+ All Categories
Home > Documents > Antagonism of IAPs Enhances CAR T-cell Efficacy · L-glutamine, nonessential amino acids, 10...

Antagonism of IAPs Enhances CAR T-cell Efficacy · L-glutamine, nonessential amino acids, 10...

Date post: 01-Sep-2020
Category:
Upload: others
View: 0 times
Download: 0 times
Share this document with a friend
11
Priority Brief Antagonism of IAPs Enhances CAR T-cell Efcacy Jessica Michie 1,2 , Paul A. Beavis 1,2 , Andrew J. Freeman 1,2 , Stephin J. Vervoort 3 , Kelly M. Ramsbottom 1 , Vignesh Narasimhan 2,4 , Emily J. Lelliott 2,5 , Najoua Lalaoui 6 , Robert G. Ramsay 2,4 , Ricky W. Johnstone 2,3 , John Silke 6,7 , Phillip K. Darcy 1,2 , Ilia Voskoboinik 1,2 , Conor J. Kearney 1,2 , and Jane Oliaro 1,2 Abstract Chimeric antigen receptor (CAR) T-cell therapy has prov- en successful in the treatment of hematological malignan- cies, notably acute lymphoblastic leukemia and B-cell lym- phoma. However, the efcacy of CAR T cells against solid tumors is poor, likely due to tumor-associated immuno- suppression. Here, we demonstrated that antagonizing the "inhibitor of apoptosis proteins" with the clinical smac- mimetic, birinapant, signicantly enhanced the antitumor activity of CAR T cells in a tumor necrosis factor (TNF)- dependent manner. Enhanced tumor cell death occurred independently of the perforin-mediated granule exocytosis pathway, underscoring the cytotoxic potential of CAR T-cellderived TNF. Combining CAR T-cell therapy with birinapant signicantly reduced established tumor growth in vivo, where either therapy alone was relatively ineffective. Using patient biopsy-derived tumoroids, we demonstrated the synergistic potential of combining CAR T-cell therapy with smac-mimetics. Taken together, we identied CAR T-cellderived TNF as a potent antitumor effector, which can be further harnessed by smac-mimetics. Introduction Adoptive cellular therapy using chimeric antigen receptor (CAR) T cells is showing great promise for the treatment of cancer (1, 2). However, although CAR T cells have been successful in treating hematological malignancies (2), results in the solid tumor setting have been less positive (3). One reason for the lack of efcacy of CAR T cells in solid tumors is the immunosuppressive microenvironment encountered (1). This includes the expression of ligands on tumor cells that bind to checkpoint receptors on the T cells and inhibit their func- tion. For this reason, combining CAR T cells with additional therapies that target checkpoint receptorligand interactions may improve outcomes in solid cancers. PD-1 blockade, for example, has been shown to signicantly enhance the efcacy of both CAR T-cell (4) and adoptive cell therapy (5). The efcacy of CAR T-cell therapy may also be enhanced by agents that sensitize tumor cells to T-cell effector mechanisms, includ- ing cytokines. Smac-mimetics (SM) are a new class of antican- cer agents that can sensitize tumor cells to tumor necrosis factor (TNF)mediated cell death (6). Because TNF is key to the antitumor effector function of cytotoxic T cells (7), this class of small-molecule drugs may also enhance CAR T-cell efcacy. During apoptotic cell death, the mitochondrial protein Smac/Diablo binds to and antagonizes the major "inhibitor of apoptosis proteins" (IAPs). Because IAPs are often upregu- lated in human cancer and facilitate resistance to tumor cell death following therapy, small-molecule mimetics of Smac/Diablo (SMs) were developed (6, 8). Antagonism of IAPs also results in formation of the death-inducing signaling com- plex upon TNF stimulation, rendering the tumor cell sensitive to TNF-induced cell death via the extrinsic pathway (9). Thus, therapies that boost immunity, particularly in the context of elevated TNF secretion, are likely to synergize with SMs. Indeed, IAP antagonism can promote antitumor immunity in multiple myeloma (10) and glioblastoma (11) when combined with checkpoint inhibition, in part due to TNF secreted by immune cells such as CD8 þ T cells. Based on the positive results from preclinical studies (12, 13), several SMs have completed phase I/II trials in patients with advanced solid and hematological cancers, with the SM birina- pant showing the most promise (8). Given the critical role of TNF in antitumor effector functions of cytotoxic T cells (7) and the ability of SMs to sensitize tumor cells to TNF-dependent apopto- sis, we investigated the potential of this class of drugs to enhance CAR T-cell efcacy. We demonstrated that IAP antagonism by birinapant sensitized tumor cells to CAR T-cellderived TNF, signicantly enhancing the antitumor activity of CAR T-cell ther- apy both in vitro and in vivo. The results pave the way for a combination therapy that may improve the efcacy of adoptive cell therapy in solid malignancies. 1 Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia. 2 Sir Peter MacCallum Department of Oncology, The Univer- sity of Melbourne, Parkville, Victoria, Australia. 3 Translational Haematology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia. 4 Gastrointestinal Cancer Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia. 5 Cancer Therapeutics Program, Peter MaCallum Cancer Centre, Melbourne, Victoria, Australia. 6 Cell Signalling and Cell Death Division, The Walter and Eliza Hall Institute for Medical Research, Parkville, Victoria, Australia. 7 Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia. Note: Supplementary data for this article are available at Cancer Immunology Research Online (http://cancerimmunolres.aacrjournals.org/). C.J. Kearney and J. Oliaro contributed equally to this article. Corresponding Author: Jane Oliaro, Peter MacCallum Cancer Centre, 305 Grattan Street, Melbourne, Victoria 3000, Australia. Phone: 61-3-85597094; Fax: 61-3-85597379; E-mail: [email protected] doi: 10.1158/2326-6066.CIR-18-0428 Ó2019 American Association for Cancer Research. Cancer Immunology Research www.aacrjournals.org 183 on December 23, 2020. © 2019 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from Published OnlineFirst January 16, 2019; DOI: 10.1158/2326-6066.CIR-18-0428
Transcript
Page 1: Antagonism of IAPs Enhances CAR T-cell Efficacy · L-glutamine, nonessential amino acids, 10 mmol/L 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid (HEPES) sodium pyruvate,and(Calbiochem)].Tcellswereactivatedwithanti-CD3

Priority Brief

Antagonism of IAPs Enhances CAR T-cell EfficacyJessica Michie1,2, Paul A. Beavis1,2, Andrew J. Freeman1,2, Stephin J. Vervoort3,Kelly M. Ramsbottom1,Vignesh Narasimhan2,4, Emily J. Lelliott2,5, Najoua Lalaoui6,Robert G. Ramsay2,4, Ricky W. Johnstone2,3, John Silke6,7, Phillip K. Darcy1,2,Ilia Voskoboinik1,2, Conor J. Kearney1,2, and Jane Oliaro1,2

Abstract

Chimeric antigen receptor (CAR) T-cell therapy has prov-en successful in the treatment of hematological malignan-cies, notably acute lymphoblastic leukemia and B-cell lym-phoma. However, the efficacy of CAR T cells against solidtumors is poor, likely due to tumor-associated immuno-suppression. Here, we demonstrated that antagonizing the"inhibitor of apoptosis proteins" with the clinical smac-mimetic, birinapant, significantly enhanced the antitumoractivity of CAR T cells in a tumor necrosis factor (TNF)-dependent manner. Enhanced tumor cell death occurred

independently of the perforin-mediated granule exocytosispathway, underscoring the cytotoxic potential of CART-cell–derived TNF. Combining CAR T-cell therapy withbirinapant significantly reduced established tumor growthin vivo, where either therapy alone was relatively ineffective.Using patient biopsy-derived tumoroids, we demonstratedthe synergistic potential of combining CAR T-cell therapywith smac-mimetics. Taken together, we identified CART-cell–derived TNF as a potent antitumor effector, whichcan be further harnessed by smac-mimetics.

IntroductionAdoptive cellular therapy using chimeric antigen receptor

(CAR) T cells is showing great promise for the treatment ofcancer (1, 2). However, although CAR T cells have beensuccessful in treating hematological malignancies (2), resultsin the solid tumor setting have been less positive (3). Onereason for the lack of efficacy of CAR T cells in solid tumors isthe immunosuppressive microenvironment encountered (1).This includes the expression of ligands on tumor cells that bindto checkpoint receptors on the T cells and inhibit their func-tion. For this reason, combining CAR T cells with additionaltherapies that target checkpoint receptor–ligand interactionsmay improve outcomes in solid cancers. PD-1 blockade, forexample, has been shown to significantly enhance the efficacyof both CAR T-cell (4) and adoptive cell therapy (5). The

efficacy of CAR T-cell therapy may also be enhanced by agentsthat sensitize tumor cells to T-cell effector mechanisms, includ-ing cytokines. Smac-mimetics (SM) are a new class of antican-cer agents that can sensitize tumor cells to tumor necrosisfactor (TNF)–mediated cell death (6). Because TNF is key tothe antitumor effector function of cytotoxic T cells (7), thisclass of small-molecule drugs may also enhance CAR T-cellefficacy.

During apoptotic cell death, the mitochondrial proteinSmac/Diablo binds to and antagonizes the major "inhibitorof apoptosis proteins" (IAPs). Because IAPs are often upregu-lated in human cancer and facilitate resistance to tumorcell death following therapy, small-molecule mimetics ofSmac/Diablo (SMs) were developed (6, 8). Antagonism of IAPsalso results in formation of the death-inducing signaling com-plex upon TNF stimulation, rendering the tumor cell sensitiveto TNF-induced cell death via the extrinsic pathway (9). Thus,therapies that boost immunity, particularly in the context ofelevated TNF secretion, are likely to synergize with SMs. Indeed,IAP antagonism can promote antitumor immunity in multiplemyeloma (10) and glioblastoma (11) when combined withcheckpoint inhibition, in part due to TNF secreted by immunecells such as CD8þ T cells.

Based on the positive results from preclinical studies (12, 13),several SMs have completed phase I/II trials in patients withadvanced solid and hematological cancers, with the SM birina-pant showing themost promise (8). Given the critical role of TNFin antitumor effector functions of cytotoxic T cells (7) and theability of SMs to sensitize tumor cells to TNF-dependent apopto-sis, we investigated the potential of this class of drugs to enhanceCAR T-cell efficacy. We demonstrated that IAP antagonism bybirinapant sensitized tumor cells to CAR T-cell–derived TNF,significantly enhancing the antitumor activity of CAR T-cell ther-apy both in vitro and in vivo. The results pave the way for acombination therapy that may improve the efficacy of adoptivecell therapy in solid malignancies.

1Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne,Victoria, Australia. 2Sir Peter MacCallum Department of Oncology, The Univer-sity of Melbourne, Parkville, Victoria, Australia. 3Translational HaematologyProgram, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.4Gastrointestinal Cancer Program, Peter MacCallum Cancer Centre, Melbourne,Victoria, Australia. 5Cancer Therapeutics Program, Peter MaCallum CancerCentre, Melbourne, Victoria, Australia. 6Cell Signalling and Cell Death Division,The Walter and Eliza Hall Institute for Medical Research, Parkville, Victoria,Australia. 7Department of Medical Biology, The University of Melbourne,Parkville, Victoria, Australia.

Note: Supplementary data for this article are available at Cancer ImmunologyResearch Online (http://cancerimmunolres.aacrjournals.org/).

C.J. Kearney and J. Oliaro contributed equally to this article.

Corresponding Author: Jane Oliaro, Peter MacCallum Cancer Centre, 305Grattan Street, Melbourne, Victoria 3000, Australia. Phone: 61-3-85597094;Fax: 61-3-85597379; E-mail: [email protected]

doi: 10.1158/2326-6066.CIR-18-0428

�2019 American Association for Cancer Research.

CancerImmunologyResearch

www.aacrjournals.org 183

on December 23, 2020. © 2019 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst January 16, 2019; DOI: 10.1158/2326-6066.CIR-18-0428

Page 2: Antagonism of IAPs Enhances CAR T-cell Efficacy · L-glutamine, nonessential amino acids, 10 mmol/L 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid (HEPES) sodium pyruvate,and(Calbiochem)].Tcellswereactivatedwithanti-CD3

Materials and MethodsMice

All animal studies were performed in accordance with theNHMRC Australian Code for the Care and Use of Animals forScientific Purposes 8th edition (2013) andwith approval from thePeter MacCallum Cancer Centre Animal Experimentation EthicsCommittee. C57BL/6 human HER2 transgenic mice (14) andC57BL/6 perforin knockout mice (15) were bred in-house.C57BL/6 and C57BL/6 TNF knockout mice were obtained fromthe Walter and Eliza Hall Institute (Parkville, Victoria). All micewere housed in the Peter MacCallum Cancer Centre Animal CoreFacility under specific pathogen-free conditions.

Human samplesHuman PBMCs were isolated from healthy donor buffy coats

supplied by Australian Red Cross Blood Service. Biopsy samplesfrom 2 patients with HER2þ metastatic colorectal cancer wereincluded in this study. All human biopsy studies were conductedin accordance with the Declaration of Helsinki under a protocolapproved by the Peter MacCallum Cancer Centre Human EthicsCommittee, which included informed written consent from allpatients.

Antibodies, cytokines, and drugsNeutralizing antibodies: mouse anti-TNF (BioLegend; clone

MP6-XT22), human anti-TNF (BioLegend; clone MAb11). Enbrel(etanercept) was obtained from the Walter and Eliza HallInstitute of Medical Research (Parkville, Victoria). Antibodies forimmunofluorescence: anti-TNF (BioLegend; clone MP6-XT22),IFNg (eBiosciences; clone XMG1.2), Tubulin (Rockland Immu-nochemicals; clone 600-401-880). Secondary antibodies conju-gated to Alexa fluorophores and ProLong antifade with DAPIwere purchased fromMolecular Probes. For cell stimulation, anti-CD3 (clone 145-2C11) and anti-CD28 (clone 37.51) antibodieswere purchased from BD Biosciences and anti-myc tag (clone2276) fromCell Signaling Technology. IL2 and IL7were obtainedfrom theNIHandPeproTech, respectively. Anti-humanHER2waspurchased from BioLegend (clone 24D2). Birinapant was sup-plied by Medivir dissolved in 12% Captisol solution in water(vehicle).

Cell linesThe cell lines MC38 and E0771 were retrovirally transduced to

express the human HER2 antigen under the control of the mousestem cell virus LTR promoter as previously described (14). Mousecell lines were cultured in DMEM medium and the human celllines HeLa and AU565 in RPMI medium (Gibco), both supple-mented with 10% FCS (Thermo Scientific), penicillin/streptomy-cin (Gibco), and incubated at 37�C in 10%CO2. Tumor lineswereverified to be Mycoplasma-negative by the Victorian InfectiousDiseases Reference Laboratory by PCR analysis.

Generation of CAR T cellsTo generate activated mouse T cells, splenocytes from C57BL/6

mice were filtered through an Acrodisc 70-nm filter with 10 mLred cell lysis buffer (150mmol/LNH4Cl, 10mmol/L KHCO3, and0.1 mmol/L EDTA in 500 mL distilled water pH 7.4) and rinsedwith 10 mL RPMI medium (Gibco; Invitrogen). Human PBMCswere isolated from buffy coats using a Ficoll gradient centri-fugation. The cell suspension was pelleted at 1400 RPM for4 minutes, washed twice in 20 mL RPMI, and resuspended in

10 mL enriched T-cell media [RPMI supplemented with10% FCS (Thermo Scientific), penicillin/streptomycin (Gibco),L-glutamine, nonessential amino acids, 10 mmol/L 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid (HEPES) sodiumpyruvate, and (Calbiochem)]. T cellswere activatedwith anti-CD3(0.5 mg/mL) and anti-CD28 (0.5 mg/mL) in the presence of IL2(100 IU/mL) and IL7 (2 ng/mL) at a density of 5 � 106 cells/mL.

The generation of murine and human CAR T cells was done aspreviously described (14, 16). Briefly, retrovirus encoding a CARcomposed of an extracellular scFv–anti-humanHER2 fused to thetransmembrane domains of CD28 andCD3zwas transduced intoactivated T cells from either the spleen of mice or from humanPBMCs in the presence of IL2 (100 IU/mL) and IL7 (2 ng/mL;PeproTech). The viral packaging GP.E86 cell line containingempty (control) LXSN or LXSN-anti-HER2 CAR retroviral vectorwas generated as described previously (17). The CAR constructused in this study was a second-generation CAR composed of anextracellular single-chain variable fragment (scFv) specific forhuman-HER2, a CD8 hinge region, transmembrane and intracel-lular CD28 and CD3z domains, and a c-Myc tag domain allowingfor CAR T-cell FACS sorting (18). Primary mouse T cells werecollected from splenocytes of C57BL/6 human-HER2 transgenicor wildtype C57BL/6 mice. T cells were retrovirally transduced asdescribed previously (19). After transduction, T cells were main-tained in supplemented RPMI media with IL2 (100 U/mL) andIL7 (2 ng/mL). Tnf�/� or perforin�/� CAR T cells were generated asabove from C57BL/6 Tnf�/� or perforin�/� mice.

In vitro assaysCAR T-cell activity was measured using a standard chromium

release assay as previously described (15). The percentage-specifickilling was determined using the formula: (Sample 51Cr release –Spontaneous 51Cr release)/(Total 51Cr release – Spontaneous 51Crrelease) � 100, and represented as a Michaelis–Menten kinetictrend. All assays were performed using triplicate wells. The rate ofkilling in the control is represented as 100%killing, and treatmentgroupswere comparedwith this value. All killing assayswere doneover 18 hours unless otherwise specified. To generate relativekilling bar graphs, relative killing at the E:T ratio that resultsin 50% maximal killing of the least cytotoxic condition wascompared, using Michaelis–Menten trends, as done previously(15, 20). Cytokines were detected using a mouse inflammationCBA kit (BD Biosciences, 552364) as per manufacturer's instruc-tions and analyzed on a FACSVerse (BD Biosciences). All assayswere analyzed using triplicate determinations.

Flow cytometryCells assessed via flow cytometry were plated out in V-bottom

plates in 100 mL media before being were pelleted by centrifuga-tion at 1400 RPM for 4 minutes and resuspended in 100 mL FACSbuffer (PBS plus 2% FCS). Cells were then stained with an anti-human HER2 antibody (clone 24D2; BioLegend) for 30 minuteson ice before being washed twice in FACS buffer, resuspended in200 mL FACS buffer and transferred to bullet tubes for analysis.Analysis of samples was performed on a Fortessa X20 (BDBiosciences), and data were analyzed with FlowJo (Tree Star).

30RNA sequencing (RNA-seq)MC38 cells expressing human HER2 antigen and AU565

cells were left untreated or cocultured with CAR T cells at a2:1 ratio at 37�C for 6 hours. Cells were washed thoroughly

Michie et al.

Cancer Immunol Res; 7(2) February 2019 Cancer Immunology Research184

on December 23, 2020. © 2019 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst January 16, 2019; DOI: 10.1158/2326-6066.CIR-18-0428

Page 3: Antagonism of IAPs Enhances CAR T-cell Efficacy · L-glutamine, nonessential amino acids, 10 mmol/L 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid (HEPES) sodium pyruvate,and(Calbiochem)].Tcellswereactivatedwithanti-CD3

with PBS to remove contaminating CAR T cells, and cells werelysed. Total RNA was isolated as per the kit manufacturer'sinstructions (Nucleospin RNA extraction kit, Macherey-Nagel).

Single-end 75-bp RNA-seqwas performed in-house at the PeterMacCallum Cancer Centre Molecular Genomics Core on theNextSEquation 500 (Illumina). Demultiplexing of the reads wasperformed using CASAVAv1.8.2, and low-quality reads Q < 30were removed. Cutadapt (v1.9) was used to trim polyA-derivedsequences and biased 30 reads resulting from random hexamerpriming. HISAT2 was used to map the resulting reads to thehuman reference genome. Read counting was performed usingfeatureCounts; part of the subread package. Voom-LIMMA work-flow was used to normalize data for differential gene expression.Gene set enrichment analysis was performed using GSEA2 (v3.0)for identification of enriched signatures obtained from theMSigDB Hallmarks data sets (7). Sequencing data have beendeposited into the Gene-Expression Omnibus (www.ncbi.nlm.nih.gov/geo/) under accession number GSE124140.

Fixed and time-lapse microscopyMicroscopy of T-cell–tumor cell conjugates was done as pre-

viously described (15). The slides were imaged using a FV1000confocal microscope (Olympus). CAR T cells selected for quan-titation had a single contact site with one tumor cell, indicating asingle synapse event. The percentage of T cells that were positivefor the indicated cytokine in a synapse or not was quantitatedmanually by confocal microscopy. A minimum of 100 cells werecounted in each condition.

For time-lapse imaging, CAR T cells were sorted for double-positive CD8 andCAR (anti-myc) cells and labeledwith CellTraceViolet (CTV, Molecular Probes) as follows: 1 � 107 T cells werestained with 5 mmol/L CTV in PBS for 20 minutes at 37�C andwashed for 5 minutes with media. Cells were then pelleted andresuspended in prewarmed media for 10 minutes. Labeled T cellswere then added to adherent targets in media containing propi-dium iodide (PI) and 1 mmol/L birinapant or vehicle control(Captisol). Chamber slides were mounted on a heated stagewithin a temperature-controlled chamber maintained at 37�C,and constant CO2 concentration of 5% was infused using a gasincubation system with active gas mixer ("The Brick"; Ibidi).Optical sectionswere acquired through sequential scans or Bright-field/DIC on a TCS SP5 confocal microscope (Leica Microsys-tems) using a �40 (NA 0.85) air objective and Leica LAS AFsoftware. Image analysis was performed usingMeta-Morph Imag-ing Series 7 software (Universal Imaging).

Mouse experimentsC57BL/6 human HER2 transgenic mice were injected subcu-

taneously with 2.5 � 105 MC38-HER2 cells. At day 7 after tumorinjection, mice were preconditioned with total-body irradiation(4Gy) prior to the administration of 1� 107CAR T cells on days 7and 8. Mice were also treated with 50,000 IU IL2 on days 0 to4 after T-cell transfer. Mice were treated with either vehicle(Captisol) or birinapant (12 mg/kg per mouse) on days 0, 4, and8 after T-cell transfer. Tumor growthwasmonitored approximate-ly every second day using a caliper square to determine theproduct of 2 perpendicular tumor diameters. Themicewere culledwhen the tumor size reached the ethical limit (150 mm2) andrecorded for the survival plot.

Patient-derived tumoroidsTumoroids were grown from tumor cells derived from colo-

rectal patient biopsies. Tumor cells were plated on m-Plate 96Well (ibidi) by adding 20 mL of Matrigel/cell suspension per well,and placed to set in 37�C incubator for 30–60minutes. Followingincubation, 350 mL of OBmedia supplemented with 500 nmol/LA8301 (Torcis, Bioscience), 2� B27 (Gibco, Thermo Scientific),epidermal growth factor (50 ng/mL; Sigma-Aldrich), gastrin(1 mg/mL; Sigma-Aldrich), 1 mmol/L N-acetyl Cyst (Sigma-Aldrich), 5 mmol/L SB202190 (Sigma-Aldrich), 10 mmol/LSB431542 (Sigma-Aldrich), 10 mmol/L Y27632 (Sigma-Aldrich)was added to each well, and tumoroids were grown in hypoxicconditions (37�C, 5% O2, 5% CO2) for 5 to 7 days. Tumoroidswere grown to >50 mm and checked for viability using a lightmicroscope on the day of assay, before proceeding. Tumoroidswere deemed suitable for coculture after 10 to 14 days afterinitial biopsy.

A total volume of 350 mL supplemented OBmedia was appliedto the Matrigel-embedded tumoroids. PI (Sigma-Aldrich) wasadded to a final concentration of 2 mg/mL. CAR T cells/well(2.5 � 105) were then resuspended in this media. Wells were setup with the following conditions: tumoroids þ vehicle control,tumoroids þ CAR T cells/TNF (10 ng/mL), tumoroids þ birina-pant (1mmol/L), tumoroidsþCART cells/TNFþbirinapant, CART cells þ birinapant þ TNF-neutralizing antibody (80 mg/mL).Tumoroids were incubated at 37�C, 5% CO2 for 24 hours, andthen end-point images were acquired. U-Plate 96-well plateswere mounted on a heated stage in a temperature-controlledchamber,maintained at 37�C, 5%CO2 ("The Brick"; ibidi). Usingthe cellSens software (Olympus), imaging of the plate wasobtained on an UPLSAPO 4X (NA 0.16) air objective using aHammamatsu ORCA-Flash 4.0 camera. Z-stack images wereacquired by taking 25 sequential images (Z spacing 50 mm)of PI (emission 656 nm, exposure 368.644 ms) through theMatrigel, and Z stack images converted to one image in EFIformat. Image quantitation was conducted using Meta-MorphImaging Series 7 software. An identical region of interest wasidentified within theMatrigel, and integratedmorphometry anal-ysis was used to filter out individual T cells based on size. Athreshold was applied to delineate PI-positive pixels, and inte-grated intensity was measured. Identical conditions were appliedto compared imges.

Statistical analysesStatistical significance was determined using an unpaired Stu-

dent t test, ANOVA, or log-rank (Mantel–Cox) test for mousesurvival data using GraphPad Prism 7 software. Differences wereconsidered significant when P < 0.05.

ResultsCAR T cells produce cytokines upon tumor cell recognition

TNF is a key inflammatory cytokine produced by cytotoxicT cells upon target recognition that is critical for their antitumoreffector function (7). Here, we generated mouse CAR T cellsspecific for the human HER2 antigen (Fig. 1A; ref. 21) and foundthat upon recognition of HER2-expressing MC38 colon carci-noma cells (MC38-HER2), TNF and interferon gamma (IFNg)were key cytokines produced (Fig. 1B). Human HER2-directedCAR T cells also secreted TNF and IFNg when exposed toHeLa cells that overexpressed HER2 (Supplementary Fig.

Birinapant Enhances CAR T-cell Efficacy

www.aacrjournals.org Cancer Immunol Res; 7(2) February 2019 185

on December 23, 2020. © 2019 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst January 16, 2019; DOI: 10.1158/2326-6066.CIR-18-0428

Page 4: Antagonism of IAPs Enhances CAR T-cell Efficacy · L-glutamine, nonessential amino acids, 10 mmol/L 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid (HEPES) sodium pyruvate,and(Calbiochem)].Tcellswereactivatedwithanti-CD3

Figure 1.

CAR T cells produce inflammatory cytokines upon tumor cell recognition. A, Schematic representation of the CAR T-cell. B, MC38-HER2 cells wereexposed to CAR T cells at the indicated effector-to-target (E:T) ratio. After 4 hours, cytokines in supernatants were measured by cytometric beadarray. C, Left, MC38-HER2 or parental cells were seeded in chamber slides, then overlaid with CAR T cells. After 2 hours, cells were fixed and stainedas indicated, then visualized by confocal microscopy. Scale bar, 10 mm. Right, the percentage of CAR T cells that formed an immunologic synapsewas quantitated by confocal microscopy (>100 cells). The percentage of CAR T cells positive for the indicated cytokines was quantitated by confocalmicroscopy (>100 cells). MC38-HER2 cells were left untreated or incubated with CAR T cells (in triplicate) at low effector-to-target ratio for 6 hoursfollowed by (D) 30 RNA-seq of viable cells. Left, heat map showing significantly regulated genes following CAR T-cell treatment (P < 0.05). GSEAenrichment score plots from RNA-seq data. Right, log2 counts/million cells from RNA-seq data in D. Error bars, mean � SEM of triplicatedeterminations. � , P < 0.05; �� , P < 0.01; ��� , P < 0.001; ���� , P < 0.0001 by an unpaired Student t test.

Michie et al.

Cancer Immunol Res; 7(2) February 2019 Cancer Immunology Research186

on December 23, 2020. © 2019 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst January 16, 2019; DOI: 10.1158/2326-6066.CIR-18-0428

Page 5: Antagonism of IAPs Enhances CAR T-cell Efficacy · L-glutamine, nonessential amino acids, 10 mmol/L 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid (HEPES) sodium pyruvate,and(Calbiochem)].Tcellswereactivatedwithanti-CD3

S1A). Confocal microscopy of mouse CAR T cells coculturedwith MC38-HER2 or parental MC38 cells (Fig. 1C, left) reveal-ed an increase in immunological synapse formation betweenCAR T cells and MC38-HER2 tumor cells and a significantlyincreased number of synapses positive for TNF and IFNg(Fig. 1C, right).

We also detected significant transcriptional changes in theMC38-HER2 tumor cells in response to CAR T-cell treatment(Fig. 1D, left), with GSEA analysis revealing significant geneenrichment in pathways relating to TNF signaling through NF-kBand the IFNg response (Fig. 1D, center). Further analysis of thedata confirmed upregulation of specific genes known to beinduced by TNF and IFNg signaling, including CXCL10, NFKB1A,IRF1, and TAP1 (Fig. 1D, right). Transcriptional analysis of thehuman breast cancer line AU565 (which endogenously expressesHER2) cocultured with human CAR T cells also showed geneenrichment pathways relating to TNF signaling (SupplementaryFig. S1B). These data demonstrated that CAR T cells can producethe inflammatory cytokines, TNF and IFNg , which elicits a tran-scriptional cytokine response in tumor cells.

Birinapant enhances tumor cell death in the presence ofCAR T cells

Because the SM, birinapant, has previously been shown toenhance the death of tumor cells expressing a model antigen inthe presence of transgenic OT-I T cells (15), we wanted todetermine if birinapant could enhance the antitumor activityof CAR T cells to tumor cells expressing the tumor antigenHER2. Mouse CAR T cells were cocultured with MC38 or breastcancer E0771 tumor cells (both engineered to express humanHER2), and human CAR T cells with HeLa or AU565 tumorcells (both endogenously express HER2), and cell death wasmeasured using a chromium release killing assay. In all celllines, a significant increase in cell death in the presence ofbirinapant compared with vehicle was observed (Fig. 2A).Birinapant did not induce cell death of the CAR T cells(Supplementary Fig. S1C), as has been previously observedfor other T cells (15, 22).

To investigate the mechanism of this enhanced tumor celldeath in the presence of birinapant, we monitored the inter-action of mouse CAR T cells with MC38-HER2 cells by time-lapse imaging. We observed a significant increase in the deathof MC38-HER2 cells, indicated by red fluorescence, in thebirinapant-treated cocultures (Fig. 2B; Supplementary MoviesS1–S2). Further analyses of the imaging revealed a significantincrease in the percentage of cell death that occurred inde-pendently of direct CAR T-cell interaction, described as"bystander killing" (refs. 7, 15; Fig. 2B, right). To confirmthis, we conducted a killing assay with a 50:50 mix of unla-beled MC38-HER2 and chromium-labeled MC38 parentalcells. CAR T cells do not recognize non-HER2–expressingtumor cells (14), and therefore, any chromium releaseoccurred independently of direct CAR T-cell interaction(Fig. 2C). These data demonstrated a significant increase inbystander killing in the presence of CAR T cells and birinapant(Fig. 2D).

Birinapant enhances CAR T cell efficacy in a TNF-dependentmanner

We next wanted to determine the mechanism by whichbirinapant enhances tumor cell death in the presence of

CAR T cells. Given the ability of birinapant to sensitizecells to TNF-mediated cell death, we first added an anti-TNF–neutralizing antibody to the cocultures of both mouse andhuman CAR T cells and their cognate target tumor cells(Fig. 3A). In all cases, the addition of anti-TNF significantlyreduced the ability of birinapant to enhance tumor cell death inthe presence of CAR T cells.

To confirm this observation, we also utilized TNF-deficient(Tnf�/�) CAR T cells, which had significantly decreased effectorfunction comparedwith wild-type (WT) CAR T cells, an effect thatwas not amelioratedby the administration of birinapant (Fig. 3B).We detected no TNF production from the Tnf�/� CAR T cells, asexpected, but equivalent amounts of IFNg were secreted betweenTnf–/– and WT CAR T cells (Fig. 3C). To confirm that birinapantdid not enhance tumor cell death via the perforin-mediatedgranule exocytosis pathway, we repeated the assay withperforin�/� CAR T cells. Perforin�/� CAR T cells had decreasedeffector function compared with WT CAR T cells at 4 hours, andtumor cell death was unaffected by the addition of birinapant(Fig. 3D, left). However, over an 18-hour assay, tumor cell deathin the presence of perforin�/� CAR T cells was increased, and thiswas further augmented in the presence of birinapant (Fig. 3D,center). This increase in cell death was significantly reduced bythe addition of a TNF-neutralizing antibody (Fig. 3D, right),demonstrating that the increase in tumor cell death in the pres-ence of CAR T cells and birinapant is perforin-independent andTNF-dependent.

Combination CAR T-cell therapy and birinapant enhancestumor control

Using a human-HER2 transgenic mouse model (14), we nextinvestigated the combination therapy of CAR T cells andbirinapant in vivo. MC38-HER2 tumor–bearing C57BL/6 trans-genic human-HER2 mice were treated with 1 � 107 CAR T cellson days 0 and 1, and birinapant (12 mg/kg) on days 0, 4, and 8.Control mice established robust tumors, which were notcleared by CAR T-cell or birinapant therapy alone. However,the combination of CAR T-cell therapy and birinapant signif-icantly reduced tumor growth, with complete tumor clearanceobserved in 50% mice. Long-term overall survival was signif-icantly increased following the combination therapy (Fig. 4A),demonstrating that the addition of birinapant enhanced CART-cell therapy in vivo.

Birinapant enhances CAR T-cell efficacy using patienttumoroids

To determine if the combination of CAR T cells and birinapantwould enhance tumor cell death in a human cancer, we utilizedpatient biopsy-derived colorectal tumoroids that expressedHER2 (Supplementary Fig. S2A). The tumoroids were notsensitive to TNF or birinapant alone, but the combinationsignificantly enhanced tumor cell death, as indicated by redfluorescence (Fig. 4B; Supplementary Fig. S2B). To test the effectof birinapant on CAR T-cell efficacy using this model, humanCAR T cells were applied to Matrigel-embedded tumoroids(Fig. 4C, top), which endogenously expressed HER2 (Fig. 4C,bottom). The addition of CAR T cells resulted in minimaltumoroid death, whereas the combination of CAR T cells andbirinapant resulted in a significant increase in tumoroid deathcompared with CAR T-cell treatment alone (Fig. 4D). The addi-tion of an anti-TNF–neutralizing antibody to the combination-

Birinapant Enhances CAR T-cell Efficacy

www.aacrjournals.org Cancer Immunol Res; 7(2) February 2019 187

on December 23, 2020. © 2019 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst January 16, 2019; DOI: 10.1158/2326-6066.CIR-18-0428

Page 6: Antagonism of IAPs Enhances CAR T-cell Efficacy · L-glutamine, nonessential amino acids, 10 mmol/L 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid (HEPES) sodium pyruvate,and(Calbiochem)].Tcellswereactivatedwithanti-CD3

Figure 2.

IAP antagonism enhances tumor cell death in the presence of CAR T cells. A, Chromium release assay (18 hours) using mouse CAR T cells withE0771-HER2 and MC38-HER2 targets and human CAR T cells with HeLa-HER2 and AU565 targets (at the indicated E:T ratios) in the presence ofvehicle control (Captisol) or birinapant (1 mmol/L). B, Left, MC38-HER2 cells were seeded in chamber slides, then overlaid with mouse CAR T cells(2:1 ratio) labeled with CTV in PI-containing media. Representative still images at the indicated time points are depicted (hr:min). Scale bar, 50 mm.Right, percentage PI-positive cells without direct CAR T-cell contact was manually quantitated by live imaging. The number of tumor cells that diedthroughout the duration of the movie (as measured by PI uptake) without contact with a T-cell was quantitated and presented in the graph.Quantitation data are pooled from 3 separate camera positions. C, Schematic of chromium release assay to quantify bystander killing. D, Relativekilling of MC38 parental cells by bystander killing in the presence vehicle control (Captisol) or birinapant (1 mmol/L), as depicted in C. All relativekilling data are pooled from 3 independent experiments. Error bars, mean � SEM; �, P < 0.05; �� , P < 0.01 by an unpaired Student t test.

Michie et al.

Cancer Immunol Res; 7(2) February 2019 Cancer Immunology Research188

on December 23, 2020. © 2019 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst January 16, 2019; DOI: 10.1158/2326-6066.CIR-18-0428

Page 7: Antagonism of IAPs Enhances CAR T-cell Efficacy · L-glutamine, nonessential amino acids, 10 mmol/L 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid (HEPES) sodium pyruvate,and(Calbiochem)].Tcellswereactivatedwithanti-CD3

treated tumoroids significantly reduced the amount of tumoroiddeath, suggesting that TNF-mediated bystander killing was theeffector pathway of combination CAR T-cell and birinapant

therapy, not direct T-cell contact (Supplementary Fig. S2C).Together, these data demonstrate that the addition of birinapantto CAR T-cell treatment can significantly enhance tumor cell

Figure 3.

Birinapant enhances tumor cell death in the presence of CAR T cells in a TNF-dependent, perforin-independent manner.A, Chromium release assay (18 hours)using mouse CAR T cells with E0771-HER2 and MC38-HER2 targets and human CAR T cells with HeLa-HER2 and AU565 targets (at the indicated E:T ratio) in thepresence of vehicle control (Captisol) or birinapant (1 mmol/L) and presence or absence of a TNF-neutralizing antibody (10 mg/mL). B, Chromium release assaysusingWT or Tnf�/� CAR T cells and MC38-HER2 cells as targets (at the indicated E:T ratio) in the presence or absence of vehicle control (Captisol) or birinapant(1 mmol/L). C,WT or Tnf�/� CAR T cells were exposed to MC38-HER2 cells at the indicated E:T ratio. After 4 hours, cytokines in supernatants were measured byCBA. D, Left, chromium release assay (4 hours) usingWT or perforin�/� CAR T cells and MC38-HER2 cells as targets (at the indicated E:T ratio) in the presence ofvehicle control (Captisol) or birinapant (1 mmol/L). Center, chromium release assay after 18 hours. Right, chromium release assay using MC38 HER2 cells andWTor perforin–/– CAR T cells (at the indicated E:T ratio) untreated (vehicle) in the presence or absence of anti-TNF–neutralizing antibody (10 mg/mL). Error bars,mean� SEM of triplicate determinations from a representative of 2 independent experiments. � , P < 0.05 by an unpaired Student t test.

Birinapant Enhances CAR T-cell Efficacy

www.aacrjournals.org Cancer Immunol Res; 7(2) February 2019 189

on December 23, 2020. © 2019 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst January 16, 2019; DOI: 10.1158/2326-6066.CIR-18-0428

Page 8: Antagonism of IAPs Enhances CAR T-cell Efficacy · L-glutamine, nonessential amino acids, 10 mmol/L 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid (HEPES) sodium pyruvate,and(Calbiochem)].Tcellswereactivatedwithanti-CD3

Figure 4.

Birinapant enhances CAR T-cell therapy in vivo and in patient-derived tumoroids. C57BL/6 human HER2 transgenic mice were injected subcutaneously withMC38-HER2 cells (2.5� 105 cells/mouse) and tumors were allowed to establish for 7 days. Mice received total body irradiation (4 Gy) before adoptive transfer ofanti-HER2 CAR T cells intravenously (1� 107 cells/mouse), and a total of 5 doses of intraperitoneal IL2 (50,000 IU/injection) over 5 consecutive days. Birinapantwas administered at 12 mg/kg by intraperitoneal injection on days 0, 4, and 8 after CAR T-cell injection. A, Growth of MC38 tumors at indicated time points andtumor size at 12 days after treatment. Data are mean� SEM of 14–15 mice/group, pooled from 2 independent experiments. � , P < 0.05 by ANOVA. Right, survivalof mice to day 50 after treatment. Data are mean� SEM of 14–15 mice/group, pooled from 2 independent experiments. �� , P < 0.01; ���� , P < 0.0001 by log-rank(Mantel–Cox) test. B, Fluorescent microscopy of patient-derived colorectal tumoroids embedded in Matrigel and administered vehicle control (Captisol) orsoluble TNF (10 ng/mL) in the presence or absence of birinapant (1 mmol/L) in PI-containing media. C, Top, schematic representation of CAR T-cell treatment ofMatrigel-embedded tumoroids. Dashed lines, production of TNF by CAR T cells and bystander killing of tumoroids by TNF-mediated apoptosis. Bottom,tumoroids were dissociated into single-cell suspensions and analyzed for HER2 expression by flow cytometry compared with isotype control. D, Tumoroids weretreated with vehicle control (Captisol) or CAR T cells in the presence or absence of birinapant (1 mmol/L), or in the presence of birinapant and a neutralizing TNFantibody, in PI-containing media. Total fluorescent intensity was quantified using Meta-Morph software. Data are mean� SEM from tumoroids derived from 2individual patients in 3 independent experiments. � , P < 0.05; �� , P < 0.01 by ANOVA.

Michie et al.

Cancer Immunol Res; 7(2) February 2019 Cancer Immunology Research190

on December 23, 2020. © 2019 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst January 16, 2019; DOI: 10.1158/2326-6066.CIR-18-0428

Page 9: Antagonism of IAPs Enhances CAR T-cell Efficacy · L-glutamine, nonessential amino acids, 10 mmol/L 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid (HEPES) sodium pyruvate,and(Calbiochem)].Tcellswereactivatedwithanti-CD3

death and is likely to be a combination for more effectiveadoptive cellular therapy.

DiscussionAlthough the development of CAR T-cell therapy has been

revolutionary within the field of immunotherapy, the clinicalsuccess has been limited to hematological cancers (2) and ahandful of solid cancers (23, 24). A limited number of studieshave reported combination therapies that enhance CAR T-celltherapy in solid cancers. Here, we demonstrated that the SM,birinapant, significantly enhanced the efficacy of CAR T cells in asolid tumor setting, in a TNF-dependent manner.

Birinapant is currently undergoing clinical trials as a singleagent for a number of hematological and solid tumor can-cers (25, 26). Many tumors upregulate IAPs (27) to facilitateresistance to apoptosis following standard chemotherapies(28). The degradation of IAPs by SMs results in the secretion ofTNF, which can then induce apoptosis in an autocrinemanner viathe extrinsic pathway (6). Tumor cells that do not secrete TNFfollowing IAP degradation can also be killed by the exogenousaddition of TNF (29). We have previously shown the potentialof cytotoxic lymphocytes as a source of TNF that can be utilizedby SMs in vitro (15). Using a human-HER2 self-antigen mousemodel, we demonstrated here that HER2þ MC38 tumor–bearingmice treated with CAR T cells and birinapant had a significantsurvival advantage compared with either monotherapy, withcomplete remission achieved in half the mice. Given the lack ofrobust responses to CAR T-cell therapy observed in this mod-el (14), these data may have significant implications for previ-ously difficult-to-treat solid malignancies.

In order to demonstrate the potency of this combinationtherapy on patient tissue, we utilized HER2þ patient-derivedcolorectal tumoroids embedded in Matrigel as a model of asolid tumor. We have previously uncovered a critical role of theTNF-mediated apoptotic pathway as a mechanism of inducingtumor cell death in the absence of direct T-cell contact (7, 15).We demonstrated that, even with low CAR T-cell penetration, inthe presence of birinapant, CAR T-cell–derived TNF can pen-etrate through the Matrigel and trigger apoptosis of buriedtumor cells through this "bystander" effect (7, 15). This maytherefore represent a therapy that overcomes the limitation of

small numbers of CAR T cells reaching and penetrating thetumor efficiently, while allowing for potent killing of antigen-negative tumor cells by TNF. Capitalizing on the enhanced TNFproduced by T cells following checkpoint blockade (15) mayalso provide a therapy by combining CAR T-cell administrationalong with birinapant and anti–PD-1 treatment. Given all 3therapies are either in the clinic or currently being assessed inclinical trials, we expect there can be rapid translation of thistherapy into patients.

Disclosure of Potential Conflicts of InterestR.W. Johnstone reports receiving commercial research grants fromRoche and

AstraZeneca and is a consultant/advisory board member for MecRx. No poten-tial conflicts of interest were disclosed by the authors.

Authors' ContributionsConception and design: J. Michie, C.J. Kearney, J. OliaroDevelopment of methodology: J. Michie, P.A. Beavis, V. Narasimhan,R.G. Ramsay, J. OliaroAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): J. Michie, P.A. Beavis, A.J. Freeman,K.M. Ramsbottom, V. Narasimhan, E.J. Lelliott, R.G. Ramsay, J. Silke, J. OliaroAnalysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis): J. Michie, S.J. Vervoort, K.M. Ramsbottom,V. Narasimhan, N. Lalaoui, R.G. Ramsay, P.K. Darcy, I. Voskoboinik, J. OliaroWriting, review, and/or revision of the manuscript: J. Michie, P.A. Beavis,V. Narasimhan, R.G. Ramsay, R.W. Johnstone, P.K. Darcy, C.J. Kearney, J. OliaroAdministrative, technical, or material support (i.e., reporting or organizingdata, constructing databases): V. Narasimhan, J. SilkeStudy supervision: S.J. Vervoort, R.G. Ramsay, I. Voskoboinik, C.J. Kearney,J. Oliaro

AcknowledgmentsC.J. Kearney is funded by an NHMRC Early Career Fellowship, S.J. Vervoort

by a Rubicon Fellowship (Netherlands Organization for Scientific Research),R.G. Ramsay by an NHMRC project grant, P.K. Darcy by an NHMRC programgrant, I. Voskoboinik by an NHMRC fellowship and project grant, and J. Oliaroby an NHMRC and an NBCF project grant.

We thank Medivir (Sweden) for supply of birinapant, A/Professor MichaelKershaw for the HER2-expressing tumor lines, Professor Joseph Trapani forperforin-deficient mice, and Elhadi Lich for assistance with tumoroidanalyses.

Received June 26, 2018; revised October 6, 2018; accepted January 10, 2019;published first January 16, 2019.

References1. Beavis PA, Milenkovski N, Henderson MA, John LB, Allard B, Loi S, et al.

Adenosine receptor 2Ablockade increases the efficacy of anti-PD-1 throughenhanced antitumor T-cell responses. Cancer Immunol Res 2015;3:506–17.

2. Maude SL, Teachey DT, Porter DL, Grupp SA. CD19-targeted chimericantigen receptor T-cell therapy for acute lymphoblastic leukemia. Blood2015;125:4017–23.

3. Kershaw MH, Westwood JA, Darcy PK. Gene-engineered T cells for cancertherapy. Nat Rev Cancer 2013;13:525–41.

4. John LB, Kershaw MH, Darcy PK. Blockade of PD-1 immunosup-pression boosts CAR T-cell therapy. Oncoimmunology 2013;2:e26286.

5. Blake SJ, Ching AL, Kenna TJ, Galea R, Large J, Yagita H, et al. Blockade ofPD-1/PD-L1 promotes adoptive T-cell immunotherapy in a tolerogenicenvironment. PLoS One 2015;10:e0119483.

6. Vince JE, Wong WW, Khan N, Feltham R, Chau D, Ahmed AU, et al. IAPantagonists target cIAP1 to induce TNFalpha-dependent apoptosis. Cell2007;131:682–93.

7. Kearney CJ, Vervoort SJ, Hogg SJ, RamsbottomKM, Freeman AJ, Lalaoui N,et al. Tumor immune evasion arises through loss of TNF sensitivity.Sci Immunol 2018;3.

8. Condon SM, Mitsuuchi Y, Deng Y, LaPorte MG, Rippin SR, Haimowitz T,et al. Birinapant, a smac-mimetic with improved tolerability for thetreatment of solid tumors and hematologicalal malignancies.J Med Chem 2014;57:3666–77.

9. Ashkenazi A,Dixit VM.Death receptors: signaling andmodulation. Science1998;281:1305–8.

10. Chesi M, Mirza NN, Garbitt VM, Sharik ME, Dueck AC, Asmann YW, et al.IAP antagonists induce anti-tumor immunity in multiple myeloma.Nat Med 2016;22:1411–20.

11. Beug ST, Beauregard CE, Healy C, Sanda T, St-Jean M, Chabot J, et al. Smacmimetics synergize with immune checkpoint inhibitors to promotetumour immunity against glioblastoma. Nat Commun 2017;8.

12. MinDJ,He S, Green JE. Birinapant (TL32711) improves responses toGEM/AZD7762 combination therapy in triple-negative breast cancer cell lines.Anticancer Res 2016;36:2649–57.

Birinapant Enhances CAR T-cell Efficacy

www.aacrjournals.org Cancer Immunol Res; 7(2) February 2019 191

on December 23, 2020. © 2019 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst January 16, 2019; DOI: 10.1158/2326-6066.CIR-18-0428

Page 10: Antagonism of IAPs Enhances CAR T-cell Efficacy · L-glutamine, nonessential amino acids, 10 mmol/L 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid (HEPES) sodium pyruvate,and(Calbiochem)].Tcellswereactivatedwithanti-CD3

13. Krepler C, Chunduru SK, Halloran MB, He X, Xiao M, Vultur A, et al. Thenovel SMAC mimetic birinapant exhibits potent activity against humanmelanoma cells. Clin Cancer Res 2013;19:1784–94.

14. Mardiana S, John LB,HendersonMA, SlaneyCY, von Scheidt B, Giuffrida L,et al. A multifunctional role for adjuvant anti-4–1BB therapy in augment-ing antitumor response by chimeric antigen receptor T cells. Cancer Res2017;77:1296–309.

15. Kearney CJ, Lalaoui N, Freeman AJ, Ramsbottom KM, Silke J, Oliaro J. PD-L1 and IAPs co-operate to protect tumors from cytotoxic lymphocyte-derived TNF. Cell Death Differ 2017;24:1705–16.

16. Beavis PA, Henderson MA, Giuffrida L, Mills JK, Sek K, Cross RS, et al.Targeting the adenosine 2A receptor enhances chimeric antigen receptorT cell efficacy. J Clin Invest 2017;127:929–41.

17. Darcy PK, Haynes NM, Snook MB, Trapani JA, Cerruti L, Jane SM, et al.Redirected perforin-dependent lysis of colon carcinoma by ex vivo genet-ically engineerd CTL. J Immunol 2000;164:3705–12.

18. Haynes NM, Trapani JA, Teng MW, Jackson JT, Cerruti L, Jane SM, et al.Single chain antigen recognition receptors that costimulate potent rejec-tion of established experimental tumours. Blood 2002;100:3155–63.

19. Wang LX, Westwood JA, Moeller M, Duong CP, Wei WZ, Malaterre J, et al.Tumor ablation by gene modified T cells in the absence of autoimmunity.Cancer Res 2010;70:9591–8.

20. House IG, Thia K, Brennan AJ, Tothill R, Dobrovic A, Yeh WZ, et al.Heterozygosity for the common perforin mutation, p.A91V, impairs thecytotoxicity of primary natural killer cells from healthy individuals.Immunol Cell Biol 2015;93:575–80.

21. Kershaw MH, Jackson JT, Haynes NM, Teng MW, Moeller M, Hayakawa Y,et al. Gene-engineered T cells as a superior adjuvant therapy for metastaticcancer. J Immunol 2004;173:2143–50.

22. DouganM, Dougan S, Slisz J, Firestone B, VannemanM, Draganov D, et al.IAP inhibitors enhance co-stimulation to promote tumor immunity.J Exp Med 2010;207:2195–206.

23. Ahmed N, Brawley VS, Hegde M, Robertson C, Ghazi A, Gerken C, et al.Human epidermal growth factor receptor 2 (HER2)-specific chimericantigen receptor-modified T cells for the immunotherapy ofHER2-positivesarcoma. J Clin Oncol 2015;33:1688–96.

24. Louis CU, Savoldo B, Dotti G, Pule M, Yvon E, Myers GD, et al.Antitumor activity and long-term fate of chimeric antigen receptor-positive T cells in patients with neuroblastoma. Blood 2011;118:6050–6.

25. Noonan AM, Bunch KP, Chen JQ, HerrmannMA, Lee JM, Kohn EC, et al.Pharmacodynamic markers and clinical results from the phase 2 studyof the SMAC mimetic birinapant in women with relapsed platinum-resistant or -refractory epithelial ovarian cancer. Cancer 2016;122:588–97.

26. Amaravadi RK, Schilder RJ, Martin LP, Levin M, Graham MA, Weng DE,et al. A Phase I study of the SMAC-mimetic birinapant in adultswith refractory solid tumors or lymphoma. Mol Cancer Ther 2015;14:2569–75.

27. Deveraux QL, Reed JC. IAP family proteins—suppressors of apoptosis.Genes Dev 1999;13:239–52.

28. Hong X, Lei L, Glas R. Tumors acquire inhibitor of apoptosis protein (IAP)-mediated apoptosis resistance through altered specificity of cytosolicproteolysis. J Exp Med 2003;197:1731–43.

29. Petersen SL, Wang L, Yalcin-Chin A, Li L, Peyton M, Minna J, et al.Autocrine TNFalpha signaling renders human cancer cells suscepti-ble to Smac-mimetic-induced apoptosis. Cancer Cell 2007;12:445–56.

Cancer Immunol Res; 7(2) February 2019 Cancer Immunology Research192

Michie et al.

on December 23, 2020. © 2019 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst January 16, 2019; DOI: 10.1158/2326-6066.CIR-18-0428

Page 11: Antagonism of IAPs Enhances CAR T-cell Efficacy · L-glutamine, nonessential amino acids, 10 mmol/L 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid (HEPES) sodium pyruvate,and(Calbiochem)].Tcellswereactivatedwithanti-CD3

2019;7:183-192. Published OnlineFirst January 16, 2019.Cancer Immunol Res   Jessica Michie, Paul A. Beavis, Andrew J. Freeman, et al.   Antagonism of IAPs Enhances CAR T-cell Efficacy

  Updated version

  10.1158/2326-6066.CIR-18-0428doi:

Access the most recent version of this article at:

  Material

Supplementary

  http://cancerimmunolres.aacrjournals.org/content/suppl/2019/01/16/2326-6066.CIR-18-0428.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://cancerimmunolres.aacrjournals.org/content/7/2/183.full#ref-list-1

This article cites 27 articles, 16 of which you can access for free at:

  Citing articles

  http://cancerimmunolres.aacrjournals.org/content/7/2/183.full#related-urls

This article has been cited by 2 HighWire-hosted articles. Access the articles 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

  Permissions

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

.http://cancerimmunolres.aacrjournals.org/content/7/2/183To request permission to re-use all or part of this article, use this link

on December 23, 2020. © 2019 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst January 16, 2019; DOI: 10.1158/2326-6066.CIR-18-0428


Recommended