in parental 451-Lu and WM983B cells (Fig. 5A and B;Supplementary Fig. S6A and S6B). Whereas in 451-Lu-BRAFiR andWM983B-BRAFiR cells, both TTM and trametinib markedlydecreased ABT-737 IC50 (Fig. 5C and D; Supplementary Fig. S6Cand S6D), whereas only TTM was sufficient to sensitize the MEK1/2i-resistant 451-Lu cells to the BH3 mimetic (Fig. 5E and F). Further,cotreatment with TTM and ABT-737 was the only synergistic com-bination in both the parental and resistant cell lines based on BlissIndexes (Fig. 5G–I; Supplementary Fig. S6E and S6F). As such, TTMtreatment enhanced the efficacy of a BH3 mimetic in the setting ofMAPKi resistance.

Combining the Cu chelator and BH3 mimetic in the clinicallyrelevant setting of MAPKi resistance revealed that the combinationwas more efficacious than the FDA-approved combination of vemur-afenib and trametinib in both the na€�ve and MAPKi-resistant 451-Luand WM983B cells (Fig. 5L–O; Supplementary Fig. S6G–S6J). Inaddition, TTM significantly increased the efficacy of the MAPKis andBH3 mimetic in WM983B cells (Supplementary Fig. S6G and S6H).Most importantly, TTM treatment significantly improved the efficacyof ABT-737 combined with the MAPKis in each of the MAPKi-resistant BRAF mutation–positive melanoma cells (Fig. 5L–O; Sup-plementary Fig. S6I and S6J). Thus, Cu chelators may be advantageousin improving the response to the combination of a BRAFi, MEK1/2i,and BH3 mimetic being tested clinically and more importantly be anadditional treatment option in the context of MAPKi resistance.

TTM dampens the apoptotic threshold in response to BH3mimetics by limiting antiapoptotic protein expression inMAPKi-resistant BRAFV600E-positive melanoma cells

Next, we tested and found in parental cells, MAPK signaling wasabrogated by either TTM or MAPKi treatment, but unlike TTM thatsignificantly decreased the expression of BCL-W, BCL-XL, and/orMCL1, trametinib failed to impact the levels of BCL2 proteins (Fig. 6A;Supplementary Fig. S7A). Interestingly, vemurafenib significantlydecreased the expression of BCL2 and BCL-W in 451-Lu cells only(Fig. 6A; Supplementary Fig. S7A). Thus, surprisingly the pattern ofBCL2 protein expression was divergent among the 451-Lu orWM983B cells treated with TTM or the MAPKis (Fig. 6A; Supple-mentary Fig. S7A–S7C). The BRAFi-resistant 451-Lu and WM983Bcells (20, 21), which harbor upregulation of receptor tyrosine kinases asa resistance mechanism, and MEK1/2i-resistant 451Lu cells (20, 21),which harbor a Q60P-activating mutation in MEK2 and BRAFamplification as a resistance mechanism, displayed elevated MEK1/2 and ERK1/2 phosphorylation (Fig. 6A–C; SupplementaryFig. S7A–S7D). Increased MAPK signaling in the BRAFi-resistant variants was dampened by TTM and trametinib treatmentbut not the mutant selective BRAFi vemurafenib (Fig. 6B; Supple-mentary Fig. S7B–S7D). Of clinical relevance, TTM treatment ofthe dual MAPKi-resistant 451-Lu cells was sufficient to reduceERK1/2 phosphorylation, along with BCL-W, BCL-XL, and MCL1expression, which may explain the maintained sensitivity to thecombination of TTM and ABT-737 in these cells (Fig. 5C, E,and F; Supplementary Fig. S7D). These findings indicate thatthe cooperativity between the Cu chelator and BH3 mimetic in

TTM and BH3 Mimetics Synergize to Inhibit BRAFV600E Melanoma

AACRJournals.org Cancer Res; 80(7) April 1, 2020 1391

on March 1, 2021. © 2020 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 31, 2020; DOI: 10.1158/0008-5472.CAN-19-1784

Page 6: InhibitionofBCL2FamilyMembersIncreasestheEfficacy · Cu chelator tetrathiomolybdate (TTM) is supported by efficacy in BRAFV600E-driven melanoma models, due in part to inhibition

-2 -1 0 1 2 30

50

100

150

Log [TTM(mmol/L)]

%A

TP(N

orm

aliz

ed to

DM

SO)

A375WM88

0

50

100

150

%A

TP(N

orm

aliz

ed to

DM

SO

) ***********

**

-20

0

20

40

60

NPI

A375 NPI COMPOUNDA375 NPI COMPOUND + TTMWM88 NPI COMPOUNDWM88 NPI COMPOUND +TTM

-1.5 -1.0 -0.5 0.5 1.0 1.5

-1.5-1.0-0.5

0.51.01.5

EXPECTED EFFECT

OB

SE

RV

ED

EF

FE

CT

-100 -50 50 100

-100

-50

50

100

NPI COMPOUND

NP

I TT

M +

CO

MP

OU

ND

A

+

100 nmol/L

TTM IC20

A375

F A375

I

WM88G

WM88

Total = 2,123

11.59% CANCER1.84% ENDOCRINE4.57% EPIGENETIC13.19% GPCR3.67% ION CHANNEL17.57% KINASE3.91% METABOLISM8.10% MICROBIOLOGY32.93% OTHER2.64% PROTEASE

B

-100 -50 50 100

-100

-50

50

100

NPI COMPOUND

NP

I TT

M +

CO

MP

OU

ND A375 C WM88 D

-1.5 -1.0 -0.5 0.5 1.0 1.5

-1.5-1.0-0.5

0.51.01.5

EXPECTED EFFECT

OB

SE

RV

ED

EF

FE

CT E

Total = 50

8.00% CANCER10.00% EPIGENETIC18.00% KINASE12.00% METABOLISM46.00% OTHER6.00% MICROBIOLOGY

Total = 233

16.31% CANCER3.43% ENDOCRINE3.43% EPIGENETIC18.88% GPCR3.00% ION CHANNEL11.59% KINASE5.58% METABOLISM7.30% MICROBIOLOGYOTHERPROTEASE

J

A375 WM88H

5 23350Compound

Hits

Compound

Hits

NPI TTM + Compound: > 20Bliss Index: > 1.5

0

50

100

150

%A

TP(N

orm

aliz

ed to

DM

SO) ********

********

A375 WM88K

ABT-737

FOSAPREPITANT

FOSCARNET SODIUM

ICARIIN

TTM IC 50 (7

.69 mm

ol/L)

VEH

ABT-737

(100

nmol/L)

TTM IC 20 (0

.85 mm

ol/L)

TTM IC 50 +

ABT-737

TTM IC 50 (1

1.85 m

mol/L)

VEH

ABT-737 (

100 n

mol/L)

TTM IC 20 (3

.9 mm

ol/L)

TTM IC 20 +

ABT-737

TTM IC 50 +

ABT-737

TTM IC 20 +

ABT-737

0

50

100

150

% A

TP(N

orm

alize

d to

DMS

O)

L

0

50

100

150

% A

TP(N

orm

alize

d to

DMS

O)

+- -- - - + + + +- - --- -+ - + - + - ++ - +-- +- - + - - + +- + +-- -- + - + + + ++ + +

A375 M

0

50

100

150

%AT

P(N

orm

alize

d to

DMS

O)

A375

O WM88

****

***********

************ * ***

0

50

100

150

% A

TP(N

orm

alize

d to

DMS

O)

N

TTM IC20 (3.9 mmol/L):

ABT-737 IC20 (6.72 mmol/L):

VEM IC20 (0.19 mmol/L):TRAM IC20 (26 nmol/L):

TTM IC20 (0.85 mmol/L):

ABT-737 IC20 (5.09 mmol/L):

VEM IC20 (0.15 mmol/L):TRAM IC20 (50 nmol/L):

TTM IC50 (11.85 mmol/L):

ABT-737 IC50 (26.86 mmol/L):

VEM IC50 (0.48 mmol/L):TRAM IC50 (65 nmol/L):

TTM IC50 (7.69 mmol/L):

ABT-737 IC50 (20.36 mmol/L):

VEM IC50 (0.46 mmol/L):TRAM IC50 (90 nmol/L):

WM88****

****

*****

******** ****

*****

*

****

******

***

*

********

********

****

**********

****

DIMEGLUMIN

E SALT

HYDRO-

CHLOROTHIAZID

E

+- -- - - + + + +- - --- -+ - + - + - ++ - +-- +- - + - - + +- + +-- -- + - + + + ++ + +

+- -- - - + + + +- - --- -+ - + - + - ++ - +-- +- - + - - + +- + +-- -- + - + + + ++ + +

+- -- - - + + + +- - --- -+ - + - + - ++ - +-- +- - + - - + +- + +-- -- + - + + + ++ + +

Kim et al.

Cancer Res; 80(7) April 1, 2020 CANCER RESEARCH1392

on March 1, 2021. © 2020 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 31, 2020; DOI: 10.1158/0008-5472.CAN-19-1784

Page 7: InhibitionofBCL2FamilyMembersIncreasestheEfficacy · Cu chelator tetrathiomolybdate (TTM) is supported by efficacy in BRAFV600E-driven melanoma models, due in part to inhibition

MAPKi-resistant BRAF mutation–positive melanoma cells may bedriven in part by diminished antiapoptotic protein expression.

To address whether the threshold for apoptosis induction is tippedwhenMAPKi-resistant BRAFV600E-mutantmelanoma cells are treatedwith the combination ofABT-737 and TTMor aMAPKi, wemeasuredthe percentage of apoptotic cells and the levels of markers of apoptosis.Cotreatment with TTM and ABT-737 significantly elevated the num-ber of Annexin V–positive parental and BRAFi-resistant cells, whereasvemurafenib failed to increase the apoptotic response of ABT-737 inthe 451-Lu BRAFiR and WM983B BRAFiR cells (Fig. 6D and E;Supplementary Fig. S7E and S7F). Although the dual combination oftrametinib and ABT-737 was sufficient to increase the percentage ofapoptotic parental and 451-Lu BRAFiR cells to a similar degree tocotreatment with TTM and ABT-737, apoptotic cell death was onlyobserved in MEK1/2i-resistant 451-Lu cells when TTM and ABT-737were coadministered (Fig. 6F). Further, cotreatment of TTM withABT-737 was the only combination to result in elevated cleavage ofCASPASE-3 andPARP in both the parental andMAPKi-resistant 451-Lu cells (Fig. 6G–I; Supplementary Fig. S7G–S7I). These resultsprovide a rationale for a cotreatment strategy with a Cu chelator andBH3 mimetics in the context of MAPKi-resistant forms of BRAFV600E

melanoma.Finally, to extend our findings to in vitro models with well-

characterized mechanisms of resistance to MAPKi, A375 cells wereengineered to stably expressing the p61BRAFV600E splice variant,MEK1C121S, or PDGFRb (18). As expected, each protein conveyedresistance to vemurafenib, whereas only p61BRAFV600E andMEK1C121S imparted resistance to trametinib (18), as measured byan increase in the IC50 of the inhibitors (Supplementary Fig. S8A andS8B). In contrast, TTM reduced the number of viable A375 cellsexpressing these MAPK-dependent drivers of MAPKi resistance(Supplementary Fig. S8A and S8B). Mechanistically, we previouslyshowed that TTM treatment elicits a reduction in ERK1/2 phosphor-ylation in A375 cells stably expressing either the p61BRAFV600E splicevariant, MEK1C121S, or PDGFRb (18). TTM was the only compoundcapable of significantly increasing the cleavage of CASPASE-3 andPARP when combined with ABT-737 in each cell line (SupplementaryFig. S8C and S8D). In summary, these data indicate that MAPK-dependent forms of resistance can be countered by a Cu chelator bymechanistically inhibiting MEK1/2 kinase activity, and apoptosisinduction can be achieved when the Cu chelator is combined withBH3 mimetics.

TTM in combination with ABT-263 suppresses growth of naïveand MEK1/2i-resistant BRAFV600E-positive melanomaxenografts

To evaluate the therapeutic potential of combining TTM withBH3 mimetics in vivo, we utilized mouse xenograft tumor models ofparental and MAPKi-resistant BRAF mutation–positive melanoma.Instead of using ABT-737 for in vivo experiments, we evaluated its

clinically relevant, orally available analog navitoclax, ABT-263 (36).Mice bearing approximately 100 mm3 A375 or 451-Lu parentaltumors were randomly assigned to one of five treatment arms: (i)vehicle, (ii) TTM, (iii) TTM and ABT-263, (iv) BRAFi, MEK1/2i,and ABT-263, and (v) BRAFi, MEK1/2i, ABT-263, and TTM. Thesetreatments arms were prioritized based on their ability to exam thebenefit of combining a Cu chelator and a BH3 mimetic, along with aCu chelator and the clinically investigated combination of MAPKisand a BH3 mimetic. The growth of established xenograft tumorsderived from 451-Lu and A375 cells was reduced when mice weretreated with TTM alone and was enhanced when TTM was com-bined with ABT-263 (Fig. 7A–F). Further, growth of the parentaltumors was severely reduced when mice were treated for 30 dayswith the combination of MAPKis and ABT-263, and this reductionin A375 tumor growth was significantly accentuated when TTM wasalso administered (Fig. 7A–F). Although the in vivo tumor growthresponse to the combination of MAPKis and ABT-263 was atten-uated in BRAFi-resistant 451-Lu cells, the inclusion of TTM delayedtumor growth and significantly improved the time to endpointtumor volume (Fig. 7G–I). Finally, we demonstrate that xenografttumors derived from MEK1/2i-resistant 451-Lu cells failed torespond to the combination of MAPKis and ABT-263, butremained sensitive to TTM and ABT-263 dual treatment, whichalso improved the antitumorigenic properties of the MAPKis(Fig. 7J–L). The body weights of mice, monitored as an indicatorof drugs toxicity, were similar compared with vehicle group (Sup-plementary Fig. S9A–S9D). In concordance with reduced tumorgrowth kinetics, cotreatment with either TTM and ABT-263 orMAPKis and ABT-263 significantly decreased Ki67-positive stain-ing and increased terminal deoxynucleotidyl transferase–mediateddUTP nick end labeling (TUNEL)–positive staining of parentalBRAF-mutant melanoma tumors (Fig. 7M–T; SupplementaryFig. S9E–S9L). Although the addition of TTM to the combinationof the MAPKis and ABT-263 did not increase tumor cell apoptosis(Fig. 7Q–T; Supplementary Fig. S9I–S9L), cell proliferation wasdampened in the parental and MAPKi-resistant tumors (Fig. 7M–P; Supplementary Fig. S9E–S9H). As further evidence of apoptosis,cleaved CASPASE-3 and PARP were elevated in parental tumortissues isolated from mice treated with the dual combination ofTTM and ABT-263, the triple combination of MAPKis and ABT-263, and quadruple combination of TTM, MAPKis, and ABT-263(Supplementary Fig. S9M and S9N). This observation was restrictedto the MAPKi-resistant tumors tissue isolated from mice treatedwith the aforementioned dual and quadruple combinations of TTMand ABT-263 (Supplementary Fig. S9O and S9P). Taken together,these findings support our conclusion that the efficacy of Cuchelators can be enhanced with chemical inducers of apoptosis,and this combination can cooperate with MAPKis to reduce tumorcell growth and survival, especially in the context of refractory orrelapsed BRAFV600E-driven melanoma.

Figure 2.High-throughput screen of bioactive compound library reveals BH3 mimetics as a synergistic combination with TTM in BRAFV600E-positive melanoma cells. A,Schematic diagram of high-throughput screen of 2123 Selleckchem bioactive compound library at 100 nmol/L alone or in combination with the IC20 of TTM(dashed red line) in A375 or WM88. B and C, NPI of TTM þ Compound versus NPI Compound graph for indicated cells treated with Selleckchem bioactivecompound library alone or in combination with IC20 of TTM. Hits (blue circles) are defined as NPI TTM þ Compound ≥20 (dashed red line). D and E, Observedeffect versus expected effect graph for indicated cells of hits from B and C. Hits (blue circles) are defined as Bliss Index ≥1.5 (dashed red line). F and G,Graphical representation of hits defined as NPI TTM þ Compound ≥20 and Bliss Index ≥1.5 in indicated cells. H, Venn diagram relationship betweencompound hits. I, NPI of 5 overlapping hit compounds from H alone or in combination with IC20 of TTM (dashed red line). J and K, Scatter dot plot of %ATPnormalized to vehicle � SEM of indicated cells treated with vehicle or indicated concentrations of drugs. L–O, Scatter dot plot of %ATP normalized to vehicle� SEM of indicated cells treated with vehicle or indicated concentrations of drugs. Results were compared using a one-way ANOVA followed by a Tukeymulticomparisons test. � , P < 0.05; �� , P < 0.01; ���, P < 0.001; ���� , P < 0.0001. n ¼ 3.

TTM and BH3 Mimetics Synergize to Inhibit BRAFV600E Melanoma

AACRJournals.org Cancer Res; 80(7) April 1, 2020 1393

on March 1, 2021. © 2020 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 31, 2020; DOI: 10.1158/0008-5472.CAN-19-1784

Page 8: InhibitionofBCL2FamilyMembersIncreasestheEfficacy · Cu chelator tetrathiomolybdate (TTM) is supported by efficacy in BRAFV600E-driven melanoma models, due in part to inhibition

0.0 0.5 1.00.0

0.5

1.0

EXPECTED EFFECT

OB

SE

RV

ED

EFF

EC

T

0.0 0.5 1.00.0

0.5

1.0

EXPECTED EFFECT

OB

SE

RV

ED

EFF

EC

T

0.0 0.5 1.00.0

0.5

1.0

EXPECTED EFFECT

OB

SE

RV

ED

EFF

EC

T

0

10

20

30

40

IC50

BH

3 m

imet

ic (m

mol

/L)

******

*******

*******

0

10

20

30

40IC

50BH

3 m

imet

ic (m

mol

/L)

**********

******

*******

0.0 0.5 1.0 1.5 2.00

50

100

150

Log [ABT-737(mmol/L)] Log [ABT-199(mmol/L)]

%AT

P(N

orm

aliz

ed to

DM

SO)

0.0 0.5 1.0 1.5 2.00

50

100

150

%AT

P(N

orm

aliz

ed to

DM

SO)

- Dox+ Dox

0

5

10

15

20

25

IC50

TTM

(mm

ol/L

) -Dox+Dox**** ****

********

**** ****

0

5

10

15

IC50

TTM

(μm

ol/L

) -Dox+Dox*** ***

******* ***

**

0.0 0.5 1.0 1.5 2.00

50

100

150

%AT

P(N

orm

aliz

ed to

DM

SO)

- Dox+ Dox

A375Dox shRNA: BCL2 #1A

0.0 0.5 1.0 1.5 2.00

50

100

150

Log [TTM(mmol/L)] Log [TTM(mmol/L)] Log [TTM(mmol/L)] Log [TTM(mmol/L)]

Log [TTM(mmol/L)]Log [TTM(mmol/L)]Log [TTM(mmol/L)]Log [TTM(mmol/L)]

Log [TTM(mmol/L)] Log [TTM(mmol/L)] Log [TTM(mmol/L)] Log [TTM(mmol/L)]

Log [TTM(mmol/L)]Log [TTM(mmol/L)]Log [TTM(mmol/L)]Log [TTM(mmol/L)]

%AT

P(N

orm

aliz

ed to

DM

SO)

- Dox+ Dox

BCL2 #2

B A375

Dox shRNA

BCL2 #1

BCL-XL #1

BCL-W #1

MCL1 #1

BCL2 #2

BCL-XL #2

BCL-W #2

MCL1 #2

WM88Dox shRNA: BCL2 #1

C

BCL2 #2

D WM88

Dox shRNA

BCL2 #1

BCL-XL #1

BCL-W #1

MCL1 #1

BCL2 #2

BCL-XL #2

BCL-W #2

MCL1 #2

0.0 0.5 1.0 1.5 2.00

50

100

150

%AT

P(N

orm

aliz

ed to

DM

SO)

- Dox+ Dox

E A375BCL2/-XL/-WiBH3 Mimetic:

0.0 0.5 1.0 1.5 2.00

50

100

150

%AT

P(N

orm

aliz

ed to

DM

SO)

- Dox+ Dox

0.0 0.5 1.0 1.5 2.00

50

100

150

%AT

P(N

orm

aliz

ed to

DM

SO)

- Dox+ Dox

BCL-XL #1

0.0 0.5 1.0 1.5 2.00

50

100

150

%AT

P(N

orm

aliz

ed to

DM

SO)

- Dox+ Dox

BCL-XL #2

0.0 0.5 1.0 1.5 2.00

50

100

150

%AT

P(N

orm

aliz

ed to

DM

SO)

- Dox+ Dox

BCL-XL #1

BCL-XL #2

BCL2i

0.0 0.5 1.0 1.5 2.00

50

100

150

%AT

P(N

orm

aliz

ed to

DM

SO)

- Dox+ Dox

BCL-W #1

0.0 0.5 1.0 1.5 2.00

50

100

150

%AT

P(N

orm

aliz

ed to

DM

SO)

- Dox+ Dox

BCL-W #2

0.0 0.5 1.0 1.5 2.00

50

100

150

%AT

P(N

orm

aliz

ed to

DM

SO)

- Dox+ Dox

BCL-W #1

BCL-W #2

0.0 0.5 1.0 1.5 2.00

50

100

150

%AT

P(N

orm

aliz

ed to

DM

SO)

- Dox+ Dox

BCL-XL1i

MCL1 #1

0.0 0.5 1.0 1.5 2.00

50

100

150

%AT

P(N

orm

aliz

ed to

DM

SO)

- Dox+ Dox

MCL1 #2

0.0 0.5 1.0 1.5 2.00

50

100

150

%AT

P(N

orm

aliz

ed to

DM

SO)

- Dox+ Dox

MCL1 #1

MCL1 #2

0.0 0.5 1.0 1.5 2.00

50

100

150

%AT

P(N

orm

aliz

ed to

DM

SO)

- Dox+ Dox

0.0 0.5 1.0 1.5 2.00

50

100

150

%AT

P(N

orm

aliz

ed to

DM

SO)

- Dox+ Dox

MCL1i

FA375

ABT-737 ABT-199 A1210477WEHI-539G WM88BCL2/-XL/-WiBH3 Mimetic: BCL2i BCL-XL1i MCL1i

H WM88

ABT-737 ABT-199 A1210477WEHI-539

IC75

IC50

IC250

IC75

IC50

IC250

TTM (mmol/L):

TTM (mmol/L): TTM (mmol/L):

TTM (mmol/L):

IC75

IC50

IC250

IC75

IC50

IC250

I A375

TTM + ABT-737 TTM + ABT-199 TTM + WEHI-539 TTM + A1210477

BCL2i BCL-XL1i MCL1iBCL2/-XL/-WiJ WM88

TTM + ABT-737 TTM + ABT-199 TTM + WEHI-539 TTM + A1210477

BCL2i BCL-XL1i MCL1iBCL2/-XL/-Wi

0.0 0.5 1.0 1.5 2.00

50

100

150

%AT

P(N

orm

aliz

ed to

DM

SO)

0.0 0.5 1.0 1.5 2.00

50

100

150

Log [A1210477(mmol/L)]

%AT

P(N

orm

aliz

ed to

DM

SO)

0.0 0.5 1.0 1.5 2.00

50

100

150

Log [WEHI-539(mmol/L)]

%AT

P(N

orm

aliz

ed to

DM

SO)

0.0 0.5 1.0 1.5 2.00

50

100

150

Log [ABT-737(mmol/L)]

%AT

P(N

orm

aliz

ed to

DM

SO)

0.0 0.5 1.0 1.5 2.00

50

100

150

Log [ABT-199(mmol/L)]

%AT

P(N

orm

aliz

ed to

DM

SO)

0.0 0.5 1.0 1.5 2.00

50

100

150

Log [A1210477(mmol/L)]

%AT

P(N

orm

aliz

ed to

DM

SO)

0.0 0.5 1.0 1.5 2.00

50

100

150

Log [WEHI-539(mmol/L)]

%AT

P(N

orm

aliz

ed to

DM

SO)

0.0 0.5 1.00.0

0.5

1.0

EXPECTED EFFECT

OB

SE

RV

ED

EFF

EC

T

0.0 0.5 1.00.0

0.5

1.0

EXPECTED EFFECT

OB

SE

RV

ED

EFF

EC

T

0.0 0.5 1.00.0

0.5

1.0

EXPECTED EFFECT

OB

SE

RV

ED

EFF

EC

T

0.0 0.5 1.00.0

0.5

1.0

EXPECTED EFFECT

OB

SE

RV

ED

EFF

EC

T

0.0 0.5 1.00.0

0.5

1.0

EXPECTED EFFECT

OB

SE

RV

ED

EFF

EC

T

Figure 3.

Targeting select BCL2 family proteins increases sensitivity to TTM in BRAFV600E-positive melanoma. A and C, Relative CellTiter-Glo cell viability � SEM ofindicated cells stably expressing two independent Dox-inducible shRNAs (#1 and #2) against indicated genes treated with indicated concentrations of TTMwithout or with Dox. n ¼ 3. B and D, Scatter dot plot of TTM IC50 � SEM in indicated cells stably expressing two independent Dox-inducibleshRNAs against indicated genes treated without or with Dox. Results were compared using a two-way ANOVA followed by a Sidak multicomparisonstest. ��� , P < 0.001; ����, P < 0.0001. n¼ 3. E and G, Relative CellTiter-Glo cell viability� SEM of indicated cells treated without or with indicated concentrationsof TTM and increasing concentrations of indicated BH3 mimetics. n ¼ 3. F and H, Scatter dot plot of BH3 mimetic IC50 � SEM in indicated cells treated withoutor with indicated concentrations of TTM and increasing concentrations of indicated BH3 mimetics. n ¼ 3. Results were compared using a two-way ANOVAfollowed by a Tukey multicomparisons test. n ¼ 3. I and J, Graphical representation of Bliss Index (observed effect vs. expected effect) from A375 (E) andWM88 (G) at the indicated drug combinations. Synergistic combinations are indicated by Bliss Index values >1. ��, P < 0.01; ��� , P < 0.001; ���� , P < 0.0001.

Kim et al.

Cancer Res; 80(7) April 1, 2020 CANCER RESEARCH1394

on March 1, 2021. © 2020 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 31, 2020; DOI: 10.1158/0008-5472.CAN-19-1784

Page 9: InhibitionofBCL2FamilyMembersIncreasestheEfficacy · Cu chelator tetrathiomolybdate (TTM) is supported by efficacy in BRAFV600E-driven melanoma models, due in part to inhibition

C

CYTOCHROME C

HSP60

α−TUBULIN

TTM (mmol/L): 0 5 0 5ABT-737 (mmol/L):

TTM (mmol/L):ABT-737 (mmol/L):

TTM (mmol/L):ABT-737 (mmol/L):

TTM (mmol/L):ABT-737 (mmol/L):

TTM (mmol/L):WEHI-539 (mmol/L):

TTM (mmol/L):A1210477 (mmol/L):

0 0 10 100 5 0 50 0 10 10

CytosolMitochondria

CYTOCHROME C

HSP60

α−TUBULIN

WM88

A375

0 10 0 100 0 10 10

0 10 0 100 0 10 10

CytosolMitochondriaD

G0 5 0 50 0 10 10

A375

116

89

19

17

0 5 0 50 0 10 10

- Z-DEVD-FMK + Z-DEVD-FMK

Clv. CASPASE-3

CASPASE-3

Clv. PARP

PARP

H0 10 0 100 0 10 10

WM88

116

89

19

17

0 10 0 100 0 10 10

- Z-DEVD-FMK + Z-DEVD-FMK

Clv. CASPASE-3

CASPASE-3

Clv. PARP

PARP

b-Actin b-Actin

b-Actin

b-Actin b-Actin

b-Actin

A A375

VEH

TTM

IC20

(0.8

5 mm

ol/L

)

TTM

IC50

(7.6

9 μm

ol/L

)P-MEK1/2

T-MEK1/2

P-ERK1/2

T-ERK1/2

BCL2

BCL-XL

BCL-W

MCL1

B WM88

VEH

TTM

IC20

(3.9

mm

ol/L

)

TTM

IC50

(11.

85 μ

mol

/L)

P-MEK1/2

T-MEK1/2

P-ERK1/2

T-ERK1/2

BCL2

BCL-XL

BCL-W

MCL1

I A375

0 5 0 50 0 10 10

0 10 0 100 0 10 10

WM88

116

89

19

17 Clv. CASPASE-3

CASPASE-3

Clv. PARP

PARP

J A375

0 5 0 50 0 10 10

0 10 0 100 0 10 10

WM88

116

89

19

17Clv. CASPASE-3

CASPASE-3

Clv. PARP

PARP

0

2

4

6

8

10

Ann

exin

V–p

ositi

ve (%

)

****

**

E A375

VEH

TTM + ABT-7

37

- Z-DEVD-FMK + Z-DEVD-FMK

0

5

10

15

Ann

exin

V–p

ositi

ve (%

)

****

TTM (10 mm

ol/L)

TTM (5 mm

ol/L)

VEH

ABT-737

(10 m

mol/L)

ABT-737

(10 m

mol/L)

TTM + ABT-7

37

- Z-DEVD-FMK + Z-DEVD-FMK

WM88F

VEH

TTM + ABT-7

37

TTM (10 m

mol/L)

TTM (5 mm

ol/L)

VEH

ABT-737

(10 m

mol/L)

ABT-737

(10 m

mol/L)

TTM + ABT-7

37

Fold 1.0 0.9 1.1

Fold 1.0 0.4 0.6

Fold 1.0 1.6 1.5

Fold 1.0 1.0 1.1

Fold 1.0 0.6 0.6

Fold 1.0 0.1 0.4

Fold 1.0 1.0 1.1

Fold 1.0 0.4 0.9

Fold 1.0 0.9 0.5

Fold 1.0 0.8 0.5

Fold 1.0 0.5 0.6

Fold 1.0 0.6 0.5

CYTOCHROME C/HSP60CYTOCHROME C/α−TUBULIN

1.0 0.6 0.4 0.6 - - - - - - - - 1.0 0.3 2.5 5.4

CYTOCHROME C/HSP60CYTOCHROME C/α−TUBULIN

1.0 0.6 0.7 0.9 - - - - - - - - 1.0 0.5 0.7 2.2

0.2 0.2 0.3 2.2 0.3 0.2 0.2 0.1Clv. CAS-3/CAS-3

0.0 0.0 0.2 0.9 0.0 0.0 0.0 0.4Clv. PARP/PARP

0.2 0.2 0.2 1.3 0.3 0.0 0.2 0.3Clv. CAS-3/CAS-3

0.0 0.0 0.0 0.7 0.0 0.0 0.0 0.2Clv. PARP/PARP

1.6 1.4 1.8 2.1 1.8 1.8 2.6 4.9Clv. CAS-3/CAS-3

0.0 0.1 0.1 0.5 0.3 0.3 0.5 0.9Clv. PARP/PARP

0.5 0.4 0.5 0.5 1.0 1.0 1.8 3.5Clv. CAS-3/CAS-3

0.2 0.1 0.3 0.6 0.1 0.1 0.2 0.5Clv. PARP/PARP

Figure 4.

TTM reduces BCL2 protein expression and lowers the apoptotic threshold when combined with BH3 mimetics in BRAFV600E-positive melanoma. A andB, Immunoblot detection of phosphorylated (P)-MEK1/2, total (T)-MEK1/2, P-ERK1/2, T-ERK1/2, BCL2, BCL-W, BCL-XL, MCL1, and b-actin from indicatedcells treated with vehicle (VEH) or the indicated concentrations of TTM. n ¼ 3. C and D, Immunoblot detection of CYTOCHROME-C, HSP60, and a-TUBULIN ofmitochondrial or cytosolic fractions from indicated cells treated with vehicle or the indicated concentrations of drugs. E and F, Scatter dot plot offlow cytometry analysis of Annexin V-FITC and PI � SEM stained cells pretreated without or with Z-DEVD-FMK, followed by treatment with vehicle orindicated drugs. n ¼ 3. Results were compared using a two-way ANOVA followed by a Tukey multicomparisons test. �� , P < 0.01; ���� , P < 0.0001. n ¼ 3. G andH, Immunoblot detection of Cleaved (Clv.) CASPASE-3, CASPASE-3, Clv. PARP, PARP, and b-actin from indicated cells pretreated without or with Z-DEVD-FMK, followed by treatment with vehicle or indicated drugs. I and J, Immunoblot detection of Clv.CASPASE-3, CASPASE-3, Clv. PARP, PARP, and b-actin fromindicated cells treated with vehicle or indicated drugs. Quantification: P-MEK1/2/T-ERK1/2, P-ERK1/2/T-ERK1/2, BCL2 family protein/b-actin, CYTOCHROME-C/HSP60, and CYTOCHROME-C/a-TUBULIN normalized to vehicle control. Fold change Clv. CASPASE-3/CASPASE-3 and Clv. PARP/PARP.

TTM and BH3 Mimetics Synergize to Inhibit BRAFV600E Melanoma

AACRJournals.org Cancer Res; 80(7) April 1, 2020 1395

on March 1, 2021. © 2020 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 31, 2020; DOI: 10.1158/0008-5472.CAN-19-1784

Page 10: InhibitionofBCL2FamilyMembersIncreasestheEfficacy · Cu chelator tetrathiomolybdate (TTM) is supported by efficacy in BRAFV600E-driven melanoma models, due in part to inhibition

0

5

10

15

0

50

100

150

%AT

P (N

orm

aliz

ed to

DM

SO)

*******

********

** ***

********

******

J

451-Lu

0

50

100

150

%AT

P (N

orm

aliz

ed to

DM

SO)

************

********

0

50

100

150

%AT

P (N

orm

aliz

ed to

DM

SO) 451-Lu

+- -- - - + + + +- - --- -+ - + - + - ++ - +-- +- - + - - + +- + +-- -- + - + + + ++ + +

L 451-Lu BRAFiR

VEM IC50 (0.21 mmol/L):TTM IC50 (12.49 mmol/L):

ABT-737 IC50 (9.02 mmol/L):TRAM IC50 (2.71 nmol/L):

VEM IC50 (0.21 mmol/L):TTM IC50 (12.49 mmol/L):

ABT-737 IC50 (9.02 mmol/L):TRAM IC50 (2.71 nmol/L):

VEM IC50 (0.21 mmol/L):TTM IC50 (12.49 mmol/L):

ABT-737 IC50 (9.02 mmol/L):TRAM IC50 (2.71 nmol/L):

+- -- - - + + + +- - --- -+ - + - + - ++ - +-- +- - + - - + +- + +-- -- + - + + + ++ + +

K

+- -- - - + + + +- - --- -+ - + - + - ++ - +-- +- - + - - + +- + +-- -- + - + + + ++ + +

******** ****

**

***********

N 451-Lu MEKiR

TTM IC20 (3.12 mmol/L):

ABT-737 IC20 (2.26 mmol/L):

VEM IC20 (0.05 mmol/L):TRAM IC20 (0.68 nmol/L):

TTM IC20 (3.12 mmol/L):

ABT-737 IC20 (2.26 mmol/L):

VEM IC20 (0.05 mmol/L):TRAM IC20 (0.68 nmol/L):

TTM IC20 (3.12 mmol/L):

ABT-737 IC20 (2.26 mmol/L):

VEM IC20 (0.05 mmol/L):TRAM IC20 (0.68 nmol/L):

+- -- - - + + + +- - --- -+ - + - + - ++ - +-- +- - + - - + +- + +-- -- + - + + + ++ + +

0

50

100

150

%AT

P (N

orm

aliz

ed to

DM

SO)

****

0

50

100

150

%AT

P (N

orm

aliz

ed to

DM

SO)

****

****

M 451-Lu BRAFiR

+- -- - - + + + +- - --- -+ - + - + - ++ - +-- +- - + - - + +- + +-- -- + - + + + ++ + +

********

********

****

O 451-Lu MEKiR

+- -- - - + + + +- - --- -+ - + - + - ++ - +-- +- - + - - + +- + +-- -- + - + + + ++ + +

****

0

50

100

150

%AT

P (N

orm

aliz

ed to

DM

SO)

***

******

0

5

10

15

IC50

AB

T-73

7 (m

mol

/L)

IC50

AB

T-73

7 (m

mol

/L)

IC50

AB

T-73

7 (m

mol

/L)

IC50

AB

T-73

7 (m

mol

/L)

IC50

AB

T-73

7 (m

mol

/L)

IC50

AB

T-73

7 (m

mol

/L)

IC50

AB

T-73

7 (m

mol

/L)

IC50

AB

T-73

7 (m

mol

/L)

IC50

AB

T-73

7 (m

mol

/L)

0 5 10 150

50

100

150

ABT-737 (mmol/L)

%AT

P(N

orm

aliz

ed to

DM

SO)

A 451-LuCu chelator

TTM (mmol/L):0IC25IC50IC75

BRAFiVEM (mmol/L):

0IC25IC50IC75

MEK1/2iTRAM (nmol/L):

TTM (mmol/L): VEM (mmol/L): TRAM (nmol/L):

TTM (mmol/L): VEM (mmol/L): TRAM (nmol/L):

0IC25IC50IC75

Cu chelator BRAFi MEK1/2i451-LuB

G 451-Lu

VEM + ABT-737BRAFi

TTM + ABT-737Cu chelator

TRAM + ABT-737MEK1/2i

C 451-Lu BRAFiR

Cu chelator

0IC25IC50IC75

BRAFi

0IC25IC50IC75

MEK1/2i

0IC25IC50IC75

Cu chelator BRAFi MEK1/2i451-Lu BRAFiRD

E 451-Lu MEKiR

Cu chelator

0IC25IC50IC75

BRAFi

0IC25IC50IC75

MEK1/2i

0IC25IC50IC75

Cu chelator BRAFi MEK1/2i451-Lu MEKiRF

H 451-Lu BRAFiR

VEM + ABT-737BRAFi

TTM + ABT-737Cu chelator

TRAM + ABT-737MEK1/2i

I 451-Lu MEKiR

VEM + ABT-737BRAFi

TTM + ABT-737Cu chelator

TRAM + ABT-737MEK1/2i

0 5 10 150

50

100

150

ABT-737 (mmol/L)

ABT-737 (mmol/L) ABT-737 (mmol/L)

ABT-737 (mmol/L) ABT-737 (mmol/L)ABT-737 (mmol/L)

ABT-737 (mmol/L)

%AT

P(N

orm

aliz

ed to

DM

SO)

0 5 10 150

50

100

150

ABT-737 (mmol/L)

%AT

P(N

orm

aliz

ed to

DM

SO)

0

5

10

15

0

2

4

6

8

10

0.0 0.5 1.00.0

0.5

1.0

EXPECTED EFFECT

OB

SE

RV

ED

EFF

EC

T

0.0 0.5 1.00.0

0.5

1.0

EXPECTED EFFECT

OB

SE

RV

ED

EFF

EC

T

0.0 0.5 1.00.0

0.5

1.0

EXPECTED EFFECT

OB

SE

RV

ED

EFF

EC

T

TTM (mmol/L)0 IC25 IC50 IC75 0 IC25 IC50 IC75

TRAM (nmol/L)0 IC25 IC50 IC75

VEM (mmol/L)

TTM (mmol/L) TRAM (nmol/L)VEM (mmol/L)

TTM (mmol/L) TRAM (nmol/L)VEM (mmol/L)

0 IC25 IC50 IC75 0 IC25 IC50 IC75 0 IC25 IC50 IC75

0 IC25 IC50 IC75 0 IC25 IC50 IC75 0 IC25 IC50 IC75

0 5 10 150

50

100

150

%AT

P(N

orm

aliz

ed to

DM

SO)

0 5 10 150

50

100

150%

ATP

(Nor

mal

ized

to D

MSO

)

0 5 10 150

50

100

150

%AT

P(N

orm

aliz

ed to

DM

SO)

6

8

10

12

14

0

5

10

15

20

0 5 10 150

50

100

150

%AT

P(N

orm

aliz

ed to

DM

SO)

0 5 10 150

50

100

150

%AT

P(N

orm

aliz

ed to

DM

SO)

0 5 10 150

50

100

150

%AT

P(N

orm

aliz

ed to

DM

SO)

0

5

10

15

0

10

20

30

40

0

5

10

15

0.0 0.5 1.00.0

0.5

1.0

EXPECTED EFFECT

OB

SE

RV

ED

EFF

EC

T

0.0 0.5 1.00.0

0.5

1.0

EXPECTED EFFECT

OB

SE

RV

ED

EFF

EC

T

0.0 0.5 1.00.0

0.5

1.0

EXPECTED EFFECT

OB

SE

RV

ED

EFF

EC

T

0.0 0.5 1.00.0

0.5

1.0

EXPECTED EFFECTO

BS

ER

VE

D E

FFE

CT

0.0 0.5 1.00.0

0.5

1.0

EXPECTED EFFECT

OB

SE

RV

ED

EFF

EC

T

0.0 0.5 1.00.0

0.5

1.0

EXPECTED EFFECT

OB

SE

RV

ED

EFF

EC

T

*****

********

********

******

Figure 5.

TTM sensitizes vemurafenib- or trametinib-resistant BRAFV600E-positive melanoma cells to a BCL2 protein inhibitor. A, C, and E, Relative CellTiter-Glo cellviability � SEM of indicated cells treated without or with indicated concentrations of TTM, vemurafenib (VEM), or trametinib (TRAM) and increasingconcentrations of ABT-737. n ¼ 3. B, D, and F, Scatter dot plot of ABT-737 IC50 � SEM in indicated cells treated without or with indicated concentrationsof TTM, vemurafenib, or trametinib and increasing concentrations of ABT-737. n ¼ 3. Results were compared using a one-way ANOVA followed by a Dunnettmulticomparisons test. n ¼ 3. G–I, Graphical representation of Bliss Index (observed effect vs. expected effect) from indicated cells at the indicateddrug combinations. Synergistic combinations are indicated by Bliss Index values >1. J–O, Scatter dot plot of %ATP normalized to vehicle � SEM of indicatedcells treated with vehicle or indicated concentrations of TTM, vemurafenib, trametinib, ABT-737, or the indicated combinations for 72 hours. Results werecompared using a one-way ANOVA followed by a Tukey multicomparisons test. n ¼ 3. � , P < 0.05; �� , P < 0.01; ��� , P < 0.001; ���� , P < 0.0001.

Kim et al.

Cancer Res; 80(7) April 1, 2020 CANCER RESEARCH1396

on March 1, 2021. © 2020 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 31, 2020; DOI: 10.1158/0008-5472.CAN-19-1784

Page 11: InhibitionofBCL2FamilyMembersIncreasestheEfficacy · Cu chelator tetrathiomolybdate (TTM) is supported by efficacy in BRAFV600E-driven melanoma models, due in part to inhibition

A 451-Lu

VEH

TTM

IC50

(12.

5 mm

ol/L

)

VEM

IC50

(0.2

1 mm

ol/L

)

ABT-

737

IC50

(9.0

2 mm

ol/L

)

TRA

M IC

50 (2

.7 n

mol

/L)

0

20

40

60

80

Ann

exin

V–p

ositi

ve (%

)

** * *

* * **

451-Lu 451-Lu BRAFiR

0

10

20

30

40

Anne

xin

V–po

sitiv

e (%

)

******** ****

TTM (mmol/L): 0 10 0 0 10 0 TRAM (nmol/L): 0 0 20 0 0 20

Clv. CASPASE-3

CASPASE-3

Clv. PARP

PARP

451-Lu MEKiR

ABT-737 (mmol/L): 0 0 0 5 5 5

116

89

1917

G451-Lu

TTM (mmol/L): 0 10 0 0 0 10 0 0TRAM (nmol/L): 0 0 20 0 0 0 20 0

VEM (mmol/L): 0 0 0 0.2 0 0 0 0.2ABT-737 (mmol/L): 0 0 0 0 5 5 5 5

11689

1917

D

T-MEK1/2

T-ERK1/2

BCL2

BCL-W

MCL1

P-MEK1/2

P-ERK1/2

BCL-XL

451-Lu BRAFiR

T-MEK1/2

T-ERK1/2

BCL2

BCL-W

MCL1

P-MEK1/2

P-ERK1/2

BCL-XL

B 451-Lu MEKiR

T-MEK1/2

T-ERK1/2

BCL2

BCL-W

MCL1

P-MEK1/2

P-ERK1/2

BCL-XL

C

TTM (10 m

mol/L)

VEH

ABT-737

(5 mm

ol/L)

TTM + ABT-7

37

VEM (0.2

mmol/L

)

TRAM (20 n

mol/L)

VEM + ABT-7

37

TRAM + ABT-7

37

TTM (10 m

mol/L)

VEH

ABT-737

(5 mm

ol/L)

TTM + ABT-7

37

VEM (0.2

mmol/L

)

TRAM (20 n

mol/L)

VEM + ABT-7

37

TRAM + ABT-7

37

E 451-Lu MEKiR

TTM (10 m

mo/L)VEH

ABT-737

(5 mm

ol/L)

TTM + ABT-7

37

VEM (0.2

mmo/L)

TRAM (20 n

mol/L)

VEM + ABT-7

37

TRAM + ABT-7

370

10

20

30

40

Ann

exin

V–p

ositi

ve (%

)

** **

F

TTM (mmol/L): 0 10 0 0 10 0 VEM (mmol/L): 0 0 0.2 0 0 0.2

451-Lu BRAFiR

ABT-737 (mmol/L): 0 0 0 5 5 5

11689

1917 Clv. CASPASE-3

CASPASE-3

Clv. PARP

PARP

b-Actin

b-Actin b-Actin b-Actin

b-Actin b-Actin

Clv. CASPASE-3

CASPASE-3

Clv. PARP

PARP

H I

VEH

TTM

IC50

(12.

5 mm

ol/L

)

VEM

IC50

(0.2

1 mm

ol/L

)

ABT-

737

IC50

(9.0

2 mm

ol/L

)

TRA

M IC

50 (2

.7 n

mol

/L)

VEH

TTM

IC50

(12.

5 mm

ol/L

)

VEM

IC50

(0.2

1 mm

ol/L

)

ABT-

737

IC50

(9.0

2 mm

ol/L

)

TRA

M IC

50 (2

.7 n

mol

/L)

Fold - - - - - 1.0 1.7 1.0 1.7 1.5 2.1 2.4 2.7 2.1 2.4 0.8 1.0 0.7 0.9 0.9 1.5 1.5 1.5 1.3 1.3 1.0 1.0 0.7 0.3 1.1

Fold - - - - - 1.0 0.3 0.3 0.7 0.8 0.9 0.3 0.9 1.3 0.8 1.5 1.0 0.9 1.1 1.4 3.0 2.7 3.3 5.2 3.1 1.0 0.6 0.3 0.2 1.0

Fold 1.0 0.8 0.7 0.6 0.7 0.4 0.8 0.6 0.6 0.8 0.7 0.9 1.5 1.0 0.7

Fold 1.0 0.6 1.0 0.3 0.9 0.8 1.1 0.9 1.2 0.9 1.1 0.6 1.7 1.5 1.2

Fold 1.0 1.1 1.0 1.1 1.0 1.0 1.0 0.9 0.9 0.9

0.4 0.5 0.4 0.7 0.5 0.9 1.0 1.0 1.0 1.0

- - - - - 1.0 0.6 1.7 1.0 1.0

Fold 1.0 0.5 0.7 0.8 0.7 0.9 0.4 0.7 0.9 0.8 0.9 0.5 1.0 0.9 0.9

Clv. CAS-3/CAS-3 0.5 0.8 0.8 0.8 0.9 3.0 2.8 2.6

Clv. PARP/PARP 0.1 0.1 0.1 0.1 0.1 0.5 0.2 0.2

Clv. CAS-3/CAS-3

0.7 0.4 0.2 0.4 3.3 0.9

Clv. PARP/PARP 0.3 0.3 0.3 0.5 3.1 1.1

Clv. CAS-3/CAS-3

0.2 0.1 0.2 0.5 1.6 0.9

Clv. PARP/PARP 0.0 0.1 0.1 0.2 0.5 0.2

Fold

Fold

Fold

Fold

Fold

Fold

Fold

Fold

Fold

Fold

Fold

Fold

SHORT

LONG

SHORT

LONG

SHORT

LONG

SHORT

LONG

SHORT

LONG

SHORT

LONG

SHORT

LONG

SHORT

LONG

SHORT

LONG

Figure 6.

TTM reduces BCL2 protein expression and lowers the apoptotic threshold when combined with BH3 mimetics in MAPK inhibitor–resistant BRAFV600E-positivemelanoma. A–C, Immunoblot detection of phosphorylated (P)-MEK1/2, total (T)-MEK1/2, P-ERK1/2, T-ERK1/2, BCL2, BCL-W, BCL-XL, MCL1, and b-actin fromindicated cells treatedwith vehicle (VEH) or the indicated concentrations of TTM, trametinib (TRAM), vemurafenib (VEM), or ABT-737. n¼ 3.D–F, Scatter dot plot offlow cytometry analysis of Annexin V-FITC and PI stained � SEM from indicated cells treated with indicated drugs. n¼ 3. Results were compared using a two-wayANOVA followed by a Tukey multicomparisons test. �� , P < 0.01; ���, P < 0.001; ���� , P < 0.0001. n ¼ 3. G–I, Immunoblot detection of Cleaved (Clv.) CASPASE-3,CASPASE-3, Clv. PARP, PARP, and b-actin from indicated cells treated with vehicle, TTM, trametinib, vemurafenib, and/or ABT-737. Quantification: P-MEK1/2/T-ERK1/2, P-ERK1/2/T-ERK1/2, or BCL2 family protein/b-actin, normalized to vehicle control. Fold change Clv. CASPASE-3/CASPASE-3 and Clv. PARP/PARP.

TTM and BH3 Mimetics Synergize to Inhibit BRAFV600E Melanoma

AACRJournals.org Cancer Res; 80(7) April 1, 2020 1397

on March 1, 2021. © 2020 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 31, 2020; DOI: 10.1158/0008-5472.CAN-19-1784

Page 12: InhibitionofBCL2FamilyMembersIncreasestheEfficacy · Cu chelator tetrathiomolybdate (TTM) is supported by efficacy in BRAFV600E-driven melanoma models, due in part to inhibition

0 10 20 300

50

100

Time (days)

Surv

ival

(%)

0 10 20 300 10 20 300

50

100

Time (days)

Surv

ival

(%)

0

500

1,000

1,500

Tum

or v

olum

e (m

m3 )

at e

ndpo

int o

r 30

days

** **** ******

0

500

1,000

1,500

Tum

or v

olum

e (m

m3 )

at e

ndpo

int o

r 30

days **

***** ****

*

*

0 10 20 30 400

500

1,000

1,500

0

500

1,000

1,500

Time (days)

Tum

or v

olum

e (m

m3 )

AA375

BA375

TTMVEH

TTM + ABT-737VEM + TRAM + ABT-737VEM + TRAM + ABT-737 +TTM

TTMVEH

TTM + ABT-737VEM + TRAM + ABT-737VEM + TRAM + ABT-737 +TTM

D 451-Lu E 451-Lu

TTMVEH

TTM + ABT-737VEM + TRAM + ABT-737VEM + TRAM + ABT-737 +TTM

TTMVEH

TTM + ABT-737VEM + TRAM + ABT-737VEM + TRAM + ABT-737 +TTM

G 451-Lu BRAFiR H 451-Lu BRAFiR

TTMVEH

TTM + ABT-737VEM + TRAM + ABT-737VEM + TRAM + ABT-737 +TTM

TTMVEH

TTM + ABT-737VEM + TRAM + ABT-737VEM + TRAM + ABT-737 +TTM

J 451-Lu MEKiR K 451-Lu MEKiR

TTMVEH

TTM + ABT-737VEM + TRAM + ABT-737VEM + TRAM + ABT-737 +TTM

TTMVEH

TTM + ABT-737VEM + TRAM + ABT-737VEM + TRAM + ABT-737 +TTM

0 10 20 30 400

500

1,000

1,500

Time (days)

Tum

or v

olum

e (m

m3 )

0 10 20 300

50

100

Time (days)Su

rviv

al (%

)

C

F

0 10 20 30 400

500

1,000

1,500

Time (days)

Time (days)

Tum

or v

olum

e (m

m3 )

Tum

or v

olum

e (m

m3 )

0 10 20 300

50

100

Time (days)

Surv

ival

(%)

TTMVEH

TTM + ABT-737VEM + TRAM + ABT-737VEM + TRAM + ABT-737 +TTM

TTMVEH

TTM + ABT-737VEM + TRAM + ABT-737VEM + TRAM + ABT-737 +TTM

I

0

10

20

30

Days

to e

ndpo

int

** *** ***** *

A375

451-Lu

451-Lu BRAFiR

L 451-Lu MEKiR

0

10

20

30

Day

s to

end

poin

t

****** *********

TTMVEH

TTM + ABT-737VEM + TRAM + ABT-737VEM + TRAM + ABT-737 +TTM

TTMVEH

TTM + ABT-737VEM + TRAM + ABT-737VEM + TRAM + ABT-737 +TTM

0

20

40

60

80

Ki6

7-P

osi

tive

tum

or

cells

(%

) ********** ****

***

*

M A375

0

20

40

60

80

100

Ki6

7-P

osi

tive

tum

or

cells

(%

) ** ** ****

*

*

451-LuN

0

20

40

60

80

100

Ki6

7-P

osi

tive

tum

or

cells

(%

)

*** *** ****

**

**

*

451-Lu BRAFiRO

0

20

40

60

80

100K

i67-

Po

siti

vetu

mo

r ce

lls (

%)

* **

****

P 451-Lu MEKiR

Q A375 451-LuR 451-Lu BRAFiRS T 451-Lu MEKiR

0

10

20

30

40

TUN

EL-

Pos

itive

tum

or c

ells

(%)

****** ***

0

10

20

30

40

50

TUN

EL-

Pos

itive

tum

or c

ells

(%)

******** **

0

5

10

15

20

25

TU

NE

L-P

osi

tive

tum

or

cells

(%)

*******

*** ******

0

10

20

30

TU

NE

L-P

osi

tive

tum

or

cells

(%)

******

***

***

TTMVEH

TTM + ABT-7

37

VEM + TRAM +

ABT-737

+TTM

VEM + TRAM +

ABT-737 TTM

VEH

TTM + ABT-7

37

VEM + TRAM +

ABT-737

+TTM

VEM + TRAM +

ABT-737 TTM

VEH

TTM + ABT-7

37

VEM + TRAM +

ABT-737

+TTM

VEM + TRAM +

ABT-737 TTM

VEH

TTM + ABT-7

37

VEM + TRAM +

ABT-737

+TTM

VEM + TRAM +

ABT-737

TTMVEH

TTM + ABT-7

37

VEM + TRAM +

ABT-737

+TTM

VEM + TRAM +

ABT-737 TTM

VEH

TTM + ABT-7

37

VEM + TRAM +

ABT-737

+TTM

VEM + TRAM +

ABT-737 TTM

VEH

TTM + ABT-7

37

VEM + TRAM +

ABT-737

+TTM

VEM + TRAM +

ABT-737TTM

VEH

TTM + ABT-7

37

VEM + TRAM +

ABT-737

+TTM

VEM + TRAM +

ABT-737

Figure 7.

Cotreatment with TTM and ABT-263 suppresses in vivo growth of parental and MAPK inhibitor–resistant BRAFV600E-positive melanoma.A, D, G, and J,Mean tumorvolume (mm3)�SEMversus time (days) inmice (n¼ 5or 6) injectedwith indicated cells and treatedwith indicated drugs until tumor volume 1.0 cm3or for 30days.B,E, H, and K, Kaplan–Meier analysis of percentage of mice (n ¼ 5 or 6) with tumor volume ≥ 1.0 cm3 versus time injected with indicated cells treated with indicateddrugs. C and F, Scatter dot plot of tumor volume (mm3)� SEM at endpoint of 1.0 cm3 or at 30 days in mice (n¼ 5 or 6) injected with indicated cells and treated withindicated drugs. Results were compared using unpaired Student t test. n ¼ 5 or 6. I and L, Scatter dot plot of days to endpoint tumor volume � SEM of 1.0 cm3 or30 days of mice (n¼ 5 or 6) injected with indicated cells treated with indicated drugs. Results were compared using unpaired Student t test. n¼ 5 or 6.M–T, Scatterdot plot of % Ki67 or TUNEL-positive cells per tumor � SEM from mice injected with indicated cells treated with indicated drugs. Results were compared usingunpaired Student t test. n ¼ 5 or 6. �, P < 0.05; �� , P < 0.01; ��� , P < 0.001; ���� , P < 0.0001. n ¼ 5 or 6.

Kim et al.

Cancer Res; 80(7) April 1, 2020 CANCER RESEARCH1398

on March 1, 2021. © 2020 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 31, 2020; DOI: 10.1158/0008-5472.CAN-19-1784

Page 13: InhibitionofBCL2FamilyMembersIncreasestheEfficacy · Cu chelator tetrathiomolybdate (TTM) is supported by efficacy in BRAFV600E-driven melanoma models, due in part to inhibition

DiscussionHere, we identified several compounds with the capacity to enhance

TTM efficacy in BRAFV600E-driven melanoma, but focused ourefforts on an inhibitor of BCL2 antiapoptotic proteins. Although thecombination of ABT-737 and a BRAFi induces apoptosis inBRAFV600E-mutant melanoma cells (52–54), it is not effective inBRAFi-resistant cells and required additional targeted or conventionalchemotherapeutics to tip the apoptotic threshold toward celldeath (54). The absence of efficacy achieved with ABT-737 and BRAFiin the context of resistance may be driven by a unique repertoire ofBCL2 proteins in melanocytes (45, 46, 49–51, 54, 56). In addition,proapoptotic BH3-only proteins' expression patterns or their differentbinding affinity to the antiapoptotic proteins can induce differentdependencies on BCL2 proteins (34, 45, 57). Recently, Lee andcolleagues defined that BCL-XL and MCL1 dual targeting is criticalto blunt melanoma cell survival, but it remains to be determinedwhether their combination targeting would reduce MAPKi-resistantmelanoma cell viability (58).

Mechanistically, genetic knockdown of BCL-XL, BCL-W, orMCL1,but not BCL2, revealed that several BCL2 proteins were involved in thesynergistic reduction in cell viability and induction of apoptotic celldeath elicited by cotargeting the MAPK pathway with TTM andantiapoptotic machinery with BH3 mimetics (Figs. 3 and 4; Supple-mentary Figs. S4 and S6). Mechanistically, the Cu chelator lowered thethreshold for apoptotic cell death by limiting the expression of severalBCL2 proteins (Fig. 4; Supplementary Fig. S7). Encouragingly,BRAFV600E-mutant melanoma tumor growth was effectively sup-pressed and controlled when TTM was combined with ABT-263without negatively affecting animal welfare, suggesting that Cu che-lation could be added to current efforts to evaluate MAPKi withnavitoclax in BRAF mutation–positive melanoma (NCT01989585;Fig. 7; Supplementary Fig S9). Our results provide the possibility thatBH3 mimetics can be an effective therapeutic with less adverse effectswhen dosed with a Cu chelator based on the synergistic combination,which is not achieved with vemurafenib or trametinib (Figs. 3 and 5;Supplementary Fig. S5). Finally, in the setting of MAPKi-resistantBRAFV600E-driven melanoma, TTM in combination with ABT-737in vitro or ABT-263 in vivo reduced cell viability, decreased tumorgrowth kinetics, reduced tumor proliferation, and triggered apoptoticcell death (Figs. 5–7; Supplementary Figs. S6–S9). These findingshighlight a need for additional exploration of the interplay between

Cu-dependent MAPK signaling and cellular intrinsic apoptotic path-ways. Thus, we provide a proof of concept that Cu chelation therapysynergistically improves BH3 mimetic efficacy by inducing apoptosisand supports the utility of this combination in patients with BRAFmutation–positive melanoma that do not respond to targeted therapyor immunotherapy, have severe side effects, or acquire resistance.

Disclosure of Potential Conflicts of InterestD.C. Brady has an ownership interest (including patents) in Merlon Inc. No

potential conflicts of interest were disclosed by the other authors.

Authors’ ContributionsConception and design: Y.-J. Kim, D.C. BradyDevelopment of methodology: Y.-J. Kim, T. Tsang, D.C. BradyAcquisition of data (provided animals, acquired and managed patients, providedfacilities, etc.): Y.-J. Kim, J.M. PosimoAnalysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis): Y.-J. Kim, T. Tsang, G.R. Anderson, D.C. BradyWriting, review, and/or revision of the manuscript: Y.-J. Kim, T. Tsang,G.R. Anderson, J.M. Posimo, D.C. BradyAdministrative, technical, or material support (i.e., reporting or organizing data,constructing databases): Y.-J. Kim, D.C. BradyStudy supervision: D.C. Brady

AcknowledgmentsWe thank J. Villanueva (The Wistar Institute) for cell lines, S. Cherry and D.C.

Schultz of the University of Pennsylvania High-Throughput Screening Core fortechnical support, I.A. Asangani, L. Busino, T.P. Gade, B.J. Kim, S.W. Ryeom, and E.S.Witze (University of Pennsylvania) for technical support, discussions, and/or reviewof the article, andD. Sneddon for administrative support. This work was supported bythe PewCharitable Trust Pew Scholars Program in Biomedical Science Award #50359(D.C. Brady) and by a pilot project from Tara Miller Foundation at University ofPennsylvania/Wistar Institute supported by Skin Cancer SPORE Career Enhance-ment Program Award to D.C. Brady (P50CA174523). T. Tsang is supported by NCINRSA Fellowship (F31) F31CA243294. G.R. Anderson is supported by NCI Pre-doctoral to Postdoctoral Transition Fellowship (F99/K00) K00CA222728. J.M.Posimo is supported by American Cancer Society Postdoctoral Fellowship131203PF1714701CCG. This work is dedicated in the memory of Maria Whitehead.

The costs of publication of this article were defrayed in part by thepayment of page charges. This article must therefore be hereby markedadvertisement in accordance with 18 U.S.C. Section 1734 solely to indicatethis fact.

Received June 6, 2019; revised December 17, 2019; accepted January 27, 2020;published first January 31, 2020.

References1. DaviesH, Bignell GR, CoxC, Stephens P, Edkins S, Clegg S, et al.Mutations of the

BRAF gene in human cancer. Nature 2002;417:949–54.2. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA Cancer J Clin 2019;

7–34.3. The Cancer Genome Atlas Network. Genomic classification of cutaneous

melanoma. Cell 2015;161:1681–96.4. Holderfield M, Deuker MM, McCormick F, McMahon M. Targeting RAF

kinases for cancer therapy: BRAF-mutated melanoma and beyond. Nat RevCancer 2014;14:455–67.

5. Wan PTC, Garnett MJ, Roe SM, Lee S, Niculescu-Duvaz D, Good VM, et al.Mechanism of activation of the RAF-ERK signaling pathway by oncogenicmutations of B-RAF. Cell 2004;116:855–67.

6. Flaherty KT, Infante JR, Daud A, Gonzalez R, Kefford RF, Sosman J, et al.Combined BRAF andMEK inhibition inmelanomawith BRAFV600mutations.N Engl J Med 2012;367:1694–703.

7. Robert C, Karaszewska B, Schachter J, Rutkowski P, Mackiewicz A, StroiakovskiD, et al. Improved overall survival in melanoma with combined dabrafenib andtrametinib. N Engl J Med 2015;372:30–9.

8. Larkin J, Ascierto PA, Dreno B, Atkinson V, Liszkay G,MaioM, et al. Combinedvemurafenib and cobimetinib in BRAF-mutated melanoma. N Engl J Med 2014;371:1867–76.

9. Long G V., Stroyakovskiy D, Gogas H, Levchenko E, De Braud F, Larkin J, et al.Dabrafenib and trametinib versus dabrafenib and placebo for Val600 BRAF-mutant melanoma: a multicentre, double-blind, phase 3 randomised controlledtrial. Lancet 2015;386:444–51.

10. Caunt CJ, Sale MJ, Smith PD, Cook SJ. MEK1 and MEK2 inhibitorsand cancer therapy: the long and winding road. Nat Rev Cancer 2015;15:577–92.

11. Solit DB, Rosen N. Towards a unified model of RAF inhibitor resistance.Cancer Discov 2014;4:27–30.

12. Friedman A, Perrimon N. High-throughput approaches to dissecting MAPKsignaling pathways. Methods 2006;40:262–71.

13. Friedman A, Perrimon N. A functional RNAi screen for regulators of receptortyrosine kinase and ERK signalling. Nature 2006;444:230–4.

14. TurskiML, Brady DC, KimHJ, Kim B-E, Nose Y, Counter CM, et al. A novel rolefor copper in Ras/MAPK signaling. Mol Cell Biol 2012;32:1284–95.

TTM and BH3 Mimetics Synergize to Inhibit BRAFV600E Melanoma

AACRJournals.org Cancer Res; 80(7) April 1, 2020 1399

on March 1, 2021. © 2020 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 31, 2020; DOI: 10.1158/0008-5472.CAN-19-1784

Page 14: InhibitionofBCL2FamilyMembersIncreasestheEfficacy · Cu chelator tetrathiomolybdate (TTM) is supported by efficacy in BRAFV600E-driven melanoma models, due in part to inhibition

15. Solit DB, Garraway LA, Pratilas CA, Sawai A, Getz G, Basso A, et al. BRAFmutation predicts sensitivity to MEK inhibition. Nature 2006;439:358–62.

16. BradyDC, CroweMS, TurskiML,HobbsGA, YaoX, ChaikuadA, et al. Copper isrequired for oncogenic BRAF signalling and tumorigenesis. Nature 2014;509:492–6.

17. Brewer GJ, Askari F, Lorincz MT, Carlson M, Schilsky M, Kluin KJ, et al.Treatment of wilson disease with ammonium tetrathiomolybdate. Arch Neurol2006;63:521–7.

18. Brady DC, Crowe MS, Greenberg DN, Counter CM. Copper chelation inhibitsBRAFV600E-driven melanomagenesis and counters resistance to BRAFV600Eand MEK1/2 inhibitors. Cancer Res 2017;77:6240–52.

19. Chan N, Willis A, Kornhauser N, Mward M, Lee SB, Nackos E, et al.Influencing the tumor microenvironment: a phase II study of copperdepletion using tetrathiomolybdate in patients with breast cancer at highrisk for recurrence and in preclinical models of lung metastases. Clin CancerRes 2017;23:666–76.

20. Villanueva J, Vultur A, Lee JT, Somasundaram R, Fukunaga-Kalabis M, CipollaAK, et al. Acquired resistance to BRAF inhibitors mediated by a RAF kinaseswitch in melanoma can be overcome by cotargeting MEK and IGF-1R/PI3K.Cancer Cell 2010;18:683–95.

21. Villanueva J, Infante JR, Krepler C, Reyes-Uribe P, Samanta M, Chen HY, et al.Concurrent MEK2mutation and BRAF amplification confer resistance to BRAFand MEK inhibitors in melanoma. Cell Rep 2013;4:1090–9.

22. Foucquier J, Guedj M. Analysis of drug combinations: current methodologicallandscape. Pharmacol Res Perspect 2015;3:e00149.

23. Hamad NM, Elconin JH, Karnoub AE, Bai W, Rich JN, Abraham RT, et al.Distinct requirements for Ras oncogenesis in human versus mouse cells.Genes Dev 2002;16:2045–57.

24. Vermes I, Haanen C, Steffens-Nakken H, Reutelingsperger C. A novel assay forapoptosis. Flow cytometric detection of phosphatidylserine expression on earlyapoptotic cells using fluorescein labelled annexin V. J Immunol Methods 1995;184:39–51.

25. Nicholson DW, Ali A, Thornberry NA, Vaillancourt JP, Ding CK, Gallant M,et al. Identification and inhibition of the ICE/CED-3 protease necessary formammalian apoptosis. Nature 1995;376:37–43.

26. Li P, Nijhawan D, Budihardjo I, Srinivasula SM, Ahmad M, Alnemri ES, et al.Cytochrome c and dATP-dependent formation of Apaf-1/caspase-9 complexinitiates an apoptotic protease cascade. Cell 1997;91:479–89.

27. Chipuk JE, Moldoveanu T, Llambi F, Parsons MJ, Green DR. The BCL-2 familyreunion. Mol Cell 2010;37:299–310.

28. Boise LH, Gonz�alez-García M, Postema CE, Ding L, Lindsten T, Turka LA, et al.bcl-x, a bcl-2-related gene that functions as a dominant regulator of apoptotic celldeath. Cell 1993;74:597–608.

29. Gibson L, Holmgreen SP, Huang DC, Bernard O, Copeland NG, Jenkins NA,et al. bcl-w, a novelmember of the bcl-2 family, promotes cell survival. Oncogene1996;13:665–75.

30. Kozopas KM,Yang T, BuchanHL, ZhouP, Craig RW.MCL1, a gene expressed inprogrammed myeloid cell differentiation, has sequence similarity to BCL2.Proc Natl Acad Sci U S A 1993;90:3516–20.

31. Oltvai ZN, Milliman CL, Korsmeyer SJ. Bcl-2 heterodimerizes in vivo with aconserved homolog, Bax, that accelerates programmed cell death. Cell 1993;74:609–19.

32. Chittenden T, Flemington C, Houghton AB, Ebb RG, Gallo GJ, Elangovan B,et al. A conserveddomain in Bak, distinct fromBH1 andBH2,mediates cell deathand protein binding functions. EMBO J 1995;14:5589–96.

33. O'Connor L, Strasser A, O'Reilly LA, HausmannG, Adams JM, Cory S, et al. Bim:a novel member of the Bcl-2 family that promotes apoptosis. EMBO J 1998;17:384–95.

34. Letai AG. Diagnosing and exploiting cancer's addiction to blocks in apo-ptosis. Nat Rev Cancer 2008;8:121–32.

35. Delbridge ARD, Strasser A. The BCL-2 protein family, BH3-mimetics andcancer therapy. Cell Death Differ 2015;22:1071–80.

36. Tse C, Shoemaker AR, Adickes J, Anderson MG, Chen J, Jin S, et al. ABT-263: apotent and orally bioavailable Bcl-2 family inhibitor. Cancer Res 2008;68:3421–8.

37. 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 tumours.Nature 2005;435:677–81.

38. 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 whilesparing platelets. Nat Med 2013;19:202–8.

39. Lessene G, Czabotar PE, Sleebs BE, Zobel K, Lowes KN, Adams JM, et al.Structure-guided design of a selective BCL-XL inhibitor. Nat Chem Biol 2013;9:390–7.

40. Leverson JD, Zhang H, Chen J, Tahir SK, Phillips DC, Xue J, et al. Potent andselective small-molecule MCL-1 inhibitors demonstrate on-target cancer cellkilling activity as single agents and in combination with ABT-263 (navitoclax).Cell Death Dis 2015;6:e1590.

41. Davids MS, Seymour JF, Gerecitano JF, Kahl BS, Pagel JM, Wierda WG, et al.Phase I study ofABT-199 (GDC-0199) in patients with relapsed/refractory (R/R)non-Hodgkin lymphoma (NHL): responses observed in diffuse large B-cell(DLBCL) and follicular lymphoma (FL) at higher cohort doses. J Clin Oncol2017;32:8522.

42. KonoplevaM, Pollyea DA, Potluri J, Chyla B, Hogdal L, Busman T, et al. Efficacyand biological correlates of response in a phase II study of venetoclax mono-therapy in patients with acute myelogenous leukemia. Cancer Discov 2016;6:1106–17.

43. Vogler M, Dinsdale D, Dyer MJS, Cohen GM. ABT199 selectivelyinhibits BCL2 but not BCL2L1 and efficiently induces apoptosis ofchronic lymphocytic leukaemic cells but not platelets. Br J Haematol2013;163:139–42.

44. Seymour JF, Davids MS, Pagel JM, Kahl BS, Wierda WG, Miller TP, et al. Bcl-2inhibitor ABT-199 (GDC-0199)monotherapy shows anti-tumor activity includ-ing complete remissions in high-risk relapsed/refractory (R/R) chronic lym-phocytic leukemia (CLL) and small lymphocytic lymphoma (SLL). Blood 2013;122:872.

45. Soderquist RS, Crawford L, Liu E, Lu M, Agarwal A, Anderson GR, et al.Systematic mapping of BCL-2 gene dependencies in cancer reveals moleculardeterminants of BH3 mimetic sensitivity. Nat Commun 2018;9:3513.

46. Anvekar RA, Asciolla JJ, Missert DJ, Chipuk JE. Born to be alive: a role for theBCL-2 family in melanoma tumor cell survival, apoptosis, and treatment.Front Oncol 2011;1.

47. Gautschi O, Tschopp S, Olie RA, Leech SH, Sim~oes-W€ust AP, Ziegler A, et al.Activity of a novel bcl-2/bcl-xL-bispecific antisense oligonucleotide againsttumors of diverse histologic origins. J Natl Cancer Inst 2001;93:463–71.

48. Del Bufalo D, Trisciuoglio D, Scarsella M, Zangemeister-Wittke U, Zupi G.Treatment of melanoma cells with a bcl-2/bcl-xL antisense oligonucleotideinduces antiangiogenic activity. Oncogene 2003;22:8441–7.

49. Thallinger C,WolschekMF,Wacheck V,Maierhofer H, Gunsberg P, Polterauer P,et al. Mcl-1 antisense therapy chemosensitizes human melanoma in a SCIDmouse xenotransplantation model. J Invest Dermatol 2003;120:1081–6.

50. Keuling AM, Felton KEA, Parker AAM, Akbari M, Andrew SE, Tron VA. RNAsilencing ofMcl-1 enhances ABT-737-mediated apoptosis inmelanoma: role fora caspase-8-dependent pathway. PLoS One 2009;4:e6651.

51. Nguyen M, Marcellus RC, Roulston A, Watson M, Serfass L, Madiraju SRM,et al. Small molecule obatoclax (GX15-070) antagonizes MCL-1 and over-comes MCL-1-mediated resistance to apoptosis. Proc Natl Acad Sci U S A2007;104:19512–7.

52. Cragg MS, Jansen ES, Cook M, Harris C, Strasser A, Scott CL. Treatment of B-RAF mutant human tumor cells with a MEK inhibitor requires Bim and isenhanced by a BH3 mimetic. J Clin Invest 2008;118:3651–9.

53. Serasinghe MN,Missert DJ, Asciolla JJ, Podgrabinska S, Wieder SY, Izadmehr S,et al. Anti-apoptotic BCL-2 proteins govern cellular outcome following B-RAF(V600E) inhibition and can be targeted to reduce resistance. Oncogene 2015;34:857–67.

54. Wroblewski D, Mijatov B, Mohana-Kumaran N, Lai F, Gallagher SJ, Haass NK,et al. The BH3-mimetic ABT-737 sensitizes human melanoma cells to apoptosisinduced by selective BRAF inhibitors but does not reverse acquired resistance.Carcinogenesis 2013;34:237–47.

55. McGill GG, Horstmann M, Widlund HR, Du J, Motyckova G, Nishimura EK,et al. Bcl2 regulation by the melanocyte master regulator Mitf modulates lineagesurvival and melanoma cell viability. Cell 2002;109:707–18.

56. Tang L, Tron VA, Reed JC, Mah KJ, Krajewska M, Li G, et al. Expression ofapoptosis regulators in cutaneousmalignant melanoma. Clin Cancer Res 1998;4:1865–71.

57. Certo M, Del Gaizo Moore V, Nishino M, Wei G, Korsmeyer S, Armstrong SA,et al. Mitochondria primed by death signals determine cellular addiction toantiapoptotic BCL-2 family members. Cancer Cell 2006;9:351–65.

58. Lee EF, Harris TJ, Tran S, Evangelista M, Arulananda S, John T, et al. BCL-XLand MCL-1 are the key BCL-2 family proteins in melanoma cell survival.Cell Death Dis 2019;10:342.

Cancer Res; 80(7) April 1, 2020 CANCER RESEARCH1400

Kim et al.

on March 1, 2021. © 2020 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 31, 2020; DOI: 10.1158/0008-5472.CAN-19-1784

Page 15: InhibitionofBCL2FamilyMembersIncreasestheEfficacy · Cu chelator tetrathiomolybdate (TTM) is supported by efficacy in BRAFV600E-driven melanoma models, due in part to inhibition

2020;80:1387-1400. Published OnlineFirst January 31, 2020.Cancer Res   Ye-Jin Kim, Tiffany Tsang, Grace R. Anderson, et al.  

-Driven MelanomaV600ECopper Chelation in BRAFInhibition of BCL2 Family Members Increases the Efficacy of

  Updated version

  10.1158/0008-5472.CAN-19-1784doi:

Access the most recent version of this article at:

  Material

Supplementary

  http://cancerres.aacrjournals.org/content/suppl/2020/01/31/0008-5472.CAN-19-1784.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://cancerres.aacrjournals.org/content/80/7/1387.full#ref-list-1

This article cites 56 articles, 12 of which you can access for free at:

   

  E-mail alerts related to this article or journal.Sign up to receive free email-alerts

  Subscriptions

Reprints and

  [email protected]

To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at

  Permissions

  Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

.http://cancerres.aacrjournals.org/content/80/7/1387To request permission to re-use all or part of this article, use this link

on March 1, 2021. © 2020 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 31, 2020; DOI: 10.1158/0008-5472.CAN-19-1784