the dna repair inhibitor dbait is specific for malignant ... · hematologic malignancies account...

12
Large Molecule Therapeutics The DNA Repair Inhibitor Dbait Is Specic for Malignant Hematologic Cells in Blood Sylvain Thierry 1 , Wael Jdey 1,2 , Solana Alculumbre 3 , Vassili Soumelis 3 , Patricia Noguiez-Hellin 1 , and Marie Dutreix 1 Abstract Hematologic malignancies are rare cancers that develop refractory disease upon patient relapse, resulting in decreased life expectancy and quality of life. DNA repair inhibitors are a promising strategy to treat cancer but are limited by their hematologic toxicity in combination with conventional che- motherapies. Dbait are large molecules targeting the signaling of DNA damage and inhibiting all the double-strand DNA break pathways. Dbait have been shown to sensitize resistant solid tumors to radiotherapy and platinum salts. Here, we analyze the efcacy and lack of toxicity of AsiDNA, a cholesterol form of Dbait, in hematologic malignancies. We show that AsiDNA enters cells via LDL receptors and activates its molec- ular target, the DNA dependent protein kinase (DNA-PKcs) in 10 lymphoma and leukemia cell lines (Jurkat-E6.1, MT-4, MOLT-4, 174xCEM.T2, Sup-T1, HuT-78, Raji, IM-9, THP-1, and U-937) and in normal primary human PBMCs, resting or activated T cells, and CD34 þ progenitors. The treatment with AsiDNA induced necrotic and mitotic cell death in most cancer cell lines and had no effect on blood or bone marrow cells, including immune activation, proliferation, or differentiation. Sensitivity to AsiDNA was independent of p53 status. Survival to combined treatment with conventional therapies (etoposide, cyclophosphamides, vincristine, or radiotherapy) was analyzed by isobolograms and combination index. AsiDNA synergized with all treatments, except vincristine, without increasing their toxicity to normal blood cells. AsiDNA is a novel, potent, and wide-range drug with the potential to specically increase DNA- damaging treatment toxicity in tumor without adding toxicity in normal hematologic cells or inducing immune dysregulation. Mol Cancer Ther; 16(12); 281727. Ó2017 AACR. Introduction Hematologic malignancies account for approximately 10% of all newly diagnosed neoplasms. Their common characteristic is the presence of chromosomal translocations, a trait not typically observed in solid tumors (1). This, among other characteristics, makes hematologic malignancies a unique class of neoplasms resulting in a unique set of challenges for treatment and the prevention of relapse. A major underlying complication of hema- tologic malignancies is the development of refractory disease upon patient relapse. DNA repair systems that can reverse the cytotoxicity of many clinically used DNA-damaging agents are among the more widely studied targets to overcome tumor resistance. Targeting DNA repair for cancer therapy has been well validated in solid tumor oncology. PARP inhibitors are the most advanced repair inhibitors, but their apparent hematology toxic- ity (24) could limit their use in treatment of hematologic neoplasia. Human cancers typically arise after a long process of random mutations, accompanied by continual selection of more rapidly adapting and proliferating tumor cells. Several of the founder mutations that lead to cancers involve genetic changes in key DNA repair pathways (5). In addition, epigenetic changes via DNA methylation and/or histone methylation and acetylation at the site of DNA repair genes are found in numerous cancers (6). Thus, cancer treatments that target a specic DNA repair defect can be selectively toxic (lethal) to cancer cells, with various DNA repair capacities, while sparing normal (DNA repair procient) tissues. Genetic instability plays a critical role in inherited and sporadic leukemia and is a common feature of blood malignancy. Chro- mosomal abnormalities in chronic lymphocytic leukemia (CLL) are detected in up to 80% of patients (7). In general, clonal chromosome aberrations are found in approximately half of acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS) patients (8). Moreover, several mutations in repair genes are responsible for hereditary lymphoma susceptibility disorders, including ataxia-telangiectasia (A-T), Nijmegen breakage syn- drome (NBS), and Bloom syndrome. In the absence of a genetic predisposition, exposure to toxins or immune system alterations (e.g., such as those that occur in acquired immunodeciency syndrome) may interfere with the mechanisms that assure genetic safety by resolving DNA breaks. Inherited DNA repair defects that predispose individuals to cancer, such as mutations of the ataxia telangiectasia mutated (ATM) gene, are found in several types of cancer, including acute leukemia (9). CLL is another hematologic malignancy with a poor prognosis in which there are mutations in the ATM gene. Such CLL cells have homologous recombination (HR) defects and are amenable to therapy with PARP1 inhibitors (10). AML are sensitive to PARP1 inhibition because of their 1 Institut Curie, PSL Research University, CNRS UMR 3347, INSERM U1021, Paris- Sud University, Orsay, France. 2 DNA-Therapeutics, Onxeo, Paris, France. 3 Insti- tut Curie, PSL Research University, INSERM U932, Paris, France. Note: Supplementary data for this article are available at Molecular Cancer Therapeutics Online (http://mct.aacrjournals.org/). Corresponding Author: Marie Dutreix, Institut Curie, Centre Universitaire, Bati- ment 112, Orsay 91405, France. Phone: 331-6986-7186; Fax: 331-6986-3141; E-mail: [email protected] doi: 10.1158/1535-7163.MCT-17-0405 Ó2017 American Association for Cancer Research. Molecular Cancer Therapeutics www.aacrjournals.org 2817 on January 24, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from Published OnlineFirst September 25, 2017; DOI: 10.1158/1535-7163.MCT-17-0405

Upload: others

Post on 26-Sep-2020

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: The DNA Repair Inhibitor Dbait Is Specific for Malignant ... · Hematologic malignancies account for approximately 10% of all newly diagnosed neoplasms. Their common characteristic

Large Molecule Therapeutics

The DNA Repair Inhibitor Dbait Is Specific forMalignant Hematologic Cells in BloodSylvain Thierry1,Wael Jdey1,2, Solana Alculumbre3, Vassili Soumelis3,Patricia Noguiez-Hellin1, and Marie Dutreix1

Abstract

Hematologic malignancies are rare cancers that developrefractory disease upon patient relapse, resulting in decreasedlife expectancy and quality of life. DNA repair inhibitors are apromising strategy to treat cancer but are limited by theirhematologic toxicity in combination with conventional che-motherapies. Dbait are large molecules targeting the signalingof DNA damage and inhibiting all the double-strand DNAbreak pathways. Dbait have been shown to sensitize resistantsolid tumors to radiotherapy and platinum salts. Here, weanalyze the efficacy and lack of toxicity of AsiDNA, a cholesterolform of Dbait, in hematologic malignancies. We show thatAsiDNA enters cells via LDL receptors and activates its molec-ular target, the DNA dependent protein kinase (DNA-PKcs) in10 lymphoma and leukemia cell lines (Jurkat-E6.1, MT-4,MOLT-4, 174xCEM.T2, Sup-T1, HuT-78, Raji, IM-9, THP-1,

and U-937) and in normal primary human PBMCs, resting oractivated T cells, and CD34þ progenitors. The treatment withAsiDNA induced necrotic and mitotic cell death in most cancercell lines and had no effect on blood or bone marrow cells,including immune activation, proliferation, or differentiation.Sensitivity to AsiDNA was independent of p53 status. Survivalto combined treatment with conventional therapies (etoposide,cyclophosphamides, vincristine, or radiotherapy) was analyzedby isobolograms and combination index. AsiDNA synergizedwith all treatments, except vincristine, without increasing theirtoxicity to normal blood cells. AsiDNA is a novel, potent, andwide-range drug with the potential to specifically increase DNA-damaging treatment toxicity in tumor without adding toxicity innormal hematologic cells or inducing immune dysregulation.Mol Cancer Ther; 16(12); 2817–27. �2017 AACR.

IntroductionHematologic malignancies account for approximately 10% of

all newly diagnosed neoplasms. Their common characteristic isthe presence of chromosomal translocations, a trait not typicallyobserved in solid tumors (1). This, among other characteristics,makes hematologic malignancies a unique class of neoplasmsresulting in a unique set of challenges for treatment and theprevention of relapse. Amajor underlying complication of hema-tologic malignancies is the development of refractory diseaseupon patient relapse. DNA repair systems that can reverse thecytotoxicity of many clinically used DNA-damaging agents areamong the more widely studied targets to overcome tumorresistance. Targeting DNA repair for cancer therapy has been wellvalidated in solid tumor oncology. PARP inhibitors are the mostadvanced repair inhibitors, but their apparent hematology toxic-ity (2–4) could limit their use in treatment of hematologicneoplasia.

Human cancers typically arise after a long process of randommutations, accompanied by continual selection of more rapidlyadapting and proliferating tumor cells. Several of the foundermutations that lead to cancers involve genetic changes in keyDNArepair pathways (5). In addition, epigenetic changes via DNAmethylation and/or histone methylation and acetylation at thesite of DNA repair genes are found in numerous cancers (6). Thus,cancer treatments that target a specific DNA repair defect can beselectively toxic (lethal) to cancer cells, with various DNA repaircapacities, while sparing normal (DNA repair proficient) tissues.Genetic instability plays a critical role in inherited and sporadicleukemia and is a common feature of blood malignancy. Chro-mosomal abnormalities in chronic lymphocytic leukemia (CLL)are detected in up to 80% of patients (7). In general, clonalchromosome aberrations are found in approximately half of acutemyeloid leukemia (AML) and myelodysplastic syndrome (MDS)patients (8). Moreover, several mutations in repair genes areresponsible for hereditary lymphoma susceptibility disorders,including ataxia-telangiectasia (A-T), Nijmegen breakage syn-drome (NBS), and Bloom syndrome. In the absence of a geneticpredisposition, exposure to toxins or immune system alterations(e.g., such as those that occur in acquired immunodeficiencysyndrome)may interfere with themechanisms that assure geneticsafety by resolving DNA breaks. Inherited DNA repair defects thatpredispose individuals to cancer, such as mutations of the ataxiatelangiectasia mutated (ATM) gene, are found in several types ofcancer, including acute leukemia (9). CLL is another hematologicmalignancywith a poor prognosis inwhich there aremutations inthe ATM gene. Such CLL cells have homologous recombination(HR) defects and are amenable to therapy with PARP1 inhibitors(10). AML are sensitive to PARP1 inhibition because of their

1Institut Curie, PSL Research University, CNRS UMR 3347, INSERM U1021, Paris-Sud University, Orsay, France. 2DNA-Therapeutics, Onxeo, Paris, France. 3Insti-tut Curie, PSL Research University, INSERM U932, Paris, France.

Note: Supplementary data for this article are available at Molecular CancerTherapeutics Online (http://mct.aacrjournals.org/).

Corresponding Author: Marie Dutreix, Institut Curie, Centre Universitaire, Bati-ment 112, Orsay 91405, France. Phone: 331-6986-7186; Fax: 331-6986-3141;E-mail: [email protected]

doi: 10.1158/1535-7163.MCT-17-0405

�2017 American Association for Cancer Research.

MolecularCancerTherapeutics

www.aacrjournals.org 2817

on January 24, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst September 25, 2017; DOI: 10.1158/1535-7163.MCT-17-0405

Page 2: The DNA Repair Inhibitor Dbait Is Specific for Malignant ... · Hematologic malignancies account for approximately 10% of all newly diagnosed neoplasms. Their common characteristic

intrinsic genomic instability, and PARP1 inhibition has beenfound to increase the cytotoxicity of alkylating agents in thisleukemia (11). However, the high toxicity of the combinedtreatment, mainly myelosuppression (neutropenia and throm-bocytopenia), was shown to limit the dose in various trialspreventing combined treatment to achieve a complete tumorresponse (12)

Most conventional cancer treatments, consisting of radio- orchemotherapy, target DNA to induce genotoxicity. However,these chemotherapeutic drugs also target normal proliferatingcells in naturally proliferating tissues. These tissues, which includebone marrow, gastrointestinal tract, liver, and hair follicles, arealso sensitive to DNA-targeting treatments. This leads to doselimitations andmay limit the success of treatment. The differencebetween treatment efficacy on malignant cells and toxicity innormal tissues determines the so-called "therapeutic index." Thekey to effective cancer treatment is to target tumor cells whilesparing normal tissue. This has led to the idea that the search forcures should focus on "disrupting the broader biological path-ways that support cancer growth" (13). Given that DNA repairdefects are early drivers of many cancers, there is great interest indeveloping therapeutics that exploits these potential weaknesses.We have designed a new strategy called Dbait, which uses shortDNA molecules, to inhibit all DNA repair pathways involved indouble-strand (DSB) and single-strand break (SSB) repair. Dbaitbind and activate DNA damage signaling enzymes, PARP, andDNA-dependent kinase (DNA-PK). Once activated, the twoenzymes modify their targets, generating a false DNA damagesignal. This inhibits recruitment of key repair proteins, such asRad51, Nbs1, Mre11, 53BP1, BRCA1, and XRCC1, to the site ofdamage (14, 15), (15, 16). Thus, repair enzymes do not aggregateat damage site and do not form foci after irradiation in Dbait-treated cellswith a pannuclear phosphorylation ofH2AX inducedby DNA-PK activation. Repair of single-strand and double-strandDNA breaks [via SSB repair (17), nonhomologous end joining(15), and homologous recombination (18)] is impaired inDbait-treated cells. AsiDNA is the clinical form of Dbait. It is composedof the active part of Dbait, a stable 32-bp long double-strandedDNAwith a blunt end, covalently bound to a cholesterol, to allowmolecules to enter the cells without the help of transfectionvectors. AsiDNA (called DT01 for the first-in-man trial) wasrecently tested in a phase I clinical trial on melanoma skinmetastases in association with radiotherapy (19). The resultsdemonstrated the lack of toxicity of locally administered mole-cules and suggested an increase in radiotherapy efficacy. AsiDNAactivitywas never tested in vitro. Here, wedemonstrate its ability toenter hematologic cancer cells and treat lymphoma and leukemiawithout toxic effects on hematopoietic cells. We explore thespecificity of the uptake and target activation and toxicity ofAsiDNA in lymphoma and leukemia models as well as in normalhuman blood cells.

Materials and MethodsCulture of malignant hematologic cell lines

Ten hematologic malignant cell lines including THP-1 and U-937myeloid leukemia cells; Jurkat-E6.1,MOLT-4, 174xCEM.T2mandMT4 acute CD4þ T-cell leukemia; Raji and IM-9 B-cell Burkittlymphoma, and HuT-78 CD4þ T-cell cutaneous lymphoma andSupT-1 CD4þ T-cell pleural lymphoma were purchased from theATCC (Raji, CCL-86; IM-9, CCL-159; HuT-78, CRMB-TIB-161;

Sup-T1, CRL-1942; Jurkat-E6.1, TIB-152; MOLT-4, CRL-1582;174xCEM.T2, CRL-1992; U-937, CRL-1593.2, and THP-1, TIB-202 respectively), except for MT-4, which was a kind gift fromDr.Olivier Delelis (CNRS UMR-1021, ENS de Cachan, France). Celllines were authenticated at the beginning and at the end of thestudy by short tandem repeat profiling (Geneprint 10, Promega)at 9 different loci (TH01, D5S818, D13S317, D7S820, D16S539,CSF1PO, AMEL, vWA, and TPOX). Cell lines were verified to benegative for mycoplasma contamination using the VenorGeMAdvance Kit (Biovalley). Cells were grown according to thesupplier's instructions in RPMI1640medium supplemented with10% FBS and 1%penicillin/streptomycin at 37�C in a humidifiedatmosphere at 5% CO2. Reagents for cell culture were purchasedfrom Gibco Invitrogen.

Culture of primary blood-derived cellsPeripheral blood mononuclear cells (PBMC) were isolated

from the blood of healthy donors (Etablissement Francais duSang Cabanel, Paris, France). Written informed consent to use thecells for research was obtained from each donor. PBMCs wereextracted by Ficoll–Hypaque density gradient, washed twice inRPMI1640medium, anddirectly cultured. The isolation of restingCD4þ T cells, negatively selected from PBMCs, was performedusing the Isolation Kit from Miltenyi Biotec according to themanufacturer's protocol with the addition of bead-conjugatedantibodies against both early (CD69 and CD25) and late (HLA-DR) activation markers. Cells were depleted using LS columns asrecommended by the manufacturer (Miltenyi Biotec) with apurity >98%. When necessary, CD4þ T cells were activated usingthe T-Cell Activation/Expansion Kit from Miltenyi Biotec andexpanded in culture for 10 days. Resting and activated CD4þ Tcells were cultured in RPMI1640 supplemented with 10%humanserum (Gibco Invitrogen) and 10 to 20 ng/mL rh-IL2. Plasmo-cytoid dendritic cells (pDC) were purified as described previously(20). Briefly, PBMCs were isolated using Ficoll gradient (Amer-sham). The pDCs were isolated by flow cytometry (21) asLin�CD11c�CD4þ cells with a purity >98%. The cells werecultured at a density of 106 cells/mL in complete RPMIGlutaMAX(Gibco) containing 10% FBS (HyClone).

Dbait moleculesAsiDNA is the clinical form of Dbait, a 64-nucleotide (nt)

oligodeoxyribonucleotide consisting of two complementary 32-nt strands, connected through a 1,19-bis(phospho)-8-hydraza-2-hydroxy-4-oxa-9-oxo-nonadecane linker, with a cholesterol at the50-end and three phosphorothioate internucleotide linkages ateach of the 50 and the 30 0 ends (Agilent). The sequence is: 50- XGsCsTs GTG CCC ACA ACC CAG CAA ACA AGC CTA GA L -CLTCT AGG CTT GTT TGC TGG GTT GTG GGC AC sAsGsC -30,where L is an amino linker, X a cholesteryl tetraethyleneglycol, CL

a carboxylic (hydroxyundecanoic) acid linker and s a phosphor-othioate linkage. The CpG-Dbait molecule only differs fromAsiDNA by its sequence, which includes seven CpGs (in bold):50-X AsCsGs CAC GGG TGT TGG GTC GTT TGT TCG GAT CT L -CL AGA TCC GAA CAA ACG ACC CAA CAC CCG TsGsCs GT-30

(Agilent).

Chemicals and antibodies4-hydroxycyclophosphamide (the active form of the cyclo-

phosphamide prodrug) was purchased from Santa Cruz Bio-technology SA (#sc-206885) and dissolved in cell culture

Thierry et al.

Mol Cancer Ther; 16(12) December 2017 Molecular Cancer Therapeutics2818

on January 24, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst September 25, 2017; DOI: 10.1158/1535-7163.MCT-17-0405

Page 3: The DNA Repair Inhibitor Dbait Is Specific for Malignant ... · Hematologic malignancies account for approximately 10% of all newly diagnosed neoplasms. Their common characteristic

medium to make a stock solution. Etoposide (#E1383) andvincristine sulfate (#V8388) were both purchased from Sigma-Aldrich. Etoposide was dissolved in DMSO to a stock concen-tration of 500 mmol/L. All chemicals were directly dissolved inthe appropriate solvent before the addition of one tenth of thefinal volume to the cell culture medium. Anti-LDL receptor(#ab60107), anti-LDL receptor Alexa-Fluor-488 (#ab636977),anti-calreticulin PE (#ab83220), and anti-H2AX-pS139 Alexa-Fluor-647 (#ab560447) were purchased from Abcam. Anti-CD4 APC (#130-098-160), anti-CD69 FITC (#130-098-901),anti-CD25 PE (#130.098.861), anti-HLA-DR PerCP (#130-098-179), anti-CD80 PE (#130-099-200), anti-CD83 APC (#130-098-889), and anti-CD86 FITC (130-098-182) were purchasedfrom Miltenyi Biotec. Anti-CD80 PECy7 (#305217), anti-CD86PECy5 (#305407), was purchased from eBioscience.

Cell survival and proliferationRelative cell survival was measured by the mitochondrial MTT

3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide)colorimetric assay or live-cell monitoring. MTT assay was mod-ified for cells in suspension as described previously (22). Briefly,one tenth of the culture volume ofMTT solution [5mg/mL in PBSwas added to the cultures and the cells incubated at 37�C in 5%CO2 for 30 minutes to 4 hours (depending on the cells)]. Cellswere lysed by adding one culture volume of lysis buffer (50%dimethylformamide, 20% SDS, and 0.22% acetic acid) directly tothe medium and incubated for 2 hours at 37�C before reading at550 nm (Victor-X3, PerkinElmer). For proliferation studies, cellswere cultured in 96-well plates: the MT-4, MOLT-4, 174xCEM.T2,Jurket-E6.1, Sup-T1,U-937, IM-9, andRaji cell lineswere seeded at2� 105 cells/mL,HuT-78 at 4� 105 cells/mL, and THP-1, PBMCs,and activated and resting CD4þ T cells at 106 cells/mL. For thecombined treatments, AsiDNA was added to the cell cultures 24hours before the additionof other drugs or irradiation.MTTassayswere performed 4 days after the last treatment except for irradia-tions that were performed 6 days after. Cell proliferation and celldeath were monitored after staining with 0.4% trypan blue(Sigma Aldrich) by visual counting using a Burker chamber. Cellsurvival was calculated as the ratio of living treated cells to livingmock-treated cells. Cell death was calculated as the number ofdead cells divided by the total number of cells.

Colony-forming units of granulocytes and/ormacrophages assay

The colony-forming units of granulocytes and/or macrophages(CFU-GM) assay was performed as described previously (23, 24).Briefly, 2,000 human blood CD34þ progenitors from bone mar-row (Hu-BM-CD34þ, #70002.3, Stemcell Technologies) wereplated for methylcellulose assays in triplicate in 1.5 mL Metho-Cult (H4535-Enriched-without-EPO, #04535, Stemcell Technol-ogies) in a 35-mm dish with or without AsiDNA. Cells werecultured for 14 days before colonies were scored for proliferationand differentiation. Differentiation was based on cell shape andproliferation estimated by counting colonies according to themanufacturer's protocol. Results are expressed as thepercentage ofdifferentiated cells and colonies following treatment relative tothose of cells that were not.

Annexin V/propidium iodide assayCell death analyses was performed on 106 living cells harvested

and resuspended in 100 mLMACS buffer in the presence of a 1:50

dilution of Annexin V-APC (Miltenyi Biotec), for 20 minutesat room temperature. The viability dye, propidium Iodide(from Sigma Aldrich), was then added to a final concentrationof 5 mg/mL before flow cytometry analyses.

Analysis of immune cell activationThe membrane activation markers CD69 and CD25, for CD4þ

T cells, and CD80 and CD86, for pDCs, were analyzed by flowcytometry according to the manufacturers' protocols. Flow cyto-metry analysis was performed on a FACSCalibur, Aria III, or LSRII(BD- Biosciences). At least 10,000 events were collected. All datawere analyzed using FlowJo software (TreeStar). Cytokine secre-tion was quantified by ELISA for human IFNg (#SIF50, R&DSystems) according to the manufacturer's protocol.

Regression trendlines, EC50, and synergy calculationsNonlinear regression trendlines were generated using Graph-

Pad (Prism) software. The EC50 was calculated as the inflectionpoint of the curves to generate Hill equation values, taking intoaccount the standalone effect of the second drug for the drugcombinations. Synergy, additivity, and antagonism were eval-uated by the dose reduction index (DRI); combination index,according to the Lowe nonlinear regression model (CILowe);and Chou linear regression model (CIChou) as described pre-viously (25). The DRI was calculated as the ratio between theEC50 obtained for the combination compared with that of thesingle agents alone, expressed as the fold change. The CILowe,CIChou, and isobolograms were calculated using the SynerDruginterface based on R software as described previously. Drug–drug interactions can be additive (DRI¼ 1; CIChou ¼ 1; CILowe ¼0), antagonistic (DR1 < 1; CIChou > 1; CILowe > 0), or synergistic(DRI > 1; CIChou < 1; CILowe < 0). Isobolograms were calculatedas described previously (26) and consist of two parts: a two-dimensional scatterplot of the combinations of inhibitoryconcentrations and an envelope of additivity determined bytwo possible additive mechanisms, the first (mode-I) implyingindependent effects of the two agents and the second (mode-II)implying an interaction between the effects of the two agents: ifthe combined effect of the two agents falls within this envelope,their effects are additive; if it is below, they are synergistic; andif it is above, they are antagonist.

Statistical analysisComparisons between different conditions in experiments

were performed using the Wilcoxon test (nonparametric pairedtest). Correlations were evaluated using the Spearman test (non-parametric test). P < 0.05 was considered to be significant for allstatistical tests applied.

AvailabilitySynerDrug interface based on R software Maps is an open-

source collaborative initiative available in the Institut Curierepository (https://github.com/bioinfo-pf-curie/synerdrug), asdescribed previously (25)

ResultsAsiDNA is toxic to malignant hematologic cells

We tested the toxicity of AsiDNAmonotherapy on a wide rangeof hematologic cancer cells, including two myeloid leukemias,

Toxicity and Efficacy of Dbait on Hematologic Cells

www.aacrjournals.org Mol Cancer Ther; 16(12) December 2017 2819

on January 24, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst September 25, 2017; DOI: 10.1158/1535-7163.MCT-17-0405

Page 4: The DNA Repair Inhibitor Dbait Is Specific for Malignant ... · Hematologic malignancies account for approximately 10% of all newly diagnosed neoplasms. Their common characteristic

four acute T-cell leukemias, two Burkitt B-cell lymphomas, andtwo T-cell lymphomas (Table 1). We evaluated the half maximaleffective concentration (EC50) of AsiDNA for all cell lines bymeasuring the survival 4 days after treatment with various doses,ranging from 0.16 to 48 mmol/L, the maximum concentrationclose to the level observed in monkeys after intravenous injection(Fig. 1A and B). AsiDNA was toxic for eight of the 10 tested celllines, with EC50s inferior to 16mmol/L for themost sensitive (class"S": U-937, IM-9, MOLT-4, and Sup-T1) and ranging from 16 to48 mmol/L for the cells with intermediate sensitivity (class "I":MT4, Jurkat-E6.1, 174xCEM.T2, and HuT-78). Two cell lines wereresistant up to the maximum tested dose of 48 mmol/L (class "R":Raji and THP1; Table 1). Similar results were obtained with thecolorimetric MTT assay (Fig. 1), or direct counting of viable cellsby trypan blue enumeration (Supplementary Fig. S1).

Sensitivity to AsiDNA was independent of the cellular subset(myeloid, B cells, T cells) and the type of disease, either leukemiaor lymphoma (Fig. 1; Table 1). Unexpectedly, the efficacy ofAsiDNA in the various cell lines did not appear to depend onthe level of p53 activity, as themost sensitive (U-937 and Sup-T1)and resistant cells (Raji and THP-1) were all p53 deficient P ¼0.3387, ns).

AsiDNA is not toxic to normal peripheral blood cellsAsiDNAwas toxic tomost of the cancer blood cells tested. Thus,

we wanted to gain further insights on its effects on normal bloodcells. We used fresh whole PBMCs from healthy donors to testglobal blood cell toxicity. We first performed an in vitro AsiDNAdose escalation study similar to that performed for the cancer cells.No significant toxicity was observed at any tested dose, withmorethan 80% survival relative to nontreated cells at the highest doseof 48 mmol/L (Fig. 1B). We tested whether cell cycling is requiredfor AsiDNA toxicity by analyzing the fate of resting T cells (CD4r),as well as their activated and cycling ex vivo counterpart (CD4a),for up to 11 days after treatment and observed no toxicity at anytime point (Fig. 1B).

We analyzed the effect of AsiDNA on bonemarrow progenitorsusing the ex vivo CFU-GM assay to confirm its lack of toxicity tonormal cells. CFU-GM assays are generally used to investigate thedirect adverse effects of xenobiotics on the proliferative anddifferentiation capacities of blood-forming multipotential stemcells (CD34þ), depending on the humoral growth factors andlocal cytokines in their specific microenvironment (23, 24). Wetreated cellswith various doses of AsiDNA from0.48 to 48mmol/Land analyzed their ability to formcolonies, either differentiated ornot, based on their morphology. There was no statistically sig-nificant decrease either in proliferation or differentiation underthese conditions (Fig. 1C), indicating the absence of acute ordelayed toxicity of Dbait on bone marrow human progenitors atdoses sufficient for antitumor efficacy in most leukemia andlymphoma tested cell lines (Fig. 1A). These results are in accordwith the absence of toxicity found in mice after systemic admin-istration of AsiDNA (personal data).

AsiDNA triggers programmed necrotic and mitotic death inmalignant hematologic cells

The observed effects of AsiDNA on malignant hematologiccells were related to the DNA portion of the molecule, ascholesterol alone did not induce any toxicity in the same rangeof doses (Fig. 2A). This confirms that the effect is due to thecapacity of AsiDNA to mimic DSBs. The decrease in the number Ta

ble

1.Effects

ofAsiDNAonhe

matologic

maligna

ntcelllines

p53

Doub

ling

AsiDNAstan

dalone

þEtoposide

þCyclopho

spha

mide

þIrrad

iations

þVincristine

Nam

estatus

time(h)

EC50(m

mol/L)

Sensitivity

Combo

CI Lowe

CI Chou

Combo

CI Lowe

CI C

hou

Combo

CI Lowe

CI C

hou

Combo

CI Lowe

CI Chou

Raji

M,D

EL

18>4

8R

Add

�0.046

0.943

Syn

�0.377

0.432

-nd

ndSyn

�0.111

0.817

IM-9

WT

1912.76

SSyn

�0.251

0.354

Syn

�0.245

0.575

Syn

�0.10

20.842

Add

�0.064

0.914

HuT

-78

M,S

TOP

3629

.28

ISyn

�0.430

0.461

Syn

�0.676

0.255

Syn

�0.256

0.561

Add

0.034

1.013

Sup

-T1

MM

207.57

5S

Syn

�0.219

0.446

Add

�0.021

0.943

Syn

�0.18

50.674

Add

�0.052

0.923

Jurkat-E6.1

MM

2424

.22

IAdd

�0.010

0.968

Syn

�0.254

0.376

Syn

�0.13

40.801

Add

0.043

1.026

MOLT

-4WT

249.756

SSyn

�0.203

0.666

Syn

�0.266

0.482

Add

�0.033

0.942

Syn

�0.117

0.811

MT-4

WT

2219.64

ISyn

�0.265

0.542

—nd

nd—

Nd

ndAdd

0.039

1.096

174xC

EM.T2

WT

1821.21

ISyn

�0.468

0.461

Syn

�0.309

0.405

Syn

�0.18

60.628

Add

�0.072

0.893

U-937

M,D

EL

200.899

SSyn

�0.485

0.322

Syn

�0.386

0.311

—nd

ndAdd

�0.043

0.949

THP-1

M48

>48

R—

ndnd

—nd

nd—

ndnd

—nd

nd

NOTE:P53

status

(M,singlemutationa

leve

ntinthetumor;MM,m

ultiplemutations;STOP,stopmutation;DEL,deletion).Sen

sitivity

totrea

tmen

tswithAsiDNA:I:intermed

iate,S:sen

sitive

,R:resistant;Syn

:syn

ergic,A

dd:

additive;

models:CI Lowe:L

oweno

nlinea

rregression,

CI Chou:C

houlinea

rregression.

Thierry et al.

Mol Cancer Ther; 16(12) December 2017 Molecular Cancer Therapeutics2820

on January 24, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst September 25, 2017; DOI: 10.1158/1535-7163.MCT-17-0405

Page 5: The DNA Repair Inhibitor Dbait Is Specific for Malignant ... · Hematologic malignancies account for approximately 10% of all newly diagnosed neoplasms. Their common characteristic

of living cells was associated with a corresponding increase inthe number of dead cells, characteristic of the cytotoxic activityof AsiDNA, rather than an arrest of division. The absence ofa perturbation of the cell cycle was confirmed by analysisof the distribution of the treated cells in the cycle (sensitiveor resistant to Dbait), which was unaffected after 12, 24, or48 hours of Dbait treatment, regardless of the dose (Supple-mentary Fig. S2).

However, a global increase of cells in the G2 phase started toappear after 3 days of treatment and increased by the fourth day incell lines that displayed sensitivity to AsiDNA (Fig. 2B). Thisincrease of cells in G2 was concomitant with cell death. The mostsensitive cells (U-937) showed the strongest increase in theproportion of cells in G2. In contrast, the resistant and interme-diate sensitive cell lines, which did not die at these doses ofAsiDNA did not show any short- or long-term variations of thecell cycle (Fig. 2B). By 4 days of treatment, the number ofpolyploid cells and those in the sub-G0–G1 subset, increasedslightly, but this increase was significant only for p53-proficientIM-9 cells (Fig. 2B). Annexin V/propidium analysis after 4 days oftreatment revealed a large increase of necrosis and, to a lesserextent, late apoptosis in cells treated with AsiDNA (Fig. 3C).Cholesterol, without the DNA moiety to activate DNA-PK, hadno effect on cell survival or the cell cycle. Calreticulin exposure atmembrane of dying cells (ecto-CRT) is observed in all the cellstreated by AsiDNA in a dose-dependent manner as amarker of anendoplasmic reticulum stress and programmed cell death (27).Altogether, these results strongly suggest that AsiDNA inducesmitotic cell death in malignant hematologic cells independentlyof their p53 status.

Cell membrane LDL receptors control AsiDNA uptakeThe lack of sensitivity of normal and resistant cells could be

due to the poor uptake of AsiDNA by these cells. We analyzedintracellular AsiDNA levels 24 hours after treatment usingCy5.5-labeled fluorescent AsiDNA (Fig. 3A). All cell lines dis-played AsiDNA uptake with 80% to 100% showing Cy5.5fluorescence. The individual cell fluorescence intensity variedlittle among different cells in the same cell culture. However,the mean value of fluorescence intensities (MFI) varied by up to10-fold between cell lines, indicating that all cell lines canuptake AsiDNA, but with different efficiencies. The MFI did notsignificantly reflect the variation of cell size (Spearman correla-tions: cell volume/Cy5.5 MFI, P ¼ 0.1237 ns). AsiDNA cellularuptake is promoted by its cholesterol moiety (28). We thusevaluated the cell membrane concentration of the low-density

Figure 1.Survival of malignant hematologic and normal human primary immune cells inresponse to AsiDNA dose escalation. Cell survival was determined byMTT assayafter 4 days of treatment. A, Malignant hematologic cells were classifiedaccording to their sensitivity to AsiDNA. Sensitive (green): U-937 (cross), Sup-T1(asterisk), MOLT-4 (diamond), IM-9 (triangle); intermediate (blue): Jurkat-E6.1(circle), MT-4 (square),174xCEM.T2 (rectangle), HuT-78 (star); resistant (red):Raji (triangle), THP1 (cross). Data are the mean of at least six independentexperiments. B, Normal human primary blood cells: PBMCs (black diamonds),activated CD4 T cells (blue circles), and resting CD4 T cells (red squares). Cellsurvival was determined by MTT assay after 4 (plain lines), 7 (dashed lines), and11 days of treatment (dotted lines). Data are the mean of three to fourindependent experiments with cells from different donors. C, CFU-GM assay onbone marrow stem cells. Proliferation (squares) and differentiation (diamonds)were assayed after 14 days of treatmentwithAsiDNA. Data are themean of threeto four independent experiments. Error bars, SD.

Toxicity and Efficacy of Dbait on Hematologic Cells

www.aacrjournals.org Mol Cancer Ther; 16(12) December 2017 2821

on January 24, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst September 25, 2017; DOI: 10.1158/1535-7163.MCT-17-0405

Page 6: The DNA Repair Inhibitor Dbait Is Specific for Malignant ... · Hematologic malignancies account for approximately 10% of all newly diagnosed neoplasms. Their common characteristic

lipoprotein receptor (LDL-R), which plays a key role in cho-lesterol-rich LDL endocytosis. The LDL-R is differentiallyexpressed in malignant hematologic cells (29, 30), and itsmembrane concentration varied by approximately 10-foldwithin our set of cell lines (Fig. 3A). The respective mean

concentration of AsiDNA and the level of LDL-R at the mem-brane were highly correlated (Spearman P < 0.0001; Fig. 3C).

LDL-R expression is induced by IL2 in NK cells (31). Here,the level of LDL receptors at the membranes of T cells activatedby IL2 and anti-CD3/CD28 TCR were 5-fold higher than in

Figure 2.

AsiDNA triggers necrotic and mitotic death of malignant cells. Four cell lines were treated with cholesterol or AsiDNA at 4.8 mmol/L for Sup-T1, MOLT-4, and IM-9,or 0.48 mmol/L for the highly sensitive U-937. A, Proliferation after 2, 3, and 4 days of treatment measured by counting live (solid black) and dead (grey)cells in untreated sample (circles) or treated with cholesterol (triangles) or AsiDNA (squares). Flow cytometry analysis.B,Cell cycle after 2, 3, or 4 days of treatment:percentage of cells in polyploidy (black), G2–M (dark gray), S (light gray), G0–G1 (white), and sub-G0–G1 (hatched). C, Cell death after 4 days of treatmentmonitored by Annexin V/propidium iodide (AV/PI) staining: early apoptosis (AVþ/PI�, white), late apoptosis (AVþ/PIþ, hatched), or necrosis (AV�/PIþ, black).Results are the mean of three to four independent experiments. Error bars, SD. D, Membrane calreticulin exposure (ecto-CRT) after 3-day AsiDNA treatment:nontreated (gray), treated with AsiDNA 1.6 mmol/L (dashed line), 4.8 (dotted line), and 16 mmol/L AsiDNA (plain line). One representative experiment out ofthree is shown.

Thierry et al.

Mol Cancer Ther; 16(12) December 2017 Molecular Cancer Therapeutics2822

on January 24, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst September 25, 2017; DOI: 10.1158/1535-7163.MCT-17-0405

Page 7: The DNA Repair Inhibitor Dbait Is Specific for Malignant ... · Hematologic malignancies account for approximately 10% of all newly diagnosed neoplasms. Their common characteristic

resting T cells (Fig. 1A). This increase was associated with asimilar increase in AsiDNA uptake (Fig. 3A and C). The acti-vation of DNA-PK, revealed by the extent of H2AX phosphor-ylation, was also higher in activated than resting cells (Fig. 3B).However, this increase was not associated with an increase ofsensitivity to AsiDNA, as resting and activated T cells weresimilarly resistant to AsiDNA treatment (Fig. 1A). The resistanceto AsiDNAmay reflect the incapacity of the cells to trigger DNA-PK activation, one of the first steps required for AsiDNA/Dbaitefficacy (14). However, there was no correlation between thelevel of phosphorylated H2AX and intracellular AsiDNA levels(Fig. 3D). Moreover, neither AsiDNA cell uptake (Supplemen-tary Fig. S3A and S3B) nor activation of H2AX phosphorylation(Supplementary Fig. S3C) are predictive biomarkers for sensi-tivity to AsiDNA, contrary to the micronuclei frequency (Sup-plementary Fig. S3D) that have been recently reported on breastcancer cell lines (18).

Synergistic effects of AsiDNA in combination withchemotherapies

Genotoxic chemotherapies are often used to treat leukemia andlymphomas. They consist mostly of alkylating agents, such ascyclophosphamide (4-HC), which represents the spearhead ofthis drug class, drugs targeting DNA topoisomerase II, such as thenonintercalating poison etoposide, and mitotic poisons, such asvincristine. We analyzed the effect of combining AsiDNA withthese drugs or irradiation (a treatment inducing a wide range of

DNA damage) on the various blood cancer cell lines already usedin this study.We first determined the EC50 for eachmonotherapy,using the same protocol used for AsiDNA (Fig. 4A). The cell linesdisplayed different sensitivities to conventional chemo- andradiotherapy (Fig. 4B). All chemotherapies demonstrated toxicityfor normal hematologic cells after 4 days, except for etoposides,for which toxicity was only detectable after 7 days of treatment.AsiDNA was not toxic at the concentrations tested even after 14days of culture (Figs. 5Aand 1B and C).

Various concentrations of AsiDNA were added to the cultures24 hours prior to the chemotherapy/radiotherapy to monitorits effect on conventional therapy efficacy. We determined theEC50 of conventional treatments with and without AsiDNA andanalyzed the DRI; combination index and isobolograms of thevarious combinations (Fig. 4; Supplementary Figs. S4–S7; Table1). All cell lines were sensitized by AsiDNA to at least one specifictreatment (Fig. 4B), whereas nonmalignant cells did not evenshow sensitization to the highly toxic 4-HC, confirming thespecificity of AsiDNA for cancer cells. AsiDNA strongly synergizedwith etoposide, 4-HC, and radiotherapy treatments, which main-ly act by directly inducing DNA damage, and to a much lesserextent with vincristine, which is a mitotic spindle poison (Fig. 4;Supplementary Figs. S4–S7; Table 1).

AsiDNA does not affect primary immune cell functionsMost immune cells can be activated or repressed by small

molecules. AsiDNA molecules have a DNA moiety that could

Figure 3.

Cellular uptake of AsiDNA correlateswith LDL-receptor levels but not withH2AX phosphorylation. A and B, Flowcytometry analyses were performedon the 10 malignant cell lines, PBMCs,and monocytes (Mono) in twoindependent experiments:Cy5.5-AsiDNA uptake 24 hours after4.8 mmol/L treatment (A; gray); cellsurface LDL-receptor levels (black);and percentage of g-H2AX–positivecells, relative to the negative control(B; % positive). C and D, Spearmancorrelations analysis in sensitive(circle), intermediate (square), andresistant (diamond) cell lines:Cy5.5-AsiDNA and cell surface LDL-RMFI (C), Cy5.5-AsiDNA MFI andg-H2AX % positive cells (D).

Toxicity and Efficacy of Dbait on Hematologic Cells

www.aacrjournals.org Mol Cancer Ther; 16(12) December 2017 2823

on January 24, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst September 25, 2017; DOI: 10.1158/1535-7163.MCT-17-0405

Page 8: The DNA Repair Inhibitor Dbait Is Specific for Malignant ... · Hematologic malignancies account for approximately 10% of all newly diagnosed neoplasms. Their common characteristic

activate immune cells due to their recognition by Toll-like recep-tors (TLR). AsiDNA does not contain any CpG sequence, which isknown to be the major dinucleotide site detected by the TLRs. Amolecule containing seven CpG in the DNA sequence (CpG-Dbait) was used as a positive control of immune stimulation.We assessed the ability of these molecules to trigger pDCs andCD4þ T-cell activation. Although pDCs display high levels ofsurface TLR-9 and are highly sensitive to the CpG response,AsiDNA had no effect on pDC. In contrast, CpG-Dbait triggeredupregulation of the dendritic cell-surface activation markersCD80 and CD86 between 1 and 3 days after treatment, whichdecayed by the fourth day (Fig. 5C; Supplementary Fig. S8). IFNasecretion by the pDCs (assayed by ELISA) peaked after two days ofDbait-CpG treatment (Supplementary Fig. S8) but no significantvariation after AsiDNA treatment (data not shown). We analyzedthe effect of AsiDNA on resting CD4þ T cells (Fig. 5A) and their exvivo activated and cycling counterparts (Fig. 5B). Treatment ofresting T cells with AsiDNA or CpG-Dbait neither induced orreduced the surface expression of CD69 or CD25 activationmarkers (Fig. 5A). No effects were observed for AsiDNA treat-

ments on activated T cells, whereas CpG-Dbait–treated cellsshowed a slight increase in the level of activation markers at thehigher doses. These findings are concordant with our previousexperiments on bonemarrow stem cell differentiation by theGM-CFU assay that showed no effects on their differentiation at anyconcentration tested (Fig. 1C).

DiscussionDbait is an innovative inhibitor of DSB and SSB repair

(14, 15). The molecules were developed and tested to increasesensitivity of solid tumors to radiation (16) and chemotherapy(32, 33). However, no tests have yet been performed in bloodcells. Here, we tested the effects of AsiDNA, the form of Dbaitmolecules used in the clinic (19), on the treatment of malig-nant blood cells and the toxicity of combined treatment tohuman blood cells. All tests were performed on human cells, asmouse blood cells have been shown to contain 50-fold lessDNA-PK, one of the main targets of Dbait, than human bloodcells (34).

Figure 4.

Synergistic effects of AsiDNA in combination with conventional therapies. EC50 of malignant cell lines (white) and normal human blood cells (blue) aftervarious treatments combined with 4.8 mmol/L AsiDNA (hatched bars) or not (white bars). A, AsiDNA single treatment. B, Etoposide, cyclophosphamide (4-HC),irradiation, and vincristine sulfate. Results are expressed as the mean of three to four independent experiments. Error bars, SD. C, DRI is the ratio of theEC50 of treatments in combination with AsiDNA or alone for etoposide (blue squares), cyclophosphamide (red triangles), irradiation (black diamonds), andvincristine sulfate (green circles). Interactions can be additive (DRI ¼ 1), antagonistic (DR1 < 1), or synergistic (DRI > 1).

Thierry et al.

Mol Cancer Ther; 16(12) December 2017 Molecular Cancer Therapeutics2824

on January 24, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst September 25, 2017; DOI: 10.1158/1535-7163.MCT-17-0405

Page 9: The DNA Repair Inhibitor Dbait Is Specific for Malignant ... · Hematologic malignancies account for approximately 10% of all newly diagnosed neoplasms. Their common characteristic

All the cells were able to take up AsiDNA, although withdifferent efficiencies. The sensitivity to AsiDNA did not correlatewith uptake of the molecule or H2AX phosphorylation. Thisdiscrepancy probably reflects the numerous parameters involvedin cell death induced by AsiDNA treatment. The fact that all thecell lines were sensitized to at least one treatment indicates thatAsiDNA uptake and activity in malignant cells is not the limitingfactor. Normal cells are fully resistant to the sensitizing effect ofAsiDNA, although they incorporate AsiDNA and activate H2AX

phosphorylation. These results suggest that specific alterations inmalignant cells are required for sensitivity to AsiDNA. The pos-sibility that P53 proficiency could protect cells from AsiDNA wasexcluded as in the three p53-proficientmalignant cells lines (IM-9,MOLT-4, MT-4, and 174-CEM), AsiDNA show a synergisticeffect in combination with at least one chemotherapy treatment.In contrast, AsiDNA alone or in combination with other treat-ments had no significant activity either in normal resting oractivated cells, eliminating proliferation as a major condition for

Figure 5.

AsiDNAdoes not affect primary immune cell functions. Activation and treatmentwith AsiDNAor CpG-Dbaitmolecules of CD4þ T cells.A–C,Resting CD4þT cells (A),activated CD4þ T cells (B), and pDCs (C). Left, one representative flow cytometry analysis; right, mean of at least three independent experiments ondifferent donors. Results are expressed as the percentage of cells for each compartment. A and B, CD69þ/CD25þ (hatched), CD69þ/CD25� (gray), CD69�/CD25þ

(black), and CD69�/CD25� (white). C, CD86þ/CD80þ (hatched), CD86þ/CD80� (gray), CD86�/CD80þ (black), and CD86�/CD80� (white). Isotype,unactivated, and activated cell controls are shown. pDC-activated controls were treated by CpG oligonucleotides. Error bars, SD.

Toxicity and Efficacy of Dbait on Hematologic Cells

www.aacrjournals.org Mol Cancer Ther; 16(12) December 2017 2825

on January 24, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst September 25, 2017; DOI: 10.1158/1535-7163.MCT-17-0405

Page 10: The DNA Repair Inhibitor Dbait Is Specific for Malignant ... · Hematologic malignancies account for approximately 10% of all newly diagnosed neoplasms. Their common characteristic

responsiveness to AsiDNA. Jdey and colleagues recently demon-strated in breast cancer cell lines that genetic instability revealedby transcriptional defects in DNA repair pathways and cell-cyclecontrol, micronuclei formation, and large-scale chromosomerearrangements predicts sensitivity to AsiDNA (18). Our resultsindicate that genetic instability could also be a biomarker ofAsiDNA sensitivity in hematologic malignancies. Indeed, micro-nuclei frequency was found to correlate with the sensitivity of thehematologic cell lines to AsiDNA (Supplementary Fig. S3D).Moreover, the most resistant cell line to AsiDNA, THP1, is con-sidered to be the most physiologic model of the monocytephagocyte lineage and can differentiate into macrophages ormoDCs in response to cytokine treatment.

White blood cells are highly prone to enter apoptosis whenexposed to DNA-damaging agents. However, we only observeda moderate increase (from 2 to 5-fold) of late apoptotic cells.Even p53-proficient IM-9 cells did not show higher levels ofapoptosis than other p53-deficient cells, further eliminating apotential role of apoptotic cell death in the sensitivity of cancercells to AsiDNA. The death induced by AsiDNA was delayed,detectable only at late stages, stochastic, and asynchronous, aswe did not observe a large drop in the number of viable cells atany time.

AsiDNA sensitized cells to etoposide, 4-HC, and radiother-apy, which act by inducing chromosomal DNA damage, but notto the mitotic spindle poison vincristine. This result confirmsthat AsiDNA sensitizes liquid tumors to treatments by inhibit-ing the repair of DNA damage induced by the associatedtreatments, as it does in solid tumors. One of the most strikingresults was the high sensitization of the lymphoma models toradiotherapy. Radiotherapy is often used for the treatment ofcancer, such as non-Hodgkin lymphoma, Hodgkin lymphoma,and all types of leukemia and the addition of AsiDNA couldincrease the efficacy of such treatment. We previously demon-strated in a first-in-man clinical trial that AsiDNA does notincrease local toxicity of the irradiation (19). Treating lympho-mas by combining treatment with AsiDNA and radiotherapy isa potentially promising method.

Overall, the addition of AsiDNA to radiotherapy and DNA-damaging chemotherapy increased cytotoxicity of these DNA-

damaging drugs in most malignant hematologic cell lines. Giventhe lack of toxicity of this combination in normal cells, thecombination of AsiDNA and DNA damage–inducing agentsshould be considered for the treatment of such pathologies, inparticular, at recurrence when resistance to treatments appears.

Disclosure of Potential Conflicts of InterestM. Dutreix is a consultant at and reports receiving a commercial research

grant fromOnxeo. No potential conflicts of interest were disclosed by the otherauthors.

Authors' ContributionsConception and design: S. Thierry, M. DutreixDevelopment of methodology: S. Thierry, W. JdeyAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): S. Thierry, W. Jdey, S. AlculumbreAnalysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis): S. Thierry, W. Jdey, V. Soumelis, M. DutreixWriting, review, and/or revision of the manuscript: S. Thierry, V. Soumelis,P. Noguiez-Hellin, M. DutreixAdministrative, technical, or material support (i.e., reporting or organizingdata, constructing databases): P. Noguiez-Hellin, M. DutreixStudy supervision: M. Dutreix

AcknowledgmentsWe thankNathalie Berthault andWendy Philippon for their technical help in

this project. This work benefitted from the help of David Gentien for genomicanalysis and Charlene Lasgi for flow cytometry. This work was supported by theSIRIC-Curie, the Institut Curie, theCentreNational de la Recherche Scientifique,the Institut National de la Sant�e Et de la Recherche M�edicale and the AgenceNationale de la Recherche. Funding for open access charge was from InstitutCurie.

W. Jdey was supported by fellowship CIFFRE-ANRT (2013/0907). S. Thierrywas supported by the Institut National du Cancer (TRANSLA13-081).

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 May 10, 2017; revised July 26, 2017; accepted September 13, 2017;published OnlineFirst September 25, 2017.

References1. Vardiman JW, Thiele J, ArberDA, BrunningRD, BorowitzMJ, Porwit A, et al.

The 2008 revision of the World Health Organization (WHO) classificationof myeloid neoplasms and acute leukemia: rationale and importantchanges. Blood 2009;114:937–51.

2. Vinayak S, Ford JM. PARP inhibitors for the treatment and prevention ofbreast cancer. Curr Breast Cancer Rep 2010;2:190–7.

3. Konecny GE, Kristeleit RS. PARP inhibitors for BRCA1/2-mutated andsporadic ovarian cancer: current practice and future directions. Br J Cancer2016;115:1157–73.

4. Davar D, Beumer JH, Hamieh L, Tawbi H. Role of PARP inhi-bitors in cancer biology and therapy. Curr Med Chem 2012;19:3907–21.

5. Jeggo PA, Pearl LH, Carr AM. DNA repair, genome stability and cancer: ahistorical perspective. Nat Rev Cancer 2016;16:35–42.

6. Lahtz C, PfeiferGP. Epigenetic changes ofDNA repair genes in cancer. JMolCell Biol 2011;3:51–8.

7. Puiggros A, Blanco G, Espinet B. Genetic abnormalities in chronic lym-phocytic leukemia: where we are and where we go. Biomed Res Int 2014;2014:435983.

8. HaaseD,GermingU, Schanz J, PfeilstockerM,Nosslinger T,Hildebrandt B,et al. New insights into the prognostic impact of the karyotype inMDS and

correlation with subtypes: evidence from a core dataset of 2124 patients.Blood 2007;110:4385–95.

9. McKinnon PJ, Caldecott KW. DNA strand break repair and human geneticdisease. Annu Rev Genomics Hum Genet 2007;8:37–55.

10. Weston VJ, Oldreive CE, Skowronska A, Oscier DG, Pratt G, Dyer MJ, et al.The PARP inhibitor olaparib induces significant killing of ATM-deficientlymphoid tumor cells in vitro and in vivo. Blood 2010;116:4578–87.

11. Horton TM, Jenkins G, Pati D, Zhang L, Dolan ME, Ribes-Zamora A, et al.Poly(ADP-ribose) polymerase inhibitor ABT-888 potentiates the cytotoxicactivity of temozolomide in leukemia cells: influence of mismatch repairstatus andO6-methylguanine-DNAmethyltransferase activity.Mol CancerTher 2009;8:2232–42.

12. Gojo I, Beumer JH, Pratz KW,McDevitt MA, Baer MR, Blackford AL, et al. Aphase 1 study of the PARP inhibitor veliparib in combination withtemozolomide in acute myeloid leukemia. Clin Cancer Res 2017;23:697–706.

13. Check Hayden E. Cancer complexity slows quest for cure. Nature 2008;455:148.

14. Quanz M, Chassoux D, Berthault N, Agrario C, Sun JS, Dutreix M.Hyperactivation of DNA-PK by double-strand break mimicking moleculesdisorganizes DNA damage response. PLoS One 2009;4:e6298.

Mol Cancer Ther; 16(12) December 2017 Molecular Cancer Therapeutics2826

Thierry et al.

on January 24, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst September 25, 2017; DOI: 10.1158/1535-7163.MCT-17-0405

Page 11: The DNA Repair Inhibitor Dbait Is Specific for Malignant ... · Hematologic malignancies account for approximately 10% of all newly diagnosed neoplasms. Their common characteristic

15. Quanz M, Berthault N, Roulin C, Roy M, Herbette A, Agrario C,et al. Small-molecule drugs mimicking DNA damage: a new strategyfor sensitizing tumors to radiotherapy. Clin Cancer Res 2009;15:1308–16.

16. Biau J, Devun F, JdeyW, Kotula E, QuanzM, Chautard E, et al. A preclinicalstudy combining the DNA repair inhibitor Dbait with radiotherapy for thetreatment of melanoma. Neoplasia 2014;16:835–44.

17. Croset A, Cordelieres F, Berthault N, Buhler C, Sun J-S, Quanz M, et al.Inhibition of DNA damage repair by artificial activation of PARP withsiDNA. Nucleic Acids Res 2013;41:7344–55.

18. Jdey W, Thierry S, Russo C, Devun F, Al Abo M, Noguiez-Hellin P, et al.Drug-driven synthetic lethality: bypassing tumor cell genetics with acombination of AsiDNA and PARP inhibitors. Clin Cancer Res 2017;23:1001–11.

19. Le Tourneau C, Dreno B, Kirova Y, Grob JJ, Jouary T, Dutriaux C, et al. First-in-human phase I study of the DNA-repair inhibitor DT01 in combinationwith radiotherapy in patients with skin metastases from melanoma. Br JCancer 2016;114:1199–205.

20. Kadowaki N, Antonenko S, Lau JY, Liu YJ. Natural interferon alpha/beta-producing cells link innate and adaptive immunity. J Exp Med 2000;192:219–26.

21. Alculumbre S, Pattarini L. Purification of humandendritic cell subsets fromperipheral blood. Methods Mol Biol 2016;1423:153–67.

22. Delelis O, Thierry S, Subra F, Simon F, Malet I, Alloui C, et al. Impact ofY143 HIV-1 integrase mutations on resistance to raltegravir in vitro and invivo. Antimicrob Agents Chemother 2010;54:491–501.

23. Frisch BJ, Calvi LM. Hematopoietic stem cell cultures and assays. MethodsMol Biol 2014;1130:315–24.

24. Sarma NJ, Takeda A, Yaseen NR. Colony forming cell (CFC) assay forhuman hematopoietic cells. J Vis Exp 2010; Dec 18(46). pii: 2195. doi:10.3791/2195.

25. Foucquier J, Guedj M. Analysis of drug combinations: current methodo-logical landscape. Pharmacol Res Perspect 2015;3:e00149.

26. Steel GG, Peckham MJ. Exploitable mechanisms in combined radiother-apy-chemotherapy: the concept of additivity. Int J Radiat Oncol Biol Phys1979;5:85–91.

27. ObeidM, Tesniere A,Ghiringhelli F, FimiaGM,Apetoh L, Perfettini JL, et al.Calreticulin exposure dictates the immunogenicity of cancer cell death.NatMed 2007;13:54–61.

28. BerthaultN,Maury B, Agrario C,Herbette A, Sun JS, PeyrierasN,DutreixM.Comparison of distribution and activity of nanoparticles with shortinterfering DNA (Dbait) in various living systems. Cancer Gene Ther2011;18:695–706.

29. Radwan AA, Alanazi FK. Targeting cancer using cholesterol conjugates.Saudi Pharm J 2014;22:3–16.

30. Peeters SD, van der KolkDM, deHaanG, Bystrykh L, Kuipers F, de Vries EG,et al. Selective expression of cholesterol metabolism genes in normalCD34þCD38- cells with a heterogeneous expression pattern in AML cells.Exp Hematol 2006;34:622–30.

31. De Sanctis JB, Blanca I, Radzioch D, Bianco NE. Expression and function oflow-density lipoprotein receptors in CD3-CD16þCD56þ cells: effect ofinterleukin 2. Cell Immunol 1996;167:18–29.

32. Devun F, BousquetG,Biau J,Herbette A, RoulinC, Berger F, et al. Preclinicalstudy of the DNA repair inhibitor Dbait in combination with chemother-apy in colorectal cancer. J Gastroenterol 2012;47:266–75.

33. Herath NI, Devun F, Lienafa MC, Herbette A, Denys A, Sun JS, et al. TheDNA repair inhibitor DT01 as a novel therapeutic strategy for chemosen-sitization of colorectal liver metastasis. Mol Cancer Ther 2016;15:15–22.

34. Finnie NJ, Gottlieb TM, Blunt T, Jeggo PA, Jackson SP. DNA-dependentprotein kinase activity is absent in xrs-6 cells: implications for site-specificrecombination and DNA double-strand break repair. Proc Natl Acad SciU S A 1995;92:320–24.

www.aacrjournals.org Mol Cancer Ther; 16(12) December 2017 2827

Toxicity and Efficacy of Dbait on Hematologic Cells

on January 24, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst September 25, 2017; DOI: 10.1158/1535-7163.MCT-17-0405

Page 12: The DNA Repair Inhibitor Dbait Is Specific for Malignant ... · Hematologic malignancies account for approximately 10% of all newly diagnosed neoplasms. Their common characteristic

2017;16:2817-2827. Published OnlineFirst September 25, 2017.Mol Cancer Ther   Sylvain Thierry, Wael Jdey, Solana Alculumbre, et al.   Hematologic Cells in BloodThe DNA Repair Inhibitor Dbait Is Specific for Malignant

  Updated version

  10.1158/1535-7163.MCT-17-0405doi:

Access the most recent version of this article at:

  Material

Supplementary

  http://mct.aacrjournals.org/content/suppl/2017/09/22/1535-7163.MCT-17-0405.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://mct.aacrjournals.org/content/16/12/2817.full#ref-list-1

This article cites 33 articles, 11 of which you can access for free at:

  Citing articles

  http://mct.aacrjournals.org/content/16/12/2817.full#related-urls

This article has been cited by 1 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 at

  Permissions

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

.http://mct.aacrjournals.org/content/16/12/2817To request permission to re-use all or part of this article, use this link

on January 24, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst September 25, 2017; DOI: 10.1158/1535-7163.MCT-17-0405