15
Translational Science Inhibition of Ataxia-Telangiectasia Mutated and RAD3-Related (ATR) Overcomes Oxaliplatin Resistance and Promotes Antitumor Immunity in Colorectal Cancer Eve Comb es 1,2 , Augusto F. Andrade 1,2 , Diego Tosi 1,2 , Henri-Alexandre Michaud 1,2 , Flavie Coquel 3 , Veronique Garambois 1,2 , Delphine Desigaud 1,2 , Marta Jarlier 2 , Arnaud Coquelle 1,2 , Philippe Pasero 3 , Nathalie Bonnefoy 1,2 , Jerome Moreaux 3 , Pierre Martineau 1,2 , Maguy Del Rio 1,2 , Roderick L. Beijersbergen 4 , Nadia Vezzio-Vie 1,2 , and Celine Gongora 1,2 Abstract © 2019 American Association for Cancer Research ICD signals ATP HMGB1 CRT Immune cell death IFN production Antitumoral immunity Oxaliplatin + ATRi Cell autonomous cell death Cytoplasmic DNA Proliferation inhibition Apoptosis induction DNA damage Replication stress Although many patients with colorectal cancer initially respond to the chemotherapeutic agent oxa- liplatin, acquired resistance to this treatment remains a major chal- lenge to the long-term management of this disease. To identify molecu- lar targets of oxaliplatin resistance in colorectal cancer, we performed an shRNA-based loss-of-function genetic screen using a kinome library. We found that silencing of ataxia-telangiectasia mutated and RAD3-related (ATR), a serine/thre- onine protein kinase involved in the response to DNA stress, restored oxaliplatin sensitivity in a cellular model of oxaliplatin resistance. Combined application of the ATR inhibitor VE-822 and oxaliplatin resulted in strong synergistic effects in six different colorectal cancer cell lines and their oxaliplatin-resistant subclones, promoted DNA single- and double-strand break formation, growth arrest, and apoptosis. This treatment also increased replicative stress, cytoplasmic DNA, and signals related to immunogenic cell death such as calreticulin exposure and HMGB1 and ATP release. In a syngeneic colorectal cancer mouse model, combined administration of VE-822 and oxaliplatin signicantly increased survival by promoting antitumor T-cell responses. Finally, a DNA repair gene signature discriminated sensitive from drug-resistant patients with colorectal cancer. Overall, our results highlight the potential of ATR inhibition combined with oxaliplatin to sensitize cells to chemotherapy as a therapeutic option for patients with colorectal cancer. Signicance: These ndings demonstrate that resistance to oxaliplatin in colorectal cancer cells can be overcome with inhibitors of ATR and that combined treatment with both agents exerts synergistic antitumor effects. Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/79/11/2933/F1.large.jpg. 1 Institut de Recherche en Canc erologie de Montpellier, INSERM U1194 Universit e Montpellier, CNRS, France. 2 Institut r egional du CancerMontpellierVal d'Aurelle, Montpellier, France. 3 IGH UMR9002, CNRS-Universit e de Montpellier, Montpellier, France. 4 Division of Molecular Carcinogenesis and NKI Robotics and Screening Center, The Netherlands Cancer Institute, Amsterdam, the Netherlands. Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/). N. Vezzio-Vie and C. Gongora contributed equally to this article. E. Comb es and A.F. Andrade are the co-rst authors of this article. Corresponding Author: Celine Gongora, IRCM INSERM U1194, 208 rue des Apothicaires, Montpellier 34298, France. Phone: 334-6761-3745; Fax: 334- 6761-3787; E-mail: [email protected] Cancer Res 2019;79:293346 doi: 10.1158/0008-5472.CAN-18-2807 Ó2019 American Association for Cancer Research. Cancer Research www.aacrjournals.org 2933 on January 14, 2021. © 2019 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from Published OnlineFirst April 15, 2019; DOI: 10.1158/0008-5472.CAN-18-2807

Inhibition of Ataxia-Telangiectasia Mutated and RAD3 ... · Apothicaires, Montpellier 34298, France. Phone: 334-6761-3745; Fax: 334-6761-3787; E-mail: [email protected] Cancer

  • Upload
    others

  • View
    2

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Inhibition of Ataxia-Telangiectasia Mutated and RAD3 ... · Apothicaires, Montpellier 34298, France. Phone: 334-6761-3745; Fax: 334-6761-3787; E-mail: celine.gongora@inserm.fr Cancer

Translational Science

Inhibition of Ataxia-Telangiectasia Mutated andRAD3-Related (ATR) Overcomes OxaliplatinResistance and Promotes Antitumor Immunityin Colorectal CancerEve Comb�es1,2, Augusto F. Andrade1,2, Diego Tosi1,2, Henri-Alexandre Michaud1,2,Flavie Coquel3, Veronique Garambois1,2, Delphine Desigaud1,2, Marta Jarlier2,Arnaud Coquelle1,2, Philippe Pasero3, Nathalie Bonnefoy1,2, Jerome Moreaux3,Pierre Martineau1,2, Maguy Del Rio1,2, Roderick L. Beijersbergen4, Nadia Vezzio-Vie1,2,and Celine Gongora1,2

Abstract

© 2019 American Association for Cancer Research

ICD signals

ATP

HMGB1

CRTImmune cell death

IFN production

Antitumoralimmunity

Oxaliplatin + ATRi

Cell autonomous cell death

CytoplasmicDNA

Proliferation inhibitionApoptosis induction

DNA damageReplication stress

Although many patients withcolorectal cancer initially respondto the chemotherapeutic agent oxa-liplatin, acquired resistance to thistreatment remains a major chal-lenge to the long-termmanagementof this disease. To identify molecu-lar targets of oxaliplatin resistancein colorectal cancer, we performedan shRNA-based loss-of-functiongenetic screen using a kinomelibrary. We found that silencing ofataxia-telangiectasia mutated andRAD3-related (ATR), a serine/thre-onineprotein kinase involved in theresponse to DNA stress, restoredoxaliplatin sensitivity in a cellularmodel of oxaliplatin resistance.Combined application of the ATRinhibitor VE-822 and oxaliplatinresulted in strong synergistic effects in six different colorectal cancer cell lines and their oxaliplatin-resistant subclones,promoted DNA single- and double-strand break formation, growth arrest, and apoptosis. This treatment also increasedreplicative stress, cytoplasmic DNA, and signals related to immunogenic cell death such as calreticulin exposure and HMGB1and ATP release. In a syngeneic colorectal cancer mouse model, combined administration of VE-822 and oxaliplatinsignificantly increased survival by promoting antitumor T-cell responses. Finally, a DNA repair gene signature discriminatedsensitive from drug-resistant patients with colorectal cancer. Overall, our results highlight the potential of ATR inhibitioncombined with oxaliplatin to sensitize cells to chemotherapy as a therapeutic option for patients with colorectal cancer.

Significance: These findings demonstrate that resistance to oxaliplatin in colorectal cancer cells can be overcome withinhibitors of ATR and that combined treatment with both agents exerts synergistic antitumor effects.

Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/79/11/2933/F1.large.jpg.

1Institut de Recherche en Canc�erologie de Montpellier, INSERM U1194 Universit�eMontpellier, CNRS, France. 2Institut r�egionalduCancer–Montpellier–Vald'Aurelle,Montpellier, France. 3IGHUMR9002, CNRS-Universit�e deMontpellier, Montpellier,France. 4Division of Molecular Carcinogenesis and NKI Robotics and ScreeningCenter, The Netherlands Cancer Institute, Amsterdam, the Netherlands.

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

N. Vezzio-Vie and C. Gongora contributed equally to this article.

E. Comb�es and A.F. Andrade are the co-first authors of this article.

Corresponding Author: Celine Gongora, IRCM INSERM U1194, 208 rue desApothicaires, Montpellier 34298, France. Phone: 334-6761-3745; Fax: 334-6761-3787; E-mail: [email protected]

Cancer Res 2019;79:2933–46

doi: 10.1158/0008-5472.CAN-18-2807

�2019 American Association for Cancer Research.

CancerResearch

www.aacrjournals.org 2933

on January 14, 2021. © 2019 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst April 15, 2019; DOI: 10.1158/0008-5472.CAN-18-2807

Page 2: Inhibition of Ataxia-Telangiectasia Mutated and RAD3 ... · Apothicaires, Montpellier 34298, France. Phone: 334-6761-3745; Fax: 334-6761-3787; E-mail: celine.gongora@inserm.fr Cancer

IntroductionColorectal cancer is the fourth most common cancer world-

wide. In 2015, the estimated colorectal cancer incidence was of1.65 million cases worldwide, and colorectal cancer caused morethan 835, 000 deaths (1). Importantly, 30% of patients presentsynchronous metastases and 50%–60% will develop metastasesthat will require chemotherapy. The current management ofcolorectal cancer is based on various drugs [5-fluorouracil (5-FU)/leucovorin (LV), capecitabine, irinotecan, oxaliplatin, bev-acizumab, cetuximab, and panitumumab], either in combinationor as single agents (2). These treatments significantly improvedthe patients' overall survival, though tumor resistance is still afrequent cause of therapy failure.

Among the drugs used in colorectal cancer treatment, oxali-platin is a third-generation platinum compound with a 1,2-diaminocyclohexane carrier ligand, and is used in combinationwith 5-fluorouracil and leucovorin (FOLFOX regimen). It causesinter- and intrastrand DNA cross-links that stop DNA replicationand transcription, leading to apoptotic cell death (3). Oxaliplatinexerts its antitumor effect also by inducing immunogenic celldeath (ICD; ref. 4), where the host immune system is primed torecognize and eliminate tumor cells upon drug-mediated stimu-lationof the host antitumor immunity. Resistance to oxaliplatin ismediated by several factors, such as reduced cellular uptake,impaired DNA adduct formation, alterations in DNA repair genes(e.g.,ERCC1 andXRCC1), apoptosis defects, andmodifications inthe expression levels of copper transporters (ATP7A and ATP7B;refs. 3–7).

Drug combinations are often used to overcome drug resis-tance and a major effort is made to identify novel drug combi-nations to improve therapeutic efficiency. High-throughputfunctional genetic screens (using shRNA or CRISPR/Cas9approaches) provide a powerful tool to identify not only themechanisms of drug resistance, but also new targets for efficientdrug combinations. In this study, we wanted to identify mole-cules that could modify molecular alterations that occur inoxaliplatin-resistant colorectal cancer cell lines. To this aim, weperformed a kinome-specific shRNA genetic screen showingthat ataxia-telangiectasia mutated and RAD3-related (ATR) isimplicated in oxaliplatin resistance. Importantly, we coulddemonstrate that the combination of an ATR inhibitor withoxaliplatin is synergistic in in vitro and in vivo colorectal cancermodels by inducing DNA double-strand breaks, leading toapoptosis, and promoting antitumor immunity.

Materials and MethodsCell culture

The human HCT116, SW48, SW480, SW620, HT29 (from theATCC and the murine CT26, kindly provided by N. Bonnefoy,colorectal cancer cell lines were grown in RPMI1640 with 10%FCS and 2 mmol/L L-glutamine at 37�C in a humidified atmo-spherewith5%CO2.Themurine colorectal cancer cell lineMC38,providedbyN.Bonnefoy,was grown inDMEMwith10%FCSand1 mmol/L sodium pyruvate at 37�C in a humidified atmospherewith 5% CO2. The oxaliplatin-resistant clones (HCT116-R1,HCT116-R2, SW48-R) were obtained as described previously (5).Briefly, the oxaliplatin-sensitive parental HCT116 and SW48 celllines were grown in the presence of 5–10 mmol/L oxaliplatin andcloned to obtain the resistant HCT116-R1 and HCT116-R2 and

SW48-R clones, respectively. The cell lineswere tested and authen-ticated by short-tandem repeat profiling (LGC Standards andEurofins Genomics). All experiments were performed at leastthree times.

Drugs and reagentsVE-822 (ATR inhibitor) was kindly provided by A. Coquelle

and dissolved to a final concentration of 50 mmol/L in DMSO(Sigma Chemical Co.). Oxaliplatin (5 mg/mL) was from ICM(oxaliplatin ACCORD Cip: 3400957954642) and was dilutedin PBS to the final concentration of 12 mmol/L. Both stocksolutions were aliquoted and stored at �20�C. Poly-L-ornithinewas purchased from Sigma and formaldehyde (16%) fromElectron Microscopy Sciences. Antibodies against ATR#13934, ATM #2873, phosphorylated ATM (Ser1981)#13050, CDK2 #2546, phosphorylated CDK2 #2561, CHK12360#, phosphorylated CHK1 (Ser345) #2341, CHK2 #2662,phosphorylated CHK2 (Thr68) #2661, GAPDH #5174, p21WAF1/CIP1 #2947, and phosphorylated p53 (Ser15) #9284were from Cell Signaling Technology. The anti-phosphorylatedATR (Thr1989) GTX128145 antibody was from Genetex, anti–b-tubulin T4026 from Sigma, and anti-p53 (DO-1) sc-126 fromSanta Cruz Biotechnology.

Pooled shRNA "dropout" screenThe TRC kinome library included 1,798 short-hairpin RNA

vectors that target 518 genes selected from TRC collection. Thelibrary was used to generate lentiviral supernatants as describedpreviously (8). HCT-116-R1 cells were infected with the lentiviralpools at a multiplicity of infection < 0.3 and with cell numberssufficient to represent the kinome library with a 1,000 timescoverage for each shRNA present in the library. After puromycinselection, infected cells were pooled and plated in 15-cm dishes(1 � 105 cells/dish) with sufficient cell numbers to maintain the1,000-fold coverage. After incubation in the absence or presenceof 3 mmol/L oxaliplatin for 10 days, cells were harvested, genomicDNA was isolated, and used for recovery of the shRNA inserts byPCR. Indexes and adaptors for deep sequencing (Illumina) wereincorporated in the PCR primers. PCR products were purifiedusing the QIAGEN PCR Purification Kit, according to the man-ufacturer's protocol. DNA was quantified using a BioAnalyzer,and samples were combined at the same molar ratio. The shRNAsequences were extracted from the sequencing reads and alignedto the kinome library. The matched reads were counted and theread counts were used for analysis. The statistical analysis wasdone with DESeq, version 1.8.3, using the default settings for"pooled." The results of the DESeq analysis were used to calculatethe ratio between treated and nontreated cells for each individualshRNA.

ATR silencing by shRNAsFour different shRNA vectors with the puromycin selection

marker and specific to ATR were used. Lentiviral production wasperformed by cotransfecting 293T cells (for lentiviral packaging)with 1 mg of anti-ATR shRNAs, 1 mg of the packaging vector gag-pol, and 1 mg of envelope vector, using FuGene6 (Invitrogen),according to the manufacturer's instructions. Lentiviral particleswere harvested and then used to infect 1 � 106 HCT116 andHCT116-R1 cells for 48 hours. Following shRNA transduction,cells were selected with puromycin.

Comb�es et al.

Cancer Res; 79(11) June 1, 2019 Cancer Research2934

on January 14, 2021. © 2019 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst April 15, 2019; DOI: 10.1158/0008-5472.CAN-18-2807

Page 3: Inhibition of Ataxia-Telangiectasia Mutated and RAD3 ... · Apothicaires, Montpellier 34298, France. Phone: 334-6761-3745; Fax: 334-6761-3787; E-mail: celine.gongora@inserm.fr Cancer

Cell growth inhibition assay (2D assay)Cell growth was evaluated using the sulforhodamine B (SRB)

assay, as described by Skehan and colleagues (9). Briefly, 300–1,000 cells/well were seeded in 96-well plates. After 24 hours,drugs were added in serial dilution. Cells were incubated for 96hours, after which, they were fixed in trichloroacetic acid solution(final concentration of 10%), and stainedwith 0.4%SRB solutionin 1% acetic acid (Sigma Chemical Co.). Fixed SRB was dissolvedin 10 mmol/L Tris-HCl solution and absorbance at 560 nm wasread using a Thermo Fisher Scientific Multiskan EX plate reader.The IC50 was determined graphically from the cell growth curves.

3D spheroid assayFor spheroid generation, 100 mL/well of cell suspensions at

optimized densities (50–300 cells/well) were dispensed in ultra-low attachment 96-well round-bottomed plates (Corning B.V.Life Sciences) and cultured at 37�C, 5%CO2, 95%humidity, for 4days. Depending on the cell sensitivity, spheroids were incubatedwith VE-822 (0 to 2.5 mmol/L) and/or oxaliplatin (0–30 mmol/L)at day 4 and at day 10 after plating.

At day 14, images were captured with the Celigo ImagingCytometer (Nexcelom Bioscience) using the "Tumorosphere"application. Cell viability was measured using a CellTiter-GloLuminescent Cell Viability Assay (Promega), according to themanufacturer's instructions. Luminescence was measured on a1450 MicroBeta TriLux Luminescence Microplate Reader (PerkinElmer). The IC50was determined graphically from the cytotoxicitycurves.

Quantification of the interaction effectThe interaction between the drugs tested in vitro was investi-

gated with a concentration matrix test, in which increasing con-centration of each single drug were assessed with all possiblecombinations of the other drugs. For each combination, thepercentage of expected growing cells in the case of effect inde-pendence was calculated according to the Bliss equation (10):

fuc ¼ fuAfuB

where fuc is the expected fraction of cells unaffected by the drugcombination in the case of effect independence, and fuA and fuBare the fractions of cells unaffected by treatment A and B, respec-tively. The difference between the fuc value and the fraction ofliving cells in the cytotoxicity test was considered as an estimationof the interaction effect, with positive values indicating synergismand negative values antagonism.

EdU assayCell proliferation was measured with the Click-iT EdU Assay

(Invitrogen) according to the manufacturer's protocol. Briefly,2.5 � 105 HCT116 and 4 � 105 HCT-116-R1 cells were plated in25 cm2

flasks, and 24 hours later they were incubated withoxaliplatin and/or VE-822 for 48 hours. Twenty hours beforeharvesting, EdU was also added. Cells were then washed in ice-cold PBS, fixed in 75% ethanol, and then incubated with theClick-iT reaction cocktail for 30 minutes. Cells were washed withPBS and then analyzed by flow cytometry.

Cell-cycle analysisTo determine the cell-cycle distribution, 2.5� 105HCT116 and

4 � 105 HCT-116-R1 cells were plated in 25 cm2flasks, and

24 hours later they were incubated with oxaliplatin (5 mmol/L)and/or VE-822 (1 mmol/L). After 48 hours, cells were washed inice-cold PBS, fixed in 75% ethanol, and labeled with 40 mg/mLpropidium iodide (Sigma) containing 100 mg/mL RNase A(Sigma). Cell-cycle distribution was then determined with aGallios Cytometer (Beckman Coulter) and quantified using theKaluza Software (Beckman Coulter).

Apoptosis assayA total of 2.5 � 105 HCT116 and 4 � 105 HCT-116-R1 cells

were plated, and after 24 hours, they were incubated withthe indicated drugs (alone or in combination) for 48 hours.Cells were stained with FITC-labeled Annexin V and 7-amino-actinomycin D (7-AAD; Annexin V/7-AAD-Beckman Coulter).For apoptosis determination, Annexin V- and 7-ADD–positivecells were quantified using a Gallios Cytometer and the KaluzaSoftware (Beckman Coulter).

Western blot analysisAfter treatment, cells (1� 104 cells/mL) were washed (1� PBS)

and directly lysed in Laemmli buffer (4% SDS, 20% glycerol, 1%2-b mercaptoethanol, 0.004% bromophenol blue, 0.125 mol/LTris HCL)þ benzonase. After denaturation at 95�C for 5minutes,protein extracts were deposited and separated on SDS-PAGEpolyacrylamide (10%, 12%, or 4%–20% gradient) gels. Theywere then transferred (transfer buffer containing 20% ethanol)to nitrocellulose membranes (10% and 12% gels: semi-dry trans-fer for 1 hour; 4%–20% gradient gels: liquid transfer for 2 hours).Membranes were then blocked in PBS/0.1% Tween/5% milk atroom temperature for 1 hour under agitation. They were thenincubated with the primary antibodies at 4�C by gentle agitationovernight. After three washes in PBS/0.1% Tween, membraneswere incubated with the relevant antispecies secondary antibodycoupled to horseradish peroxidase diluted to 1/5,000 in PBS/0.1% Tween/5% milk with gentle agitation for 1 hour. Afterwashing, immunoreactions were revealed by chemiluminescence(ECL RevelBlot Plus, and ECL RevelBlot Intense Kits) and visu-alized using the G-Box Imaging System (Syngene). Western blotanalysis was performed using ImageJ software, and quantificationwere calculated relatively to the loading control associated. Theuntreated condition was arbitrary fixed at 1 to easily observe thechanges inducedby transfections or treatments. EachWestern blotwas performed andquantifiedbetween twoand four times in eachcell line except for ATM total in HCT116 cells, for which quan-tification could only be done once.

RPA32 and gH2AX analysisHCT-116 (4,000 to 9,000 cells/well) or HCT116-R1 (8,000 to

16,000 cells/well) cells were plated in black-sided 96-well, flat-bottomed plates (Greiner) precoated with poly-L-ornithine. After24 hours, cells were incubated with VE-822 (1 mmol/L) and/oroxaliplatin (12.5 or 50 mmol/L) for 3, 6, 16, and 24 hours. Then,RPA not bound to chromatin was extracted with PBS/0.2% TritonX-100 on ice for 90 seconds. All steps were carried out carefully tokeep cells attached to the plate. Between each step, cells werewashed with PBS containing 1 mg/mL BSA. First, cells were fixedin PBS/4% paraformaldehyde (PFA) for 30 minutes and permea-bilized with PBS/0.2% Triton X-100 at room temperature for10 minutes. After blocking with PBS/1% BSA for 30 minutes,primary antibodies against RPA32 (Abcam, ab2175) and gH2AX(Cell Signaling Technology, #9718) were diluted in PBS with

Inhibition of ATR Overcomes Resistance to Oxaliplatin in CRC

www.aacrjournals.org Cancer Res; 79(11) June 1, 2019 2935

on January 14, 2021. © 2019 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst April 15, 2019; DOI: 10.1158/0008-5472.CAN-18-2807

Page 4: Inhibition of Ataxia-Telangiectasia Mutated and RAD3 ... · Apothicaires, Montpellier 34298, France. Phone: 334-6761-3745; Fax: 334-6761-3787; E-mail: celine.gongora@inserm.fr Cancer

1 mg/mL BSA and added to each well at room temperature for1 hour. Then, the secondary antibodies (anti-mouse A448,Invitrogen, A11029; and anti-rabbit A568, Thermo Fisher Scien-tific, A11011), diluted in PBS/1 mg/mL BSA, were added atroom temperature for 45 minutes. After this step, cells were keptin the dark and incubated with 1 mg/mL DAPI at 37�C for30 minutes. Cells were then washed and left in PBS for analysiswith the Celigo cytometer imaging system.

DNA fiber spreadingHCT116-R1 cells were seeded in 6-well plates (0.5 � 10�6/

well). After 1 day, cells were incubated with VE-822 (1 mmol/L)and/or oxaliplatin (12.5 mmol/L) for 24 hours. Then, cells werelabeled by two consecutive pulses of 15 minutes/each of thethymidine analogues iodo-deoxyuridine (IdU; final concentra-tion: 20 mmol/L) and chloro-deoxyuridine (CldU; final concen-tration: 200 mmol/L). Then, the medium was removed andreplaced by fresh medium containing 400 mmol/L of thymidinefor 2 hours (chase period). After this step, cells were trypsinized,washed, and diluted in cold PBS to 1 � 10�6 cells.

In a humid chamber, the DNA of approximately 2,000 cellsfrom the cell suspension was spread along a microscopy slideusing lysing buffer (200mmol/L Tris-HCL, 50mmol/L EDTA, 0.5%SDS in milliQ H2O), and fixed in acetic acid:methanol (1:3) for10 minutes, after which, slides were dried.

For immunostaining, slides were washed with H2O (2 � 5minutes) and denatured in 2.5 mol/L HCL for 1 hour. Afterwashing in PBS, slides were saturated in PBS/5% BSA for 1 hour,and then incubated with the anti-Cldu antibody (Bio-RadOBT0030) in PBS/0.1% Triton X-100 (PBST) at 37�C for 1 hour.After washes in PBST (2� 5minutes) and in PBS (1� 5minutes),slides were incubated with the secondary antibody (ThermoFisher Scientific A11006) in PBST at 37�C for 45 minutes (in thedark). After washing, slides were incubated with anti-IdU (BDBiosciences 347580) and anti-ssDNA (DHSB supernatant) anti-bodies in PBST at 37�C for 1hour,washed, and incubatedwith therelevant secondary antibody (Thermo Fisher Scientific A21123and A21241) in PBST at 37�C for 45 minutes. Then, slides werewashed, air-dried, and mounted with Prolong Gold (ThermoFisher Scientific P10144). Micrographs of labeled fibers weretaken with a Fluorescence Microscope (Zeiss or Leica DMRM)and for each condition, at least 100 track lengths were measured.Each experiment was repeated at least two or three times.

Calreticulin exposureA total of 1.5–2 � 105 cells were plated in 6-well plates, and

after 24 hours, themediumwas changed and cells were incubatedwith oxaliplatin (25 mmol/L) and/or VE-822 (1 mmol/L) for 48hours. Cells were collected, washed in cold PBS/10% FBS, andstained with 7-AAD (Beckman Coulter) for 30 minutes. Afterwashing in PBS/10% FBS, cells were fixed with 0.25% PFA for 5minutes. After washing, cells were incubated with the anti–CRT-Alexa647 antibody (ab196159, Abcam) for 1 hour, followed bywashing, andfixation in 1%PFA for 5minutes. Calreticulin (CRT)exposure was assessed using a Gallios Cytometer (BeckmanCoulter). The fluorescent intensity of CRT-positive cells was gatedrelative to 7-AAD–negative cells.

ATP and HMGB1 releaseATP and HMGB1 release in the cell supernatant were analyzed

after incubation with oxaliplatin (12.5–100 mmol/L) and/or VE-

822 (1 mmol/L) for 24 hours and 48 hours. Supernatants werecollected and centrifuged to remove dying cells. ATP releasewas measured with the ATPlite Luminescence Assay System(PerkinElmer), according to the manufacturer's instructions.HMGB1 release was quantified with the HMGB1 ELISA Kit (IBLInternational), according to the manufacturer's instructions.

Cytosolic DNACells grown on glass coverslips were fixed in 80% ice-cold

methanol for 20 minutes. Control samples were preincubated at37�Cwith 200mg/mLRNase A (Sigma) and/orwithDNase for 20minutes. Samples were blocked with 1% BSA/PBS/Triton X-100for 1 hour, and then incubated with the primary antibodiesovernight [anti-ssDNA (Developmental Studies HybridomaBank, The University of Iowa, Iowa City, IA) or anti-dsDNA(MAB1293, Millipore) in PBS/0.1% Tween]. After removal ofunbound primary antibodies by washes with PBS/0.1% Tween,secondary antibodies were added at room temperature for 1 hour.Coverslips were then washed in PBS/0.1% Tween before mount-ing with DAPI-containing Prolong Gold. Images were taken usinga 63� objective and a Zeiss microscope.

In vivo studiesXenografts. HCT116/R1 and MC38 tumor cells (2 � 106 and 5 �105, respectively) were injected subcutaneously in the left flank of8-week-old female athymic nu/nu and C57BL/6 mice (CharlesRiver Laboratories; n ¼ 6–8). Tumors were detected by palpationandmeasured with a calliper weekly. Mice were euthanized whenthe tumor volume reached 1,500 mm3. Ethical approvals wereobtained by the local ethics committee [Ethics Committeeapproved by the French Ministry, animal facility approval C34-172-27, personal approval (C�elineGongora) 34.142andprotocolapproval CEEA-LR- 1193 and CEEA-LR-1194)].

Tumor treatment.When tumors reached the volume of 100mm3,mice were treated for 4 weeks with: (i) 0.2 mL of 0.9% sodiumchloride solution by intraperitoneal injection þ D-a-tocopherolpolyethylene glycol succinate (TPGS; gavage; control group),twice per week; (ii) 5 mg/kg oxaliplatin by intraperitoneal injec-tion once per week; (iii) 30mg/kg VE-822 (dissolved in TPGS) bygavage twice perweek; or (iv) 5mg/kg oxaliplatin once perweekþ30 mg/kg VE-822 twice per week.

Statistical analysis. A linear mixed-regression model, containingboth fixed and random effects, was used to determine the rela-tionship between tumor growth and days after grafting. Datawerefirst transformed using the natural log scale to better fit theassumptions of the linear mixed model. The fixed part of themodel included variables corresponding to the number of post-graft days and the different treatments. Interaction terms werebuilt into the model; random intercepts and random slopes wereincluded to take time into account. The model coefficients wereestimated by maximum likelihood and considered significant atthe 0.05 level. Statistical analyseswere performedusing the STATA10.0 Software (StataCorp).

Detection of CD8-positive T cellsTreated and control mice harboring mouse MC38 cell xeno-

grafts were sacrificed 3 weeks after graft/treatment. Spleens wererecovered in ice-cold PBS containing 0.5% BSA and 2 mmol/L

Comb�es et al.

Cancer Res; 79(11) June 1, 2019 Cancer Research2936

on January 14, 2021. © 2019 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst April 15, 2019; DOI: 10.1158/0008-5472.CAN-18-2807

Page 5: Inhibition of Ataxia-Telangiectasia Mutated and RAD3 ... · Apothicaires, Montpellier 34298, France. Phone: 334-6761-3745; Fax: 334-6761-3787; E-mail: celine.gongora@inserm.fr Cancer

EDTA, and were mechanically dissociated. Red blood cells werethen removed by adding 2 mL ammonium-chloride-potassiumlysing buffer. White blood (effector cells) cells were recovered bycentrifugation, washed with PBS, and resuspended in RPMI1640plus 10% FBS. MC38 target cells were cultured as describedpreviously, and then trypsinized, washed, and resuspended inRPMI1640 plus 10% FBS. A total of 10.5 cells were plated in100 mL of RPMI1640 in 96-well U-shaped plates, and thenirradiated (50 Gy). Splenocytes were added at the 1:5 (target:effector) ratio and cultured overnight. GolgiPlug and GolgiStopwere added to each well to block cytokine secretion (according tothe manufacturer's protocol; BD Biosciences) for 6 hours. Toidentify IFNg-secreting CD8-positive T cells, cells were washedonce with PBS and incubated with antibodies against surfacemarkers CD45-BV786 (clone 30F11), CD8-BV605 (clone 53-6.7),and the LIVE/DEADFixable AquaDeadCell Stain Kit (Invitrogen)at 4�C for 1 hour. After extracellular staining, cells were fixed andpermeabilized, according to the eBioscience fixation and permea-bilization procedures, and intracellular staining [(anti-IFNg anti-body V421 (clone XMG1.2)] was performed at 4�C overnight.Then, samples were washed, fixed in 1% PFA, and processed fordata acquisition and analysis using a Cytoflex Flow Cytometer(Beckman Coulter) and the Flowjo 10 software.

Gene expression profiling and DNA repair scoreATR expression was assessed using the dataset GSE62322

[(n ¼ 17 normal mucosa, n ¼ 20 primary tumor, and n ¼ 19hepatic metastasis tissue samples from patients with metastaticcolorectal cancer (mCRC)] from the prospective single-centerstudy REGP (9, 10).

To build the DNA repair score, the gene expression datasetsfrom two cohorts were used. The Tsuji dataset (GSE28702)included 80 patients with stage IV colorectal cancer treated withFOLFOX. Shingo Tsuji (Genome Science Division, ResearchCenter for Advanced Science and Technology, The University ofTokyo, Tokyo, Japan) kindly provided the OS data. The Del Riodataset (GSE72970) included 36 patients with stage IV colorectalcancer treated with FOLFOX and also the clinical data (11). Geneexpression data were normalized with the MAS5 algorithm andanalyzed with GenomicScape (http://www.genomicscape.com;ref. 12) and R.2.10.1 and Bioconductor, version 2.5. A consensuslist of genes encoding proteins involved in the DNA repairpathways was obtained using the REPAIRtoire database (http://repairtoire.genesilico.pl; ref. 13) and by reviewing Medline, asdescribed previously (14). The DNA repair score was built usingour previously published methodologies to develop prognosticscores using sets of prognostic genes (14–17). The DNA repairscore was the sum of the Cox beta coefficients of each of the12 DNA repair genes with a prognostic value, weighted by �1 ifthe patient tumor signal for a given gene was above or belowthe probeset maxstat value for that gene (14–17).

ResultsA functional genetic screen reveals that ATR is implicated inoxaliplatin resistance in colorectal cancer cells

We assessed oxaliplatin sensitivity in five colorectal cancer celllines (HCT116, SW48, SW480, SW620, and HT29) and threeoxaliplatin-resistant derivatives (HCT116-R1, HCT116-R2, andSW48-R) that were established by long-term growth in the pres-ence of oxaliplatin (5). These cell lines display differentmolecular

characteristics in terms of MSI/MSS status, origin, and KRAS,BRAF, PIK3CA, and TP53 gene mutation status (SupplementaryTable S1). Using 2D (SRB) and 3D (spheroid growth) cell cultureassays, we confirmed that the three oxaliplatin-resistant cell linesdisplayed the highest oxaliplatin IC50 values (Fig. 1A).

To identify genes the inhibition of which confers sensitivity tooxaliplatin in HCT116-R1 cells (synthetic lethal interactions withoxaliplatin; ref. 8), we performed a loss-of-function genetic screenusing the TRC lentiviral kinome shRNA library that targets 518human kinases (Fig. 1B). After infection of HCT116-R1 cells withthe library and culture in the absence/presence of 3 mmol/Loxaliplatin (the oxaliplatin concentration required to kill allsensitive HCT116 cells, but inefficient in HCT116-R1 cells,see Fig. 1B) for 10 days. shRNAs were amplified by PCR and theirrelative abundance was determined by next-generation sequenc-ing using the barcode identifiers present in each shRNA vector.Weconsidered only shRNA vectors that were significantly depleted(by at least 50%) upon incubation with oxaliplatin, and selectedgenes represented by multiple shRNAs matching this criterion.Only few of the 1,798 shRNAs in the library met these criteria(Fig. 1C), among which, three (out of four) were independentshRNAs targeting ATR. To validate this finding, we infectedHCT116-R1 cells with shRNAs against ATR or luciferase(control; Fig. 1D) and confirmed that ATR silencing sensitizedHCT116-R1 cells to oxaliplatin (Fig. 1E; Supplementary Fig. S1Aand S1B). Moreover, comparison of the oxaliplatin IC50 values ofHCT116-R1-shATR and HCT116 cells indicated that upon ATRknockdown, resistant HCT116-R1 cells became as sensitive tooxaliplatin as HCT116 cells (Fig. 1F). These findings demonstratethat inhibition can be sufficient to overcome oxaliplatin resis-tance, at least in HCT116-R1 cells.

We then tested whether ATR or its target protein checkpointkinase 1 (CHK1) were constitutively activated (i.e., phosphory-lated) in HCT116-R1 cells compared with sensitive HCT116 cells.Western blot analysis showed that neither ATR nor CHK1 wereactivated in HCT116-R1 cells (Fig. 1G). In addition, analysis of atranscriptome dataset frompatients withmCRC (18) showed thatATR expression was significantly higher in primary tumors (n ¼20) andhepaticmetastasis (n¼19) than in normal tissue (n¼17)samples (Fig. 1H). This indicates that ATR is aberrantly expressedin colorectal cancer tumor samples and might be a potentialtherapeutic target.

The ATR inhibitor VE-822 and oxaliplatin combination issynergistic in colorectal cancer cells

Currently, two ATR inhibitors (ATRi) are clinically available(VX-970 and AZD-6738). As VX-970 (called VE-822 thereafter) ismore advanced in terms of clinical development, we used thisATRi. We evaluated the interaction between oxaliplatin and VE-822 in HCT116-R1 cells and in seven additional colorectal cancercell lines, using a full-range concentration matrix approach andthe SRB cytotoxicity assay (Fig. 2A). Specifically, we quantified thepercentage of cell growth (blue matrix) and the additive, syner-gistic and antagonistic effects (black, red, and green matrices,respectively). We observed a synergistic interaction between oxa-liplatin and VE-822 in all tested colorectal cancer cell lines, withthe highest values in HCT116-R1, HCT116-R2, and SW48-R cellsand the smallest for SW480. This indicates that VE-822 canefficiently overcome oxaliplatin resistance. The distribution ofthe interaction effects was not always homogeneous over theconcentration matrices. For instance, focal areas with higher

Inhibition of ATR Overcomes Resistance to Oxaliplatin in CRC

www.aacrjournals.org Cancer Res; 79(11) June 1, 2019 2937

on January 14, 2021. © 2019 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst April 15, 2019; DOI: 10.1158/0008-5472.CAN-18-2807

Page 6: Inhibition of Ataxia-Telangiectasia Mutated and RAD3 ... · Apothicaires, Montpellier 34298, France. Phone: 334-6761-3745; Fax: 334-6761-3787; E-mail: celine.gongora@inserm.fr Cancer

synergy were often located in concentration regions that corre-sponded to high VE-822 concentration.

Next, we assessed whether ATR inhibition influenced oxaliplatincytotoxic activity inmice xenografted subcutaneouslywithHCT116-R1 cells. To this aim, we treated mice with 0.9% sodium chloridesolution (control group), oxaliplatin, VE-822, or the oxaliplatin þVE-822 combination. Oxaliplatin and VE-822 alone had no effecton tumor growth comparedwith control. Conversely, the VE-822þoxaliplatin combination was effective in limiting tumor growth,even not really marked but significant (P ¼ 0.018; Fig. 2B) and

extending survival (Kaplan–Meier survival analysis, P ¼ 0.0216;Fig. 2C). This result confirms our previous in vitro findings, andprovides evidence that ATR inhibition also enhances oxaliplatintherapeutic effect in resistant colorectal cancer cells in vivo.

The VE-822 þ oxaliplatin combination induces apoptosis,reduces cell proliferation, and activates the ATM–CHK2pathway

We then investigated whether the ATRi VE-822 þ oxaliplatincombination had a synergistic effect also on cell death and/or cell

Figure 1.

Identification of the ATR gene using a synthetic lethality assay in oxaliplatin-resistant colon cancer cells. A, The colorectal cell lines HCT116 and its resistantclones (HCT116-R1 and HCT116-R2), SW48 and its resistant clone (SW48-R), SW480, SW620, and HT29 were incubated with increasing concentrations ofoxaliplatin (0–30 mmol/L). The IC50 values were obtained using the SRB and spheroid formation assays (2D and 3D cell culture assay, respectively). Data are themean� SD (error bars) of at least three independent experiments. B, Schematic workflow of the "dropout" shRNA screen to identify genes, the inhibition ofwhich was selectively lethal in oxaliplatin-resistant colon cancer cells. Oxaliplatin-resistant HCT116-R1 cells were infected with viral particles expressing the 1,798shRNA vectors that target 518 genes from the kinome library and screened for shRNAs that cause lethality after addition of oxaliplatin. C, Three shRNAs targetingATRwere significantly depleted in HCT116-R1 cells incubated with oxaliplatin. D, ATR knockdownwas evaluated byWestern blot analysis. The shLuc vector wasused as a control. E and F, HCT116-R1 cells in which ATRwas knocked down or not (shATR and shLuc) were incubated with increasing concentrations ofoxaliplatin (x-axis) and viability was assessed with the SRB assay (y-axis; E), and the IC50 values calculated (F). G, Analysis of the ATR pathway activation inHCT116 and HCT116-R1 cells. ATR, phosphorylated ATR (p-ATR), CHK1, and p-CHK1 were evaluated byWestern blot analysis. Tubulin levels were used as loadingcontrol. H,Differential ATR gene expression in tissue samples from patients with mCRC. HM, hepatic metastasis; NM, normal mucosa; PT, primary tumor.

Comb�es et al.

Cancer Res; 79(11) June 1, 2019 Cancer Research2938

on January 14, 2021. © 2019 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst April 15, 2019; DOI: 10.1158/0008-5472.CAN-18-2807

Page 7: Inhibition of Ataxia-Telangiectasia Mutated and RAD3 ... · Apothicaires, Montpellier 34298, France. Phone: 334-6761-3745; Fax: 334-6761-3787; E-mail: celine.gongora@inserm.fr Cancer

proliferation. First, cell-cycle analysis showed that each com-pound had only a minor effect on the cell-cycle distribution ofHCT116-R1 cells. Conversely, the VE-822 þ oxaliplatin combi-nation reduced significantly the number of S-phase cells andincreased that of cells in the sub-G1 phase (Fig. 3A).We confirmedthis finding using the EdU assay. Oxaliplatin and VE-822 alonehad no effect on cell proliferation, whereas in combination theyinhibited cell proliferation, as indicated by the reduction of EdU-positive HCT116-R1 cells (from 75% in untreated cells to 18% inoxaliplatin þ VE-822–treated cells; P ¼ 0.0094; Fig. 3B).

Next, we assessed apoptosis by quantifying Annexin-Vþ

HCT116-R1 cells (Fig. 3C). Only the VE-822 þ oxaliplatin com-bination significantly and strongly induced apoptosis comparedwith untreated cells and the drugs alone. Moreover, p53 and

p21 expression levels were increased by the drug combination,whereas phosphorylation of the ATR target CDK2 was decreasedcompared with cells without treatment or incubated with eachsingle drug (Fig. 3D; Supplementary Fig. S2B and S2C). Altogeth-er, these results demonstrate that the synergy between the ATRiand oxaliplatin impairs cell-cycle progression and proliferation,and promotes cell death.

To assess the effect of these drugs on the ATR signaling pathway,we performed immunoblotting analysis using HCT116-R1(Fig. 3E) and HCT116 cells (Supplementary Fig. S2A–S2C) afterincubation with oxaliplatin and/or VE-822. In both celllines, oxaliplatin treatment promoted ATR and CHK1 phosphor-ylation. As expected, this effect was inhibited when cells wereincubated with oxaliplatin and VE-822. As the ATR and ATM

Figure 2.

ATR inhibition combined with oxaliplatin alters colorectal cancer cell viability in vitro and in vivo. A, The indicated colorectal cancer cell lines were incubated withincreasing concentrations of oxaliplatin and the ATR inhibitor VE-822, and cell viability was assessed with the SRB assay (2D) and by measuring spheroid growth(3D) to obtain the viability matrix. Drug concentrations were as follows: VE-822 (from 0.0075 to 2.56 mmol/L for 2D and from 0.01 to 2.52 mmol/L for 3D),oxaliplatin (from 0.184 and 30 mmol/L for 2D and from 0.09 and 30 mmol/L for 3D). The synergymatrix was calculated as described in Materials and Methods.B, Effect of the VE-822þ oxaliplatin combination on the tumor volume in nudemice xenografted with HCT116-R1 cells. Mice received oxaliplatin, VE-822, or bothor vehicle (NT, normal tissue). C, Kaplan–Meier survival analysis of the mice described in B.

Inhibition of ATR Overcomes Resistance to Oxaliplatin in CRC

www.aacrjournals.org Cancer Res; 79(11) June 1, 2019 2939

on January 14, 2021. © 2019 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst April 15, 2019; DOI: 10.1158/0008-5472.CAN-18-2807

Page 8: Inhibition of Ataxia-Telangiectasia Mutated and RAD3 ... · Apothicaires, Montpellier 34298, France. Phone: 334-6761-3745; Fax: 334-6761-3787; E-mail: celine.gongora@inserm.fr Cancer

pathways cross-talk with each other, we also evaluated ATM andCHK2 expression (Fig. 3E, right). VE-822 induced CHK2 phos-phorylation, whereas oxaliplatin did not have detectable effect.Conversely, the oxaliplatin and VE-822 combination significantlyincreased ATM and CHK2 phosphorylation, indicating that in thepresence of oxaliplatin, theVE-822–induced inhibitionof theATRsignaling pathway leads to activation of the ATM pathway. Alto-gether, these results demonstrate that the combination of oxali-platin þ VE-822 has a cytotoxic and cytostatic effect in colorectalcancer cell lines, and that it can effectively inhibit theATRpathwayand activate ATM signaling.

The VE-822 þ oxaliplatin combination induces replicationstress, leading to DNA break formation

ATR plays an essential role in the cell response to replicationstress (RS; ref. 19). As accumulation of ssDNAandDNAdamage isan RS feature, we quantified levels of RPA, a protein covering

persisting ssDNA and gH2AX, marker of DNA damage, inHCT116-R1 cells incubated with VE-822 and/or oxaliplatin. Forthis, we used a specific fluorimetric assay that allows to correlatethe respective protein levels in cell lysates (Fig. 4A). Comparedwithuntreated cells, RPA levelswere significantly increased in cellsincubated with oxaliplatin, but not with VE-822 (Fig. 4B). Thiseffect was even more pronounced when oxaliplatin administra-tionwas combinedwith VE-822. Similar results were obtained forgH2AX, indicating that the VE-822 þ oxaliplatin combinationacts synergistically to induce DNA damage (Fig. 4B).

Temporal analysis of RPA (ssDNA) and gH2AX (DNA damage)labeling showed that oxaliplatin alone induced only a slightincrease in RPA staining, but not in gH2AX or RPAþgH2AXlabeling (Fig. 4C, left). Notably, combined VE-822 þ oxaliplatintreatment increased the number of double gH2AX- and RPA-positive cells over time (Fig. 4C, right), indicating replicationcatastrophe driven by excessive ssDNA accumulation (20).

Figure 3.

ATR inhibition combined with oxaliplatin alters the cell-cycle distribution profile and induces cell death. HCT116-R1 cells were incubated or not with oxaliplatin(5 mmol/L) and/or VE-822 (1 mmol/L) as indicated for 48 hours, and then the cell-cycle distribution was analyzed using the propidium iodide staining method (A),and the number of EdU-positive cells (B) and Annexin-V–positive cells (C) was quantified by flow cytometry. Data are the mean� SD of at least threeindependent experiments. � , P < 0.05, compared with control; #, compared with the drug combination. D and E,Western blot analysis of p53, p21, CDK2 (D), theATR-CHK1 and ATM-CHK2 signaling pathways (E) in HCT116-R1 cells incubated or not with oxaliplatin (2.5 mmol/L) and/or VE-822 (1 mmol/L) as indicated for24 hours. Tubulin and GAPDHwere used as loading controls.

Comb�es et al.

Cancer Res; 79(11) June 1, 2019 Cancer Research2940

on January 14, 2021. © 2019 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst April 15, 2019; DOI: 10.1158/0008-5472.CAN-18-2807

Page 9: Inhibition of Ataxia-Telangiectasia Mutated and RAD3 ... · Apothicaires, Montpellier 34298, France. Phone: 334-6761-3745; Fax: 334-6761-3787; E-mail: celine.gongora@inserm.fr Cancer

To explore the impact of both drugs on RS, we analyzedreplication fork speed in HCT116-R1 cells (Fig. 4D). VE-822(1 mmol/L for 24 hours) strongly affected replication inHCT116-R1 cells, decreasing fork speed by about 40%, aspreviously shown with the ATRi VE-821 (21). Oxaliplatinadducts were also reported to affect polymerase progres-sion (22), and we describe here for the first time that oxaliplatin(12.5 mmol/L for 24 hours) decreased significantly replicationfork speed by 10%. The VE-822 þ oxaliplatin combinationinhibited replication fork speed stronger than each drug on its

own, and in some cases even led to replication fork arrest. Theseresults show that the VE-822 þ oxaliplatin combinationinduces a dramatic decrease in replication fork speed, leadingto RS and replication catastrophe.

As exposure to genotoxic agents and replication fork stallingincrease the levels of cytosolic DNA (23), we assessed whether theVE-822 þ oxaliplatin combination affected the cytosolic DNAlevels. In fact, we observed that VE-822 or oxaliplatin aloneslightly increased the cytosolic DNA level, whereas the drugcombination had a much stronger effect (Fig. 4E). This concurs

Figure 4.

ATR inhibition combined with oxaliplatin induces replication stress, DNA break formation, and cytosolic DNA. A, Illustration of the method used to determine theRPA, gH2AX, and RPAþ gH2AX signal intensity. B,Analysis of total RPA and total gH2AX signal intensity after 16 hours of incubationwith VE-822 and/oroxaliplatin (Ox) as indicated. Data are the mean� SD of at least three independent experiments. �� ,P <0.01; ��� , P <0.001, compared with control; #, comparedwith oxaliplatin. C, Separated analysis of RPA, gH2AX, and RPAþ gH2AX labeling as a function of time after addition of oxaliplatin (50 mmol/L; left) or of VE-822(1 mmol/L)þ oxaliplatin (50 mmol/L; right). D, Replication fork speed measurements (CldU track length in mmol/L) in HCT116-R1 cells incubated with oxaliplatin(12.5 mmol/L) and/or VE-822 (1 mmol/L) or not (untreated); the bottom right graph summarizes the results of all experiments relative to untreated cells set to 1.Bold line, mean. AU, arbitrary unit. �� , P < 0.01; ��� , P <0.001. E, HCT116-R1 cells were incubated or not (CTR) with 1 mmol/L VE-822 and/or 12.5 mmol/L oxaliplatin(as indicated) for 48 hours and stained for cytosolic dsDNA or ssDNA (green) and with DAPI (blue). Images were acquired by microscopy and analyzed using theZEN software. Data are representative of three independent experiments.

Inhibition of ATR Overcomes Resistance to Oxaliplatin in CRC

www.aacrjournals.org Cancer Res; 79(11) June 1, 2019 2941

on January 14, 2021. © 2019 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst April 15, 2019; DOI: 10.1158/0008-5472.CAN-18-2807

Page 10: Inhibition of Ataxia-Telangiectasia Mutated and RAD3 ... · Apothicaires, Montpellier 34298, France. Phone: 334-6761-3745; Fax: 334-6761-3787; E-mail: celine.gongora@inserm.fr Cancer

with the observation that the VE-822 þ oxaliplatin combinationincreases ssDNA and dsDNA levels what could then lead toactivation of the immune system.

The VE-822þ oxaliplatin combination promotes the antitumorT-cell response

Oxaliplatin has been described to induce ICD in vitro and in vivoin different preclinical cancer models (4, 24). ICD is a specific celldeath characterized by the release or expression of damage-asso-ciated molecular patterns (DAMP), such as adenosine-50-triphos-phate (ATP) and HMGB-1 release and CRT translocation. Wetherefore decided to test the effect of the combined VE-822 þoxaliplatin administration in vivo.

For this, wefirst askedwhether theATRi VE-822 synergizedwithoxaliplatin also inmouse CT26 andMC38 colorectal cancer cells.To this aim, we used the concentration matrix approach and SRBcytotoxicity tests with four oxaliplatin and five VE-822 concen-tration in 2D (SRB) and 3D (spheroid growth) cell assays (Sup-plementary Fig. S3A). Like in human colorectal cancer cell lines,we observed a strong synergistic interaction between oxaliplatinand VE-822 in bothmouse cell lines, with higher values in the 3Dthan in the 2D assay.

We then monitored ICD in vitro by assessing CRT exposure,ATP secretion, and HMGB1 release (25) in mouse MC38 cellsand in oxaliplatin-resistant human HCT116-R1 cells (Fig. 5A–C; Supplementary Fig. S3B and S3C for HCT116 cells). Quan-tification of CRT translocation to the outer leaflet of the plasmamembrane by flow cytometry of live cells (Fig. 5A) showed thatoxaliplatin and the drug combination, but not VE-822 alone,strongly increased CRT translocation, in both cell lines. Also,ATP secretion was promoted by the drug combination in bothcell lines (Fig. 5B). HMGB1 release was significantly increasedonly in MC38 cells incubated with the VE-822 þ oxaliplatincombination (Fig. 5C). These results reveal that the ICD induc-tion by oxaliplatin is significantly increased in combinationwith the ATRi VE-822.

To test whether the VE-822 þ oxaliplatin combinationincreased ICD also in vivo, we grafted the syngeneic MC38 cellsin immunocompetent C57/Bl6 mice (Fig. 5D). When tumorsreached a volume of 100 mm3, we treated them or not withoxaliplatin, VE-822, or the oxaliplatin þ VE-822 combination.Like in nude mice (Fig. 2B), VE-822 had no effect on tumorgrowth. Conversely, oxaliplatin and particularly the oxaliplatinþ VE-822 combination decreased tumor growth (Fig. 5D).Importantly, Kaplan–Meier survival analysis showed that oxa-liplatin þ VE-822 was more efficient than control (P ¼0.00471), and also oxaliplatin (P ¼ 0.0237) and VE-822 alone(P ¼ 0.0218; Fig. 5E). The higher efficiency of the drug com-bination in immunocompetent mice suggests the establish-ment of a specific antitumor immune response. To test whetherVE-822 amplifies oxaliplatin immunogenic effect, we sacrificedmice at week 3 after treatment and quantified IFNg-producingCD8-positive T cells in the spleen in untreated animals andmice treated with oxaliplatin alone or combined with VE-822(Fig. 5F). After stimulation with irradiated MC38 cells (targetcells), IFNg-producing CD8-positive T cells (effector cells) weresignificantly increased (P ¼ 0.0121) in mice that receivedoxaliplatin þ VE-822 compared with those treated only withoxaliplatin. Altogether, these results provide evidence that ATRinhibition can reverse resistance to oxaliplatin and amplifyoxaliplatin-induced ICD in vitro and in vivo.

Identification of DNA repair genes associated with overallsurvival and response tooxaliplatin-based treatment inpatientswith colorectal cancer

Our results indicated that the ATRi VE-822 strongly synergizeswith oxaliplatin in colorectal cancer cells. Therefore, we askedwhether someDNA repair genes that are deregulated in colorectalcancer are associated with overall survival (OS) and/or responseto oxaliplatin-based treatment. To this aim, we used microarraygene expression data from primary tumors before treatment(FOLFOX regimen) from two independent cohorts of patientswith colorectal cancer: (i) Tsuji cohort [n¼ 80 patients (26)], and(ii) Del Rio cohort (n ¼ 36 patients; ref. 27). We defined a list of176 genes involved in the six major DNA repair pathways [baseexcision repair (BER), nucleotide excision repair (NER),mismatchrepair (MMR), homologous recombination repair (HRR), non-homologous end joining (NHEJ), and FANC] using the REPAIR-toire database (http://repairtoire.genesilico.pl) and literaturedata (14).Using theMaxstat R function andBenjamini–Hochbergmultiple testing correction, we found that 12 of these 176 geneshad a prognostic value (5 genes with poor and 7 with goodprognostic values; Fig. 6A, left). We used this DNA repair genesignature to create the DNA repair score. In the Tsuji cohort,patients with a high DNA repair score were characterized bystronger expression of the five bad prognostic genes and weakerexpression of the seven good prognostic genes (Fig. 6A, right).Using the Maxstat R function, we found the maximum differenceinOSwith aDNA repair score¼�4.2464 that split patients in theTsuji cohort into a low survival group (67.5% of patients, medianOS ¼ 12 months), and a high survival group (32.5% of patients,median survival not reached; P ¼ 6.58 10�5; Fig. 6B). The DNArepair score, computedusing the Tsuji cohort parameters,was alsoprognostic in the Del Rio cohort (Fig. 6C, left). The median OS ofpatients with high DNA repair score (i.e., higher than �4.2464)was 17 months, whereas it was not reached for patients withlow DNA repair score (P ¼ 0.00181). The Del Rio cohort alsoincluded data on the response after the first-line FOLFOX che-motherapy, according to the WHO criteria. Using the sameDNA repair score threshold (�4.2464), we found a significantdifference in the median progression-free survival (PFS) betweengroups (8 months for patients with high DNA repair score and21 months for patients with low DNA repair score; P ¼ 0.0189;Fig. 6C, right). These findings confirm that the ATR and DNAdamage response pathways have an important prognostic andtherapeutic value in patients with colorectal cancer.

DiscussionDespite the advances in mCRC management, the 5-year sur-

vival rate is still 12% (28).One of the causes of treatment failure isthe resistance to therapy that occurs in 90% of patients withmCRC (29). Hence, the aim of this study was to identify newtherapeutic targets toovercomeoxaliplatin resistance in colorectalcancer. Using a "chemical synthetic lethality screening," alsocalled "dropout screen," we found that ATR inhibition synergizeswith oxaliplatin to induce cancer cell death. The combination ofoxaliplatin with the ATRi VE-822 was cytotoxic and cytostatic,both in vitro and in vivo.

The ATR protein kinase is a member of the PI3K-related kinase(PIKK) family and activates CHK1 (30). ATR is activated by RPA-coated ssDNA during S-phase to promote genomic stability inresponse to DNA damage, cell-cycle arrest, or even stabilization

Comb�es et al.

Cancer Res; 79(11) June 1, 2019 Cancer Research2942

on January 14, 2021. © 2019 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst April 15, 2019; DOI: 10.1158/0008-5472.CAN-18-2807

Page 11: Inhibition of Ataxia-Telangiectasia Mutated and RAD3 ... · Apothicaires, Montpellier 34298, France. Phone: 334-6761-3745; Fax: 334-6761-3787; E-mail: celine.gongora@inserm.fr Cancer

and repair of replication forks (31–33). Here, we found that theATRi þ oxaliplatin combination promoted cell death and inhib-ited cell proliferation, as observed in the presence of high repli-cation stress, DNA damage, and activation of the ATM pathway.These data are in agreement with the model presented by Toledoand colleagues (20), suggesting that VE-822 might increase rep-licative stress generated by oxaliplatin and causing RPA shortage.If ssDNA is not stabilized by RPA, its phosphorylation by ATRbecomes unstable. Thus, in unstable ssDNA regions, DNA dam-

age increases, promoting the activation of the ATM–CHK2 path-way and the presence of cytosolic DNA. Finally, these molecularevents lead to the inhibition of replication and induction of celldeath. Moreover, as cancer cells may be defective in G1–S check-points, theymight require the ATR–CHK1 pathway to repair DNAdamage (34), thus making them more sensitive/suitable to ATRinhibition than normal cells.

To the best of our knowledge, this is the first report that clarifiesthe molecular mechanisms underlying the synergism between

Figure 5.

Oxaliplatin combined with the ATR inhibitor VE-822 induces immune death signaling.A, CRT exposure at the cell surface of mouse MC38 and human HCT116-R1colorectal cancer cells was assessed after 48-hour incubation with oxaliplatin (25 mmol/L) and/or VE-822 (1 mmol/L), or not (control) by flow cytometry analysis.� , P < 0.05, compared with control; #, compared with the drug combination. B and C,ATP and HMGB1 release in the supernatant was assessed usingluminescence and ELISA assays, respectively, in MC38 and HCT116-R1 cells incubated with oxaliplatin (12.5 or 100 mmol/L) and/or VE-822 (1 mmol/L) for 24 hoursor 48 hours. Data are the mean� SD of at least three independent experiments. � , P < 0.05, compared with control; #, compared with the drug combination.D and E, Effect of treatment with VE-822 (30 mg/kg) and/or oxaliplatin (5 mg/kg) on tumor growth in C57BL/6J mice xenografted with MC38 colorectal cancercells (n¼ 5–7mice/group;D) and Kaplan–Meier survival curves (E). F, Autologous mixed lymphocyte-MC38 tumor cell culture. Peripheral antitumor CD8-positive T cells were detected in the spleen of treated and control mice on the basis of their capacity to secrete IFNg after coculture with target (irradiated MC38)cells. Living IFNg-secreting T cells were detected by flow cytometry. Left, dot plots, representative results for splenocytes isolated from treated or control (na€�ve)mice. Right, histogram, percentage of IFNg-secreting CD8-positive T cells from three independent experiments in na€�ve (n¼ 5), oxaliplatin (Ox)þ VE-822–treated (n¼ 8), and oxaliplatin-treatedmice (n¼ 3). � , P < 0.05 (nonparametric Mann andWhitney t test). NT, normal tissue.

Inhibition of ATR Overcomes Resistance to Oxaliplatin in CRC

www.aacrjournals.org Cancer Res; 79(11) June 1, 2019 2943

on January 14, 2021. © 2019 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst April 15, 2019; DOI: 10.1158/0008-5472.CAN-18-2807

Page 12: Inhibition of Ataxia-Telangiectasia Mutated and RAD3 ... · Apothicaires, Montpellier 34298, France. Phone: 334-6761-3745; Fax: 334-6761-3787; E-mail: celine.gongora@inserm.fr Cancer

oxaliplatin and the ATRi VE-822 in human cells, a long time afterthe studies on fission yeast by Perego and colleagues showing therelevance of ATR homologue (rad3) in the sensitivity to oxali-platin (35). It also shows that oxaliplatin can induce replicativestress, the activation of ATR–CHK1 pathway and ssDNA accumu-lation (and RPA levels) on its own, and, as well as increased DNAdamages and ICD when combined with VE-822.

Acquired resistance to oxaliplatin has limited the managementof colorectal cancer and other tumor types. Our loss-of-functiongenetic screen allowed showing that the kinase ATR is, at least inpart, responsible of this resistance, and suggests that the ATRi andoxaliplatin combination therapy could be useful in the clinic.Similar screenings have already demonstrated that ATR, CHK1,and WEE1 are involved in the sensitivity to cisplatin in triple-negative breast cancer (36). Moreover, ATR inhibition sensitizedcancer cells to chemotherapy, for instance in ovarian cancer(in combination with cisplatin and gemcitabine), in pancreaticcancer (with gemcitabine), in breast cancer (with camptothecin)and in lung cancer (with cisplatin, oxaliplatin, gemcitabine,

etoposide, and SN38; ref. 37). However, none of these studiesreported that the combination of ATRi with chemotherapeuticdrugs or radiotherapy can overcome drug resistance, or its molec-ular effects. Moreover, the role of ATR in oxaliplatin resistance issomewhat controversial because at least two studies have shownopposite effects. First, Lewis and colleagues (2009) found thatATR contributes to the survival of fibroblast and osteosarcomacell lines after cisplatin, but not oxaliplatin treatment (38).Conversely, Hall and colleagues, demonstrated in 35 lungcancer cell lines that ATR inhibition sensitizes cells to fiveDNA-damaging drugs (cisplatin, oxaliplatin, gemcitabine, eto-poside, and SN38; ref. 37). Moreover, they found that while thecombination of the ATRi VX-970 with cisplatin or etoposide isalways synergistic, the combination with oxaliplatin is syner-gistic in only 60% of the tested cell lines and antagonistic in theothers (37). These findings suggest that the response to theoxaliplatin and ATRi combination is cell type–specific. Ourresults showed synergistic effects in all tested colorectal cancercell lines, even in oxaliplatin-resistant colorectal cancer cell

Figure 6.

Identification of prognostic genes that are regulated by oxaliplatin treatment in two independent cohorts of patients. A, Left, The DNA repair gene signatureobtained after Maxstat R function and Benjamini–Hochberg multiple testing correction. Five genes have poor and seven genes good prognostic value. Right,clustergram of the DNA repair score genes ordered from best to worst prognosis. The level of the probe set signal is displayed from low (deep blue) to high(deep red) expression. B, Patients with colorectal cancer (N¼ 80; Tsuji cohort) were ordered according to the increasing DNA repair score. C and D, High DNArepair score in the primary colorectal cancer could predict shorter OS and PFS. Patients from the Tsuji cohort (N¼ 80) and the Del Rio cohort (N¼ 36) weredivided into two groups based on the DNA repair score threshold of�4.2464 (group 0, low DNA repair score, score lower than�4.2464; group 1, high DNArepair score, score higher than�4.2464).

Comb�es et al.

Cancer Res; 79(11) June 1, 2019 Cancer Research2944

on January 14, 2021. © 2019 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst April 15, 2019; DOI: 10.1158/0008-5472.CAN-18-2807

Page 13: Inhibition of Ataxia-Telangiectasia Mutated and RAD3 ... · Apothicaires, Montpellier 34298, France. Phone: 334-6761-3745; Fax: 334-6761-3787; E-mail: celine.gongora@inserm.fr Cancer

lines, suggesting that colorectal cancer is a potential target forthe synergistic interaction between oxaliplatin and ATRi.

The synergistic effects of oxaliplatin and VE-822 also promotedICD signals. It has been recently shown that oxaliplatin antitumoractivity is due, at least in part, to ICD activation (4). Tumor cellsincubated with the accurate concentration of oxaliplatin are ableto produce ATP (laborious to be measured), release the DNA-binding protein HMGB1 in the tumor microenvironment, andexpose the chaperone protein CRT to the outer part of the plasmamembrane. These three signals favor the priming of the hostantitumor immune response, and thus an efficient eliminationof tumor cells, which could result in a long-term antitumorimmunity. Here, we found that in the tested colorectal cancercell lines, oxaliplatin could induce these three signals, as expected,and for the first time that ATRi VE-822 potentiates this effect.

Cytosolic DNA can be recognized by DNA sensors, leading tothe expression of type I IFNs (39) that has tumor-suppressiveeffects (40). Moreover, replication fork stalling may induce theformation of extended ssDNA regions, dsDNA breaks, and theactivation of theDNAdamage response. Shen and colleagues (23)reported that DNA damage caused by replication fork stallingleads to the presence of genome-derived DNA in the cytosol oftumor cells and to activation of immune responses. Here, wefound that incubation of colorectal cancer cells with VE-822 andoxaliplatin increases the cytosolic DNA levels. This could be theconsequence of replicative stress, and could promote the antitu-mor immunosurveillance. These in vitro results were confirmed bythe results in immunocompetent mice, where treatment with theoxaliplatin þ VE-822 combination promoted tumor growthinhibition and tumor specific CD8-positive T-cell activation.Although the combination of oxalipatinþ ATR inhibition resultsin immunogenic cancer cell death in vivo, it is not clear howmuchof the observed tumor size reduction is actually due to a cancer cellautonomous effect of the combined therapy versus immunogeniccancer cell death.

The clinical efficacy of oxaliplatin has been largely demonstrat-ed, and its mechanism of action is well known. As a platinumderivative, its primary target is DNA, where it can form intra- andinterstrand cross-links. The repair of these lesions mostly occursthrough the NER pathway that involves ERCC1, 2, 3, 4, 5. Indeed,the association between ERCC1 expression and response toplatinum derivatives has been widely reported, particularly inadvanced non–small-cell lung, bladder, head-and-neck andesophagus cancer (for cisplatin) and in colorectal cancer (foroxaliplatin).

In agreement, the gene expression–based predictiveDNA repairscore we developed for OS and PFS of patients with colorectalcancer undergoing oxaliplatin treatment validates the importanceof ERCC1 in oxaliplatin resistance, and identifies four other NERgenes (GTF2H3, HMGN1, RPA2, and GTF2H2) as involved in

resistance to this drug. This confirms the impact of the NERpathway in the repair of oxaliplatin-induced lesions. Moreover,50% of the genes in the DNA repair signature belong to the HRRpathway (SCARP, XRCC2, DMC1, BARD1, C19orf40, and RPA2).This couldbe surprising, but is consistentwithour results showingthat ATR, the major protein of the HRR pathway, is implicated inresistance to oxaliplatin. Thus, the DNA repair score couldbecome a biomarker of oxaliplatin response in patients withcolorectal cancer and might allow identifying the patients whowill most benefit from the oxaliplatin þ ATRi combination.

Altogether, our data indicate that the ATRi þ oxaliplatin com-bination is efficient in colorectal cancer cells, and also in oxali-platin-resistant colorectal cancer cells. Our results provide arational mechanistic basis for clinical trials with the oxaliplatinþ ATRi combination.

Disclosure of Potential Conflicts of InterestNo potential conflicts of interest were disclosed.

Authors' ContributionsConception and design: H.-A. Michaud, A. Coquelle, P. Pasero, N. Bonnefoy,R.L. Beijersbergen, N. Vezzio-Vie, C. GongoraDevelopment of methodology: E. Comb�es, A.F. Andrade, D. Tosi,R.L. Beijersbergen, N. Vezzio-Vie, C. GongoraAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): E. Comb�es, A.F. Andrade, H.-A. Michaud,F. Coquel, D. Desigaud, M.D. Rio, N. Vezzio-VieAnalysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis): E. Comb�es, A.F. Andrade, D. Tosi, H.-A. Michaud,M. Jarlier, J. Moreaux, R.L. Beijersbergen, N. Vezzio-Vie, C. GongoraWriting, review, and/or revision of the manuscript: E. Comb�es, A.F. Andrade,D. Tosi, A. Coquelle, J. Moreaux, P. Martineau, C. GongoraAdministrative, technical, or material support (i.e., reporting or organizingdata, constructing databases): V. Garambois, C. GongoraStudy supervision: N. Vezzio-Vie, C. Gongora

AcknowledgmentsWe acknowledge the imaging facility MRI, member of the national infra-

structure France-BioImaging supported by the FrenchNational Research Agency(ANR-10-INBS-04, Investments for the Future) for flow cytometry, and Celigoexperiments. We thank the IRCM animal facility. This work was supported byINSERM and the Institut du cancer de Montpellier (ICM), SIRIC (SIRICMontpellier Cancer Grant, INCa-DGOS-Inserm 6045), and Canc�eropole GSO.E. Comb�es had a PhD studentship from the program Investissement D'avenir(grant agreement: Labex MabImprove, ANR-10-LABX-53-01). A.F. Andradewas supported by S~ao Paulo Research Foundation (14/06947-2) and Fondationde France (postdoctoral fellowship).

The costs of publication of this articlewere defrayed inpart by the payment ofpage charges. This article must therefore be hereby marked advertisement inaccordance with 18 U.S.C. Section 1734 solely to indicate this fact.

Received September 11, 2018; revised January 3, 2019; acceptedApril 5, 2019;published first April 15, 2019.

References1. Global Burden of Disease Cancer Collaboration, Fitzmaurice C, Allen C,

Barber RM, Barregard L, Bhutta ZA, et al. Global, regional, and nationalcancer incidence,mortality, years of life lost, years livedwith disability, anddisability-adjusted life-years for 32 cancer groups, 1990 to 2015: a sys-tematic analysis for theGlobal Burden ofDisease Study. JAMAOncol 2017;3:524–48.

2. Van Cutsem E, Nordlinger B, Cervantes A, ESMO Guidelines WorkingGroup. Advanced colorectal cancer: ESMO clinical practice guidelines fortreatment. Ann Oncol 2010;21 Suppl 5:v93–7.

3. Perego P, Robert J. Oxaliplatin in the era of personalized medicine: frommechanistic studies to clinical efficacy. Cancer Chemother Pharmacol2016;77:5–18.

4. Tesniere A, Schlemmer F, Boige V, Kepp O, Martins I, Ghiringhelli F, et al.Immunogenic death of colon cancer cells treated with oxaliplatin. Onco-gene 2010;29:482–91.

5. Gourdier I, Del Rio M, Crabb�e L, Candeil L, Copois V, YchouM, et al. Drugspecific resistance to oxaliplatin is associated with apoptosis defect in acellular model of colon carcinoma. FEBS Lett 2002;529:232–6.

Inhibition of ATR Overcomes Resistance to Oxaliplatin in CRC

www.aacrjournals.org Cancer Res; 79(11) June 1, 2019 2945

on January 14, 2021. © 2019 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst April 15, 2019; DOI: 10.1158/0008-5472.CAN-18-2807

Page 14: Inhibition of Ataxia-Telangiectasia Mutated and RAD3 ... · Apothicaires, Montpellier 34298, France. Phone: 334-6761-3745; Fax: 334-6761-3787; E-mail: celine.gongora@inserm.fr Cancer

6. Samimi G, Manorek G, Castel R, Breaux JK, Cheng TC, Berry CC, et al.cDNAmicroarray-based identification of genes and pathways associatedwith oxaliplatin resistance. Cancer Chemother Pharmacol 2005;55:1–11.

7. Virag P, Brie I, Fischer-Fodor E, Perde-Schrepler M, Tatomir C, BalacescuO,et al. Assessment of cytotoxicity, apoptosis and DNA damages in Colo320colorectal cancer cells selected for oxaliplatin resistance. Cell BiochemFunct 2011;29:351–5.

8. Prahallad A, Sun C,Huang S, Di Nicolantonio F, Salazar R, Zecchin D, et al.Unresponsiveness of colon cancer to BRAF(V600E) inhibition throughfeedback activation of EGFR. Nature 2012;483:100–3.

9. SkehanP, Storeng R, ScudieroD,MonksA,McMahon J, VisticaD, et al. Newcolorimetric cytotoxicity assay for anticancer-drug screening. J Natl CancerInst 1990;82:1107–12.

10. Greco WR, Bravo G, Parsons JC. The search for synergy: a critical reviewfrom a response surface perspective. Pharmacol Rev 1995;47:331–85.

11. Del Rio M, Mollevi C, Bibeau F, Vie N, Selves J, Emile JF, et al. Molecularsubtypes of metastatic colorectal cancer are associated with patientresponse to irinotecan-based therapies. Eur J Cancer 2017;76:68–75.

12. Kassambara A, R�eme T, Jourdan M, Fest T, Hose D, Tarte K, et al.GenomicScape: an easy-to-use web tool for gene expression dataanalysis. Application to investigate the molecular events in the differ-entiation of B cells into plasma cells. PLoS Comput Biol 2015;11:e1004077.

13. Milanowska K, Krwawicz J, Papaj G, Kosinski J, Poleszak K, Lesiak J, et al.REPAIRtoire–a database of DNA repair pathways. Nucleic Acids Res 2011;39:D788–92.

14. Bret C, Klein B, CartronG, Schved J-F, Constantinou A, Pasero P, et al. DNArepair in diffuse large B-cell lymphoma: amolecular portrait. Br JHaematol2015;169:296–9.

15. Kassambara A, Hose D, Moreaux J, R�eme T, Torrent J, Rossi JF, et al.Identification of pluripotent and adult stem cell genes unrelated to cellcycle and associated with poor prognosis in multiple myeloma. PloS One2012;7:e42161.

16. Kassambara A, HoseD,Moreaux J,Walker BA, Protopopov A, Reme T, et al.Genes with a spike expression are clustered in chromosome (sub)bandsand spike (sub)bands have a powerful prognostic value in patients withmultiple myeloma. Haematologica 2012;97:622–30.

17. Moreaux J, R�eme T, Leonard W, Veyrune JL, Requirand G, Goldschmidt H,et al. Development of gene expression-based score to predict sensitivityof multiple myeloma cells to DNA methylation inhibitors. Mol CancerTher 2012;11:2685–92.

18. Del Rio M, Mollevi C, Vezzio-Vie N, Bibeau F, Ychou M, Martineau P.Specific extracellular matrix remodeling signature of colon hepatic metas-tases. PLoS One 2013;8:e74599.

19. L�opez-Contreras AJ, Fernandez-CapetilloO. The ATRbarrier to replication-born DNA damage. DNA Repair 2010;9:1249–55.

20. Toledo LI, Altmeyer M, Rask MB, Lukas C, Larsen DH, Povlsen LK, et al.ATR prohibits replication catastrophe by preventing global exhaustion ofRPA. Cell 2013;155:1088–103.

21. Joss�e R,Martin SE, Guha R, Ormanoglu P, Pfister TD, Reaper PM, et al. ATRinhibitors VE-821 and VX-970 sensitize cancer cells to topoisomerase iinhibitors by disabling DNA replication initiation and fork elongationresponses. Cancer Res 2014;74:6968–79.

22. Chaney SG, Campbell SL, Temple B, Bassett E, Wu Y, Faldu M. Proteininteractions with platinum-DNA adducts: from structure to function.J Inorg Biochem 2004;98:1551–9.

23. Shen YJ, Le Bert N, Chitre AA, Koo CX, Nga XH, Ho SS, et al. Genome-derived cytosolic DNAmediates type I interferon-dependent rejection of Bcell lymphoma cells. Cell Rep 2015;11:460–73.

24. Wang W, Wu L, Zhang J, Wu H, Han E, Guo Q. Chemoimmunotherapy bycombining oxaliplatin with immune checkpoint blockades reduced tumorburden in colorectal cancer animalmodel. BiochemBiophys Res Commun2017;487:1–7.

25. Kepp O, Senovilla L, Vitale I, Vacchelli E, Adjemian S, Agostinis P, et al.Consensus guidelines for the detection of immunogenic cell death.Oncoimmunology 2014;3:e955691.

26. Tsuji S, Midorikawa Y, Takahashi T, Yagi K, Takayama T, Yoshida K, et al.Potential responders to FOLFOX therapy for colorectal cancer by RandomForests analysis. Br J Cancer 2012;106:126–32.

27. Del Rio M, Molina F, Bascoul-Mollevi C, Copois V, Bibeau F, Chalbos P,et al. Gene expression signature in advanced colorectal cancer patientsselect drugs and response for the use of leucovorin, fluorouracil, andirinotecan. J Clin Oncol 2007;25:773–80.

28. Ferlay J, Shin HR, Bray F, Forman D, Mathers C, Parkin DM. Estimates ofworldwide burden of cancer in 2008:GLOBOCAN2008. Int J Cancer 2010;127:2893–917.

29. Longley DB, Johnston PG. Molecular mechanisms of drug resistance.J Pathol 2005;205:275–92.

30. Engelman JA, Luo J, Cantley LC. The evolution of phosphatidylinositol3-kinases as regulators of growth and metabolism. Nat Rev Genet 2006;7:606–19.

31. Kumagai A, Lee J, Yoo HY, Dunphy WG. TopBP1 activates the ATR-ATRIPcomplex. Cell 2006;124:943–55.

32. Kumagai A, Dunphy WG. How cells activate ATR. Cell Cycle 2006;5:1265–8.

33. Cimprich KA, Cortez D. ATR: an essential regulator of genome integrity.Nat Rev Mol Cell Biol 2008;9:616–27.

34. Fokas E, Prevo R, Hammond EM, Brunner TB, McKenna WG, Muschel RJ.Targeting ATR in DNA damage response and cancer therapeutics.Cancer Treat Rev 2014;40:109–17.

35. Perego P, Zunino F, Carenini N, Giuliani F, Spinelli S, Howell SB. Sensi-tivity to cisplatin and platinum-containing compounds of Schizosacchar-omyces pombe rad mutants. Mol Pharmacol 1998;54:213–9.

36. Zheng H, Shao F, Martin S, Xu X, Deng CX. WEE1 inhibition targets cellcycle checkpoints for triple negative breast cancers to overcome cisplatinresistance. Sci Rep 2017;7:43517.

37. Hall AB, Newsome D, Wang Y, Boucher DM, Eustace B, Gu Y, et al.Potentiation of tumor responses toDNAdamaging therapy by the selectiveATR inhibitor VX-970. Oncotarget 2014;5:5674–85.

38. Lewis KA, Lilly KK, Reynolds EA, Sullivan WP, Kaufmann SH, Cliby WA.Ataxia telangiectasia and rad3-related kinase contributes to cell cycle arrestand survival after cisplatin but not oxaliplatin. Mol Cancer Ther 2009;8:855–63.

39. DesmetCJ, Ishii KJ.Nucleic acid sensing at the interface between innate andadaptive immunity in vaccination. Nat Rev Immunol 2012;12:479–91.

40. Dunn GP, Koebel CM, Schreiber RD. Interferons, immunity and cancerimmunoediting. Nat Rev Immunol 2006;6:836–48.

Cancer Res; 79(11) June 1, 2019 Cancer Research2946

Comb�es et al.

on January 14, 2021. © 2019 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst April 15, 2019; DOI: 10.1158/0008-5472.CAN-18-2807

Page 15: Inhibition of Ataxia-Telangiectasia Mutated and RAD3 ... · Apothicaires, Montpellier 34298, France. Phone: 334-6761-3745; Fax: 334-6761-3787; E-mail: celine.gongora@inserm.fr Cancer

2019;79:2933-2946. Published OnlineFirst April 15, 2019.Cancer Res   Eve Combès, Augusto F. Andrade, Diego Tosi, et al.   Immunity in Colorectal Cancer) Overcomes Oxaliplatin Resistance and Promotes Antitumor

ATRInhibition of Ataxia-Telangiectasia Mutated and RAD3-Related (

  Updated version

  10.1158/0008-5472.CAN-18-2807doi:

Access the most recent version of this article at:

  Material

Supplementary

  http://cancerres.aacrjournals.org/content/suppl/2019/04/13/0008-5472.CAN-18-2807.DC1

Access the most recent supplemental material at:

   

  Overview

Visual 

http://cancerres.aacrjournals.org/content/79/11/2933/F1.large.jpgA diagrammatic summary of the major findings and biological implications:

   

  Cited articles

  http://cancerres.aacrjournals.org/content/79/11/2933.full#ref-list-1

This article cites 40 articles, 6 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/79/11/2933To request permission to re-use all or part of this article, use this link

on January 14, 2021. © 2019 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst April 15, 2019; DOI: 10.1158/0008-5472.CAN-18-2807