achieving precision death with cell-cycle inhibitors that ......inevitable for cell-cycle...

10
Review Achieving Precision Death with Cell-Cycle Inhibitors that Target DNA Replication and Repair Aimee Bence Lin 1 , Samuel C. McNeely 2 , and Richard P. Beckmann 3 Abstract All cancers are characterized by defects in the systems that ensure strict control of the cell cycle in normal tissues. The consequent excess tissue growth can be countered by drugs that halt cell division, and, indeed, the majority of chemotherapeu- tics developed during the last century work by disrupting processes essential for the cell cycle, particularly DNA synthesis, DNA replication, and chromatid segregation. In certain con- texts, the efcacy of these classes of drugs can be impressive, but because they indiscriminately block the cell cycle of all actively dividing cells, their side effects severely constrain the dose and duration with which they can be administered, allowing both normal and malignant cells to escape complete growth arrest. Recent progress in understanding how cancers lose control of the cell cycle, coupled with comprehensive genomic proling of human tumor biopsies, has shown that many cancers have mutations affecting various regulators and checkpoints that impinge on the core cell-cycle machinery. These defects intro- duce unique vulnerabilities that can be exploited by a next generation of drugs that promise improved therapeutic win- dows in patients whose tumors bear particular genomic aberra- tions, permitting increased dose intensity and efcacy. These developments, coupled with the success of new drugs targeting cell-cycle regulators, have led to a resurgence of interest in cell- cycle inhibitors. This review in particular focuses on the newer strategies that may facilitate better therapeutic targeting of drugs that inhibit the various components that safeguard the delity of the fundamental processes of DNA replication and repair. Clin Cancer Res; 23(13); 323240. Ó2017 AACR. Introduction Tissue growth and homeostasis in multicellular organisms necessitate a variety of systems to regulate and coordinate cell division. When these systems fail, unabated and uncoordinated growth can lead to cancer. The core cell-cycle machinery required for chromosome duplication, chromosome segrega- tion, and cytokinesis can be distinguished from cell-cycle reg- ulators that restrain the core engine via cell-cycle checkpoints. Many traditional cancer therapies act on the core machinery, either at S-phase, preventing nucleotide synthesis and DNA replication (antimetabolites, DNA-damaging agents, and radio- therapy) or at mitosis (microtubule inhibitors). As they act on processes essential for all dividing cells, these classical therapies also affect normal proliferating tissues, and toxicities seemed inevitable for cell-cycle inhibitors. Hence, in the 1990s, cancer drug discovery shifted focus from cell-cycle targets to mitogenic and oncogenic signaling pathways. Targeting somatic mutations in genes encoding members of these signaling pathways offered a better therapeutic window in the cancers where they were defective. This shift in focus was vindicated by success of oncogene-targeting drugs such as trastuzumab, imatinib, erlo- tinib, vemurafenib, and crizotinib (1). During this same period, next-generation sequencing ushered in efforts to comprehensively catalog all genomic aberrations that can drive cancer growth (2). Interestingly, these studies identied key cell-cycle checkpoint genes that are commonly mutated in cancer. In particular, checkpoints at three stages are targeted by mutations in established cancer genes: (i) G 1 phase checkpoints, including the restriction point; (ii) S-phase checkpoints, including DNA damage and origin ring checkpoints; and (iii) M-phase, particularly the spindle assembly checkpoint. Recent years have seen the introduction of inhibitors that specically target kinases that facilitate DNA replication and repair such as inhibitors of ataxia telangiectasia mutated kinase (ATM), ataxia telangiectasia and Rad3-related kinase (ATR), WEE1, checkpoint kinase 1 (CHK1), and checkpoint kinase 2 (CHK2), collectively referred to herein as checkpoint kinase inhibitors. Because their mechanisms of action inhibit crucial components of DNA damage checkpoints, the initial interest in this class of inhibitors was their use in combination with standard cytotoxic (genotoxic) therapies as chemopotentiators. However, preclinical investigations led to a next-generation strategy that relies on the identication of underlying genetic or expression alterations that can potentially enhance the susceptibly of cancer cells to checkpoint kinase inhibitor monotherapy or to combi- nation therapy with targeted drugs that do not induce generalized DNA damage. Consequently, this article reviews the underlying genetic drivers in tumors that facilitate better selective targeting of these agents, with a focus on checkpoint kinase inhibitors that are currently in development (Table 1). 1 Early Phase Medical-Oncology, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana. 2 Oncology Business Unit-Patient Tailoring, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana. 3 Oncology Translational Research, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana. Corresponding Author: Richard P. Beckmann, Eli Lilly and Company, Lilly Corporate Center, Drop Code 0438, Indianapolis, IN 46285. Phone: 317-276- 4285; Fax: 317-651-6346; E-mail: [email protected] doi: 10.1158/1078-0432.CCR-16-0083 Ó2017 American Association for Cancer Research. Clinical Cancer Research Clin Cancer Res; 23(13) July 1, 2017 3232 on January 14, 2021. © 2017 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from Published OnlineFirst March 22, 2017; DOI: 10.1158/1078-0432.CCR-16-0083

Upload: others

Post on 22-Sep-2020

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Achieving Precision Death with Cell-Cycle Inhibitors that ......inevitable for cell-cycle inhibitors. Hence, in the 1990s, cancer drug discovery shifted focus from cell-cycle targets

Review

Achieving Precision Death with Cell-CycleInhibitors that Target DNA Replication andRepairAimee Bence Lin1, Samuel C. McNeely2, and Richard P. Beckmann3

Abstract

All cancers are characterized by defects in the systems thatensure strict control of the cell cycle in normal tissues. Theconsequent excess tissue growth can be countered by drugs thathalt cell division, and, indeed, the majority of chemotherapeu-tics developed during the last century work by disruptingprocesses essential for the cell cycle, particularly DNA synthesis,DNA replication, and chromatid segregation. In certain con-texts, the efficacy of these classes of drugs can be impressive, butbecause they indiscriminately block the cell cycle of all activelydividing cells, their side effects severely constrain the dose andduration with which they can be administered, allowing bothnormal and malignant cells to escape complete growth arrest.Recent progress in understanding how cancers lose control ofthe cell cycle, coupled with comprehensive genomic profiling of

human tumor biopsies, has shown that many cancers havemutations affecting various regulators and checkpoints thatimpinge on the core cell-cycle machinery. These defects intro-duce unique vulnerabilities that can be exploited by a nextgeneration of drugs that promise improved therapeutic win-dows in patients whose tumors bear particular genomic aberra-tions, permitting increased dose intensity and efficacy. Thesedevelopments, coupled with the success of new drugs targetingcell-cycle regulators, have led to a resurgence of interest in cell-cycle inhibitors. This review in particular focuses on the newerstrategies that may facilitate better therapeutic targeting ofdrugs that inhibit the various components that safeguard thefidelity of the fundamental processes of DNA replication andrepair. Clin Cancer Res; 23(13); 3232–40. �2017 AACR.

IntroductionTissue growth and homeostasis in multicellular organisms

necessitate a variety of systems to regulate and coordinate celldivision. When these systems fail, unabated and uncoordinatedgrowth can lead to cancer. The core cell-cycle machineryrequired for chromosome duplication, chromosome segrega-tion, and cytokinesis can be distinguished from cell-cycle reg-ulators that restrain the core engine via cell-cycle checkpoints.Many traditional cancer therapies act on the core machinery,either at S-phase, preventing nucleotide synthesis and DNAreplication (antimetabolites, DNA-damaging agents, and radio-therapy) or at mitosis (microtubule inhibitors). As they act onprocesses essential for all dividing cells, these classical therapiesalso affect normal proliferating tissues, and toxicities seemedinevitable for cell-cycle inhibitors. Hence, in the 1990s, cancerdrug discovery shifted focus from cell-cycle targets to mitogenicand oncogenic signaling pathways. Targeting somatic mutationsin genes encoding members of these signaling pathways offereda better therapeutic window in the cancers where they were

defective. This shift in focus was vindicated by success ofoncogene-targeting drugs such as trastuzumab, imatinib, erlo-tinib, vemurafenib, and crizotinib (1).

During this same period, next-generation sequencing usheredin efforts to comprehensively catalog all genomic aberrations thatcan drive cancer growth (2). Interestingly, these studies identifiedkey cell-cycle checkpoint genes that are commonly mutated incancer. In particular, checkpoints at three stages are targeted bymutations in established cancer genes: (i) G1 phase checkpoints,including the restrictionpoint; (ii) S-phase checkpoints, includingDNA damage and origin firing checkpoints; and (iii) M-phase,particularly the spindle assembly checkpoint.

Recent years have seen the introduction of inhibitors thatspecifically target kinases that facilitate DNA replication andrepair such as inhibitors of ataxia telangiectasia mutated kinase(ATM), ataxia telangiectasia and Rad3-related kinase (ATR),WEE1, checkpoint kinase 1 (CHK1), and checkpoint kinase 2(CHK2), collectively referred to herein as checkpoint kinaseinhibitors. Because their mechanisms of action inhibit crucialcomponents of DNA damage checkpoints, the initial interest inthis class of inhibitors was their use in combinationwith standardcytotoxic (genotoxic) therapies as chemopotentiators. However,preclinical investigations led to a next-generation strategy thatrelies on the identification of underlying genetic or expressionalterations that can potentially enhance the susceptibly of cancercells to checkpoint kinase inhibitor monotherapy or to combi-nation therapy with targeted drugs that do not induce generalizedDNA damage. Consequently, this article reviews the underlyinggenetic drivers in tumors that facilitate better selective targeting ofthese agents, with a focus on checkpoint kinase inhibitors that arecurrently in development (Table 1).

1Early Phase Medical-Oncology, Eli Lilly and Company, Lilly Corporate Center,Indianapolis, Indiana. 2Oncology Business Unit-Patient Tailoring, Eli Lilly andCompany, Lilly Corporate Center, Indianapolis, Indiana. 3Oncology TranslationalResearch, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana.

Corresponding Author: Richard P. Beckmann, Eli Lilly and Company, LillyCorporate Center, Drop Code 0438, Indianapolis, IN 46285. Phone: 317-276-4285; Fax: 317-651-6346; E-mail: [email protected]

doi: 10.1158/1078-0432.CCR-16-0083

�2017 American Association for Cancer Research.

ClinicalCancerResearch

Clin Cancer Res; 23(13) July 1, 20173232

on January 14, 2021. © 2017 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst March 22, 2017; DOI: 10.1158/1078-0432.CCR-16-0083

Page 2: Achieving Precision Death with Cell-Cycle Inhibitors that ......inevitable for cell-cycle inhibitors. Hence, in the 1990s, cancer drug discovery shifted focus from cell-cycle targets

ATM, ATR, WEE1, CHK1, and CHK2 are crucial components ofDNA damage response (DDR) signaling networks responsible forcontributing either to the detection and repair of damage or forcoordinating DNA replication (3–6). These DDR checkpointkinases function collectively to maintain genomic integrity byproviding cells time to repair any DNA damage prior to replica-tionormitosis, and to initiate an apoptotic response if thedamageis beyond repair. ATR and ATM act as sensors of single-strand anddouble-strand breaks (SSB and DSB), respectively, which activateCHK1 and CHK2 by phosphorylation (Fig. 1). CHK1 and CHK2suppress cell-cycle progression through downregulation of theCDC25A and CDC25C phosphatases, which promote progres-sion by removing inhibitory phosphates on CDK2 and CDK1,

respectively (7). WEE1 also prevents cell-cycle progression viainhibitory phosphorylation of CDK1 and CDK2 (8–11). TheATR–CHK1 response is also initiated in response to ssDNAgenerated by replication fork stalling (12, 13).

Strategies for Clinical Development: Pastand Present

The initial preclinical studies evaluating these inhibitors clearlydemonstrated that the chemical inhibition of checkpoint kinasesenhances the activity of cytotoxic therapies that damage DNAthrough diverse mechanisms. Good reviews have been publishedrecently describing this biology, and, therefore, it will not be

Table 1. Cell-cycle kinase inhibitors currently in development

Target Agent Phase Context Populations being evaluated in all clinical contexts

WEE1 AZD1775, MK1775 II MonotherapyChemotherapy combinations: cisplatin, carboplatin,docetaxel, gemcitabine, irinotecan, nab-paclitaxel,paclitaxel, peglyated liposomal doxorubicin,pemetrexedTargeted therapy combinations: olaparib, belinostatRadiation/chemoradiation combinations:monotherapy, cisplatin, gemcitabine, temozolomideImmunotherapy combinations: durvalumab

Cervical, CRC, gastric/GEJ, glioblastoma, gliomas, HNSCC,NSCLC, myeloid malignancies, ovarian, pancreatic,pediatric tumors, SCLC, squamous NSCLC, TNBCBiomarker focused:Basket trial: molecular profiling–based assignmentBladder: mutations in genes associated with cell-cycleregulation (RB1, CDKN2A, CCNE1 or MYC amp)CRC: KRAS/NRAS/BRAF mutationsGastric/GEJ: TP53 mutationsNSCLC: biomarker-directed basket trialOvarian: BRCA1 mutations, BRCA2 mutations, TP53 muta-tions, window trialPediatric: basket trial stratifying based on molecularanomalies

CHK1 Prexasertib,LY2606368

II MonotherapyChemotherapy combinations: cisplatin, 5-FU,pemetrexedTargeted therapy combinations: cetuximab,LY3023414 (PI3K/mTOR inhibitor), ralimetinib(p38 MAPK inhibitor), olaparibChemoradiation combinations: cetuximab, cisplatin

AML/high-risk MDS, CRC, HSNCC, NSCLC, ovarian,pediatric, prostate, SCC of anus, squamous NSCLC,SCLC, TNBCBiomarker focused:Basket trial: replication stress or homologous repairdeficiency (e.g., MYC or CCNE1 amp, Rb loss, FBXW7mutation, BRCA1, BRCA2, PALB2, RAD51C, RAD51D,ATR, ATM, CHK2, the Fanconi anemia pathway genes)Breast: BRCA1 mutations, BRCA2 mutationsCRC: KRAS/BRAF mutationsNSCLC: KRAS/BRAF mutationsOvarian: BRCA1 mutations, BRCA2 mutations

LY2880070, ESP-01 I MonotherapyChemotherapy combinations: gemcitabine

CRC, ovarian, TNBC

CCT245737, PNT-737 I MonotherapyChemotherapy combinations: gemcitabine, cisplatin

NSCLC, pancreatic

GDC-0575, ARRY-575 I MonotherapyChemotherapy combinations: gemcitabine

ATR VX970 II MonotherapyChemotherapy combinations: cisplatin, carboplatin,gemcitabine, etoposide, irinotecan, topotecanTargeted combinations: veliparibChemoradiation combinations: monotherapy, cisplatin

HNSCC (HPV negative), NSCLC, ovarian, SCLC, urothelial,squamous NSCLC, TNBC

AZD6738 I MonotherapyChemotherapy combinations: carboplatin, paclitaxelTargeted combinations: olaparibRadiationImmunotherapy combinations: durvalumab

B-cell malignancies, gastric/GEJ, HNSCC, NSCLCBiomarker focused:CLL: loss of chromosome 11q or ATM deficientGastric/GEJ: low ATM expressionHNSCC: window trialNSCLC: low ATM expression

VX803 I MonotherapyChemotherapy combinations: gemcitabine, carboplatin,cisplatin

Ovarian

ATM AZD0156 I MonotherapyChemotherapy combinations: irinotecan, olaparib

NOTE: Source: www.clinicaltrials.gov.Abbreviations: AML, acute myeloid leukemia; amp, amplification; CLL, chronic lymphocytic leukemia; CRC, colorectal cancer; GEJ, gastroesophageal junction; HPV,humanpapillomavirus; HNSCC, squamous cell head and neck cancer; MDS,myelodysplastic syndrome;NSCLC, non–small cell lung cancer; SCC, squamous cell cancer;SCLC, small-cell lung cancer; TNBC, triple-negative breast cancer.

Selective Targeting of Checkpoint Kinase Inhibitors

www.aacrjournals.org Clin Cancer Res; 23(13) July 1, 2017 3233

on January 14, 2021. © 2017 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst March 22, 2017; DOI: 10.1158/1078-0432.CCR-16-0083

Page 3: Achieving Precision Death with Cell-Cycle Inhibitors that ......inevitable for cell-cycle inhibitors. Hence, in the 1990s, cancer drug discovery shifted focus from cell-cycle targets

summarized here (7, 10, 12–16). As a result of these data, clinicaltrials with early CHK1 inhibitors focused on the chemopotentia-tion of cytotoxic drugs. Although phase I trials demonstratedproof of concept that CHK1 inhibitors could be safely combinedwith chemotherapy (17–25), phase II studies failed to meet theirprimary endpoints (26, 27). Early CHK1 inhibitors were notsuccessful for a variety of reasons, including pharmacokineticproperties, unacceptable toxicities, and business considerations.

However, newer inhibitors, which aremore diverse and includenot only CHK1 inhibitors but also WEE1, ATR, and ATM inhibi-tors, continue to test this hypothesis with a variety of genotoxicagents and as monotherapies (Table 1). Although no phase IIstudies have been published with checkpoint inhibitors asmono-therapy, evidence of efficacy has been observed in phase I. A phaseI evaluation of AZD1775 as a monotherapy enrolled 24 patients,nine ofwhomhadBRCA1orBRCA2mutations. Twopatientswitha BRCA1mutation [squamous cell cancer (SCC) of the base of the

tongue and papillary serous ovarian cancer] had partial responses(28). Prexasertib, a CHK1 inhibitor, demonstrated objectiveresponses in patients with SCC of the anus and head and neckcancer in a phase I study with multiple expansion cohorts (29).These preliminary results suggest that the newer inhibitorsmay bemore successful than initial CHK1 inhibitors, which were onlydeveloped as chemopotentiators.

In addition, these data suggest the optimal use of these inhi-bitors may require the identification of contexts where tumor-specific vulnerabilities are leveraged to achieve selective cytotox-icity in cancer cells. p53 is a critical mediator of the DDR, which isactivated through phosphorylation by CHK2 and ATM, andparticipates in aparallel pathway toATR,CHK1, andWEE1 (Fig. 1;ref. 7). For this reason, cancer cells that have lost either p53function or ATM may show greater reliance on ATR, CHK1, andWEE1 for efficient repair and, hence, greater sensitivity to treat-ments that inhibit these kinases (30–35). This hypothesis has

© 2017 American Association for Cancer Research

P P

P P

PP

P

P

P

P P

Endogenous or exogenous events

SSBDSB

ATM ATR

CHK2 CHK1

(Proteolysis) (Sequestration)

CDK2

G1 G2S M

Cyclin A/BCyclin A/BCyclin E Cyclin E

p53 p21

CDK2 CDK1 CDK1

CDC25A CDC25C

WEE1 WEE1

Figure 1.

ATR, ATM, CHK1, CHK2, and WEE1 inhibit cell-cycle progression into S-phase and mitosis following DNA damage. This regulation occurs ultimately through thecontrol of the cyclin-dependent kinases (CDK) that facilitate entry into the S- and M-phases of the cell cycle, namely CDK2 and CDK1, respectively. Theactivity of CDK1 and CDK2 are controlled by phosphorylation as well as by the partnership of these kinases with their regulatory cyclin subunits,whereby activation of CDK2 requires association with cyclin E and CDK1 activation requires association with either cyclin A or B. Phosphorylation of eitherCDK1 or CDK2 is regulated in part by the opposing actions of the WEE1 kinase and the CDC25A/CDC25C phosphatases. ATR and ATM are sensors ofSSBs and DSBs, respectively, which activate CHK1 and CHK2, respectively, by phosphorylation. The inhibition of cell-cycle progression by activated(phosphorylated) CHK1 and CHK2 results from the inhibition of CDK1 and CDK2, which is mediated by preventing the removal of inhibitory phosphates placedon CDK1 and CDK2, by WEE1. CHK1 and CHK2 prevent the removal of these phosphates by suppressing the CDC25A and CDC25C phosphatasesthrough phosphorylation, whereby this phosphorylation facilitates proteolytic degradation of CDC25A and the sequestration of CDC25C by 14-3-3. p53 isactivated upon DNA damage by phosphorylation by ATM and CHK2 and serves as an additional pathway to inhibit cell-cycle progression bypromoting transcription of p21, which in turn inhibits the kinase activity of the cyclinE–CDK2 complex.

Lin et al.

Clin Cancer Res; 23(13) July 1, 2017 Clinical Cancer Research3234

on January 14, 2021. © 2017 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst March 22, 2017; DOI: 10.1158/1078-0432.CCR-16-0083

Page 4: Achieving Precision Death with Cell-Cycle Inhibitors that ......inevitable for cell-cycle inhibitors. Hence, in the 1990s, cancer drug discovery shifted focus from cell-cycle targets

been tested in a phase II trial where patients with TP53-mutatedovarian cancer who were refractory or resistant (<3 months) tofirst-line therapy received AZD1775 in combination with carbo-platin. The combination demonstrated manageable toxicity, andin 21 evaluable patients, the objective response rate was 43%.Median progression-free and overall survival times were 5.3months and 12.6 months, respectively. In addition to TP53mutations, patients with an objective response had alterationsin BRCA1, KRAS, MYC, or CCNE (cyclin E; ref. 36). Similarly, in arandomized, phase II trial in platinum-sensitive, TP53-mutantovarian cancer, the WEE1 inhibitor AZD1775 combined withpaclitaxel and carboplatin met the primary and secondary end-points and significantly prolonged progression-free survival com-pared with the combination of paclitaxel and carboplatin (37). Ina phase I, multi-arm combination study assessing AZD1775 witheither gemcitabine, cisplatin, or carboplatin, 176 patients wereevaluable for response. Responses were observed in patients withovarian cancer (n ¼ 7), melanoma (n¼ 3), breast cancer (n¼ 2),head and neck cancer (n¼ 3), colorectal cancer (n¼ 1), and SCCof the skin (n¼ 1). Patientswith TP53mutations (4/19, 23%)hada partial response compared with 4/33 (12%) of patients withTP53 wild-type tumors (38).

Ongoing studies with AZD1775 continue to evaluate thishypothesis in TP53-mutated gastric cancer (with paclitaxel,NCT02448329), TP53- or KRAS-mutated solid tumors (witholaparib, NCT02576444), and TP53-mutated (with either MYCamplification or CDKN2A mutation) relapsed small-cell lungcancer (SCLC; monotherapy, NCT02688907). Similarly, theATR inhibitor AZD6738 has an ongoing clinical study(NCT02264678) in ATM-low/deficient NSCLC (with carbopla-tin) or gastric cancer (with olaparib). These studies will furthercharacterize the impact of TP53 mutations and ATM deficiencynot only in the context of cytotoxic chemotherapy but also withtargeted agents and monotherapy.

The emerging clinical data with TP53mutations are one exam-ple of potential synthetic lethality (SL), where functional altera-tions in one pathway lead to enhanced sensitivity to inhibition ofanother pathway. This is the strategy guiding the use of PARPinhibitors as therapies for BRCA1- or BRCA2-deficient breast orovarian cancers (39–42). Leveraging SL for checkpoint kinaseinhibitors requires knowledge about functional alterations incancer cells that make them more vulnerable to the inhibitionof these kinases than normal cells. Although TP53 mutations arethe most well-characterized example of SL with the checkpointinhibitors, the concept of replication stress (RS) is emerging as apotential SL mechanism (43, 44). RS can arise in any situationthat leads to inappropriate replication origin licensing or firing(45). This results in stalled replication forks and DNA breaks ifthe stalled forks are not adequately resolved (46–48). In cancercells, oncogene-driven events can contribute to higher degrees ofRS by promoting growth to a point that strains the replicativecapacity of the cell (45, 49). As a result, inhibitors of checkpointkinases may induce SL with tumors that have high RS, as all ofthese kinases contribute to sensing and reducing RS (Fig. 2). Inparticular, WEE1 and CHK1 play complementary roles in restrict-ing the initiation of replication origins by inhibiting CDK2,whichwhen activated, promotes replication (6, 50, 51). Thus, treatmentwith an inhibitor of CHK1 or WEE1 augments ongoing RS byeffectively neutralizing one of the mechanisms available forsuppressing replication origin firing, resulting in more stallingand DNA breaks. As both CHK1 and WEE1 are important

signaling components in the response to DNA damage, damagethat results from the additional RS is potentially exacerbatedbecause it cannot be repaired effectively. Likewise, ATR plays animportant role as a sensor for RS as it is recruited to regions ofssDNA that become exposed by replication fork stalling. Subse-quently, ATR is responsible for activating CHK1, which in turnsuppresses replication, as indicated above (15). An ongoingchallenge is to identify tumors that have reached near-criticallevels of RS and are likely the most susceptible to treatment withcheckpoint kinase inhibitors.

Validation of therapeutic strategies to exploit RS has recentlyemerged from several studies (52–63). For example, an SL rela-tionship was described between oncogene-induced RS by MYCactivation and ATR loss in lymphomas (49). Specifically in thecontext of ATR-deficient cells, MYC expression induced higher RSand apoptosis and contributed to greater sensitivity to ATR andCHK1 inhibitors. Analogous observations were derived fromstudies in neuroblastoma models wherein sensitivity to CHK1inhibition was correlated with MYCN expression (52). Studies inother model systems for hematologic malignancies such as MYC-driven diffuse large B-cell lymphoma, cyclin D1–driven mantlecell lymphomas, T-cell acute lymphoblastic leukemia, and acutemyeloid leukemia suggest that oncogene-induced RS in thesecancers maymake them particularly vulnerable to treatment withcheckpoint kinase inhibitors (53–55, 58, 61, 63). The hypothesisthat MYC may drive RS and increase sensitivity to checkpointkinase inhibitors is being tested in several ongoing clinical trials,including a biomarker-directed study with AZD1775 and durva-lumab in muscle-invasive bladder cancer. To be eligible, patientsmust have mutations in CDKN2A or RB1 genes and/or amplifi-cation of CCNE1, MYC, MYCL, or MYCN genes, all of whichpresumably contribute to heightened RS (Fig. 2). Other agentsare focusing on SCLC, a tumor associated with RS and MYCamplifications: AZD1775 (NCT02593019) and prexasertib(NCT02735980) are being assessed as monotherapy, whereasVX970 is being evaluated with topotecan (NCT02487095).

Another opportunity for SL may be inhibition of ATR in thecontext of ATM deficiency. In preclinical models of hematologicmalignancies, cells that lack ATM (e.g., due to 11q deletions) orhave defects in p53 signaling were particularly vulnerable to theATR inhibitor AZD6738. Treatment with AZD6738 increasedreplication initiation and fork stalling, resulting in RS and DNAdamage that likely contributed to selective cytotoxicity in cellswith p53 and/or ATM defects (64). In parallel, in a phase I trialwith the ATR inhibitor VX970 and carboplatin, a patient withcolorectal cancer and ATM loss achieved a complete response(65). This hypothesis is being further tested in a trial of AZD6738in patients with relapsed/refractory B-cell malignancies withprospectively identified 11q-deleted or ATM-deficient relapsed/refractory CLL (NCT01955668).

In addition to neuroblastomas, as mentioned above, otherpediatric cancers, particularly sarcomas, may represent anotheropportunity for exploiting oncogenic drivers of RS, as demon-strated by recent studies in models for Ewing sarcoma (ES)wherein the oncogenic drivers were fusion proteins unique tothis tumor (EWS/FLI1 or EWS/ERG; ref. 59). In these studies, EScell lines were more sensitive to inhibition of ATR compared withsarcoma cell lines that lacked the EWS fusions, and ATR inhibitormonotherapy resulted in near-complete to complete growthinhibition of humanES xenografts. Related studieswith theCHK1inhibitor prexasertib (56) showed that monotherapy resulted in

Selective Targeting of Checkpoint Kinase Inhibitors

www.aacrjournals.org Clin Cancer Res; 23(13) July 1, 2017 3235

on January 14, 2021. © 2017 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst March 22, 2017; DOI: 10.1158/1078-0432.CCR-16-0083

Page 5: Achieving Precision Death with Cell-Cycle Inhibitors that ......inevitable for cell-cycle inhibitors. Hence, in the 1990s, cancer drug discovery shifted focus from cell-cycle targets

complete regressions in xenograft models for desmoplastic smallround cell tumor, which also expresses an oncogenic EWS fusionprotein known as EWS–WT1, or alveolar rhabdomyosarcoma, amalignancy driven by another unique oncogenic fusion protein(66, 67). These nonclinical studies with prexasertib have led to aninterest in exploring prexasertib in pediatric solid tumors(NCT02808650). In addition, WEE1 was identified as a target inmedulloblastoma, the most common pediatric malignant braintumor (68). AZD1775 induced DNA damage and suppressed thegrowth of medulloblastoma cells both in vitro and in vivo (68),potentially by exploiting MYC-induced RS (69). AZD1775 hasalso demonstrated in vitro activity in rhabdomyosarcoma asmonotherapy and in combination with conventional therapies(70) and improved the efficacy of radiation in mouse models of

pediatric high-grade glioma (71). Aphase I study of AZD1775 andradiation in children with diffuse intrinsic pontine gliomas isongoing (NCT01922076). AZD1775 is also being assessed in theEuropean Proof-of-Concept Therapeutic Stratification Trial ofMolecular Anomalies in Relapsed or Refractory Tumors(ESMART). In this basket study, molecular profiling of pediatrictumors is used to direct patients to individual treatmentarms, including a combination of AZD1775 and carboplatin(NCT02813135).

Strategies exploiting RS also include combining inhibitorstargeting different checkpoint kinases. Accordingly, RNA inter-ference (RNAi) studies exploring SL with the CHK1 inhibitorPF-00477736 identified WEE1 as the most significant hit (72).In addition, synergistic growth inhibition was observed with

© 2017 American Association for Cancer Research

ATR

PCHK1

Cyclin E

CDK2

Cyclin E

CDK2

P

P

PCDC25A

RPA

RPA

RPA

RPA

ATRIPDNA lesion

• Oncogenes• Enhanced cyclin E or CDK2 expression/activity• Loss of regulation of G1 → S progression• Loss of function of ATR, CHK1, or WEE1

“Activation”

Homologousrecombination repair

Fork collapse → DSBs

Replication stress

TopBP1

WEE1

Figure 2.

The ATR–CHK1 pathway reduces RS. The kinase function of CDK2 requires activation by association with cyclin E. When activated by cyclin E, CDK2 facilitatesthe progression of cells from G1 into S-phase and subsequently promotes replication origin firing. ATR and CHK1 suppress replication origin firing in part bysuppressing the activity of CDK2 through regulation of downstream effectors that coordinate the phosphorylation of CDK2. Specifically, the presence of longstretches of single-stranded DNA, which can occur in situations where the DNA polymerases lag behind the unwinding activity of the helicases, trigger ATR, which isrecruited by a complex of proteins including replication protein A (RPA), topoisomerase-binding protein 1 (TopBP1), and ATR-interacting protein (ATRIP).ATR then activates CHK1 (through phosphorylation), which subsequently through phosphorylation of the CDC25A phosphatase, negatively regulates CDK2 bypreventing the removal of inhibitory phosphates by this phosphatase. The actions of CDC25A are also opposed by WEE1, which contributes to the inhibitionof CDK2 by catalyzing the inhibitory phosphates that are removed by CDC25A. As shown at the bottom half of the figure, RS can result from endogenous orexogenous DNA damage as well as through the action of growth-promoting oncogenes, such as MYC, which serve to drive progression into S-phase. Inaddition, RS can also arise through other alterations that disrupt control at the G1–S interface, such as changes that lead to enhanced expression of cyclinE or loss of inhibition of CDK2. Alterations that may enhance cyclin E expression or CDK2 activity possibly include (i) losses in FBXW7, which facilitatesproteasomal degradation of cyclin E or (ii) alterations that disrupt regulation of the primary restriction point controlling progression from G1 into S includingactivation of CDK4, CDK6, and D-type cyclins or loss of RB1 that serves to activate E2F, thereby promoting the transcription of the genes that encode cyclin Eand CDK2 as well as other gene products that promote DNA replication. Notably, as indicated by the red Xs, the inhibition or loss of function of ATR, CHK1,or WEE1 can elevate ongoing RS by effectively removing the control over CDK2. The result of this inhibition or loss is replication fork collapse, with eventualDNA DSBs leading to the activation of DNA damage and repair responses such as homologous recombination repair.

Lin et al.

Clin Cancer Res; 23(13) July 1, 2017 Clinical Cancer Research3236

on January 14, 2021. © 2017 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst March 22, 2017; DOI: 10.1158/1078-0432.CCR-16-0083

Page 6: Achieving Precision Death with Cell-Cycle Inhibitors that ......inevitable for cell-cycle inhibitors. Hence, in the 1990s, cancer drug discovery shifted focus from cell-cycle targets

PF-00477736 and AZD1775 in cell lines and in a xenograftmodel for ovarian cancer (72). Synergy was also observed withAZD1775 and the CHK1 inhibitor MK-8776 (73). Analogousstudies showed that treatment of cancer cells in culture with theCHK1 inhibitor AZD7762 and the ATR inhibitor VE-821 ele-vated RS, leading to replication catastrophe and death byapoptosis. The combination was associated with a significantinhibition of tumor growth and an increase in overall survivalin mice bearing human NCI-H460 NSCLC xenografts whencompared with the monotherapy treatments (74).

In addition to RS, deficiencies in homologous recombinationrepair (HRR) may also contribute to greater sensitivity to check-point kinase inhibition, as homologous recombination deficien-cies (HRD) reduce the efficiency of repair of DSBs that result fromRS generated by these inhibitors (Fig. 2). Deficiencies in theFanconi anemia (FA) genes (FANC), such as defects in BRCA1or BRCA2, also disrupt HRR (75), and studies with the CHK1inhibitor G€o6976 showed that FA-deficient cell lines were highlysensitive to G€o6976 and to RNAi silencing of CHK1 mRNAexpression when compared with isogenic lines that were FAproficient (76). An RNAi-based screen that targeted 230 DNArepair genes identified many FANC genes as SL with G€o6976treatment, including FANCA, FANCC, FANCD1, FANCD2,FANCE, FANCF, and FANCG.

A similar approach using siRNA to target 240 DNA replicationand repair genes explored SL with ATR inhibitors (57). Thesestudies uncovered that loss of expression of ATR pathway geneshad the strongest association for SL including losses of ATR itself,ATRIP, RPA, and CHK1. As the ATR pathway is crucial forresponding to RS, the observations from this study as well asothers provide support regarding the therapeutic benefits thatmay be derived from the combined inhibition of targets withinthis pathway to elevate RS further (62, 72–74). In addition, theresults confirm the benefit that may be attained through mono-therapy by selective targeting of tumors with preexisting muta-tions in this pathway. Of note, these studies demonstrated thatloss of excision repair cross-complementation group 1 protein(ERCC1), which functions in the repair of bulky DNA adducts,DSBs, and interstrand cross-links (77), was a significant SL inter-action with ATR and CHK1 inhibition. Similar SL interactionswere observed between ATR inhibition and loss of another repairgene known as XRCC1, which plays a role base excision repair(78) and with ATM deficiency (34).

These studies suggest that therapeutic strategies aimed at dis-rupting HRR may facilitate SL with checkpoint kinase inhibitors.In this regard, CHK1, in addition to its role in limiting RS, isrequired for HRR by facilitating the essential recruitment ofRAD51 to sites of repair (79). Thus, CHK1 inhibition would beexpected to promote further deficiencies in HRR in addition toexacerbating RS. In support of this concept, augmented growthinhibition by the PARP inhibitor olaparib was observed upon theRNAi silencing of genes encoding CHK1, CHK2, ATR, ATM, andRPA1 as well as genes encoding proteins that contribute to HRRsuch as RAD51, NBS1, FANCA, FANCD2, and FANCC (40). Inaddition, studies in breast carcinoma cell lines showed that thecombination of PARP inhibitors (olaparib, veliparib, NU1025, orrucaparib) with CHK1 inhibitors (UCN-01, AZD7762, orLY2603618) increased DNA damage and cytotoxicity, as com-pared with the single-agent treatments (80). More recent studieshave shown in BRCA1- or BRCA2-mutated high-grade serousovarian cancer (HGSOC) models that combination therapy with

olaparib and either MK-8776 (CHK1) or AZD6738 (ATR) actedsynergistically to decrease survival and colony formation in vitroand inhibit tumor xenograft growth in vivo (81). Clinical trialsevaluating PARP inhibition with AZD1775 (NCT02511795),AZD0156 (NCT02588105), VX970 (NCT02723864), orAZD6738 (NCT02264678) are ongoing. In addition, a studywithAZD1775 and olaparib (OLAPCO) is a molecularly directed trialthat requires mutations in TP53 or KRAS (NCT02576444).

As outlined above, the optimal context for a checkpoint kinaseinhibitor might be tumors that have both high RS and HRD. Ofnotable interest are ovarian cancers, particularly HGSOCs, asapproximately half harbor mutations in genes that modulateHRR, including mutations that impact BRCA1 and BRCA2(39, 82). In addition, nearly all HGSOCs have mutations in TP53(83). Mutations in ATM and ATR have been observed in 2% ofHGSOCs, and 5% have mutations in genes of the FA DNA repairpathway (39). Mutually exclusive with mutations in BRCA1 andBRCA2,CCNE1 is amplified in 15% to 20% of HGSOCs (82, 84).RB1 loss is also observed in about 15% of these cancers (83). Ascyclin E is required for activation of CDK2, its overexpressioninduces RS andDNAdamage that activates HRR andmay increasesensitivity to single-agent CHK1, ATR1, and/or WEE1 inhibition(85). Likewise loss of RB1 can contribute to RS by promoting theprogression of cells into S-phase. Therefore, HGSOCs present aprovocative context for therapy with the checkpoint kinase inhi-bitors by providing an opportunity to leverage the dual presenceof higher RS and HRD (28, 86). Accordingly, AZD1775, prexa-sertib, LY2880070, VX970, VX803, and AZD0156 are beingassessed in a variety of subsets of ovarian cancer (Table 1), andit may be notable that AZD1775 has demonstrated clinicalactivity in both platinum-sensitive and platinum-resistant ovar-ian cancer (36, 37).

As our understanding of SL interactions and the underlyingmechanisms of the checkpoint kinase inhibitors grows, theoptimal context may shift from a focus on a particular histologysuch as ovarian cancer, to the genetic attributes of the tumorregardless of histology. This approach is already being imple-mented in multiple clinical trials. In a prexasertib basket trial(NCT02873975), patients whose tumors show alterations con-sistent with RS (MYC amplification, CCNE1 amplification, Rbloss, or FBXW7 mutation) or HRD (BRCA1, BRCA2, PALB2,RAD51C, RAD51D, ATR, ATM, CHK2, or the FA pathway genes)are eligible. Similarly, AZD1775 is being assessed in the Molec-ular Profiling–Based Targeted Therapy (MPACT) trial, whichassigns patients with solid tumors to a treatment regimenbased on their mutation/amplification category. One of thesearms assesses AZD1775 in combination with carboplatin(NCT01827384). One obvious goal from these types of studies,besides improved efficacy, is to identify tumor-specific biomar-kers that can be used to predict response to monotherapy orcombination therapy with one or more checkpoint kinaseinhibitors. Preclinical studies indicate that alterations inducingRS or HRD contribute to greater sensitivity to these inhibitors.However, the crucial challenge from a clinical perspective isgoing beyond just identifying which of these alterations indi-cate the presence of RS or HRD to identifying situations wheretumors are at near-unstainable levels of RS or HRD and,therefore, are most susceptible to interventions that furtheraugment RS or compromise HRR.

The identification of expression or genetic alterations in a smallsubset of stand-alone biomarkers for predicting response is

Selective Targeting of Checkpoint Kinase Inhibitors

www.aacrjournals.org Clin Cancer Res; 23(13) July 1, 2017 3237

on January 14, 2021. © 2017 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst March 22, 2017; DOI: 10.1158/1078-0432.CCR-16-0083

Page 7: Achieving Precision Death with Cell-Cycle Inhibitors that ......inevitable for cell-cycle inhibitors. Hence, in the 1990s, cancer drug discovery shifted focus from cell-cycle targets

certainly desirable and may be achievable in certain cancers, butfor other cancers, additional methodologies may be required toadequately measure the extent to which RS and HRD are nearcatastrophic. However, these challenges should not dampen ourenthusiasm for the ongoing clinical trials with the various check-point kinase inhibitors, as someof these trialsmayprovidepatientdata that will allow us to validate and refine our biomarkerhypotheses. The preclinical studies have provided a solid foun-dation leading to the concept of leveraging RS andHRD to achieveSL in human tumors. The clinical datawill allowus to return to thebench to explore additional concepts that will further enhancethe therapeutic benefit of the checkpoint kinase inhibitors.

Disclosure of Potential Conflicts of InterestA.B. Lin, S.C. McNeely, and R.P. Beckmann hold ownership interest in

Eli Lilly and Company. No other potential conflicts of interest weredisclosed.

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

Received October 17, 2016; revised November 29, 2016; accepted March 15,2017; published OnlineFirst March 22, 2017.

References1. Chin L, Andersen JN, Futreal PA. Cancer genomics: from discovery science

to personalized medicine. Nat Med 2011;17:297–303.2. Stratton MR, Campbell PJ, Futreal PA. The cancer genome. Nature

2009;458:719–24.3. Ciccia A, Elledge SJ. The DNA damage response: making it safe to play with

knives. Mol Cell 2010;40:179–204.4. Harper JW, Elledge SJ. The DNA damage response: ten years after. Mol Cell

2007;28:739–45.5. Sorensen CS, Syljuasen RG. Safeguarding genome integrity: the checkpoint

kinases ATR, CHK1 and WEE1 restrain CDK activity during normal DNAreplication. Nucleic Acids Res 2012;40:477–86.

6. Jones RM, Petermann E. Replication fork dynamics and the DNA damageresponse. Biochem J 2012;443:13–26.

7. McNeely S, Beckmann R, Bence Lin AK. CHEK again: revisiting the devel-opment of CHK1 inhibitors for cancer therapy. Pharmacol Ther 2014;142:1–10.

8. Parker LL, Piwnica-WormsH. Inactivation of the p34cdc2-cyclin B complexby the human WEE1 tyrosine kinase. Science 1992;257:1955–7.

9. WatanabeN,BroomeM,Hunter T. Regulationof thehumanWEE1HuCDKtyrosine 15-kinase during the cell cycle. EMBO J 1995;14:1878–91.

10. Do K, Doroshow JH, Kummar S.Wee1 kinase as a target for cancer therapy.Cell Cycle 2013;12:3159–64.

11. MathesonCJ, BackosDS, ReiganP. TargetingWEE1kinase in cancer. TrendsPharmacol Sci 2016;37:872–81.

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

13. Weber AM, Ryan AJ. ATM and ATR as therapeutic targets in cancer.Pharmacol Ther 2015;149:124–38.

14. Ma CX, Janetka JW, Piwnica-Worms H. Death by releasing thebreaks: CHK1 inhibitors as cancer therapeutics. Trends Mol Med2011;17:88–96.

15. Toledo LI, Murga M, Fernandez-Capetillo O. Targeting ATR and Chk1kinases for cancer treatment: a new model for new (and old) drugs. MolOncol 2011;5:368–73.

16. Thompson R, Eastman A. The cancer therapeutic potential of Chk1 inhi-bitors: how mechanistic studies impact on clinical trial design. Br J ClinPharmacol 2013;76:358–69.

17. Calvo E, Braiteh F, VonHoff D, McWilliams R, Becerra C, Galsky MD, et al.Phase I study of CHK1 inhibitor LY2603618 in combination with gemci-tabine in patients with solid tumors. Oncology 2016;91:251–60.

18. Calvo E, Chen VJ, Marshall M, Ohnmacht U, Hynes SM, Kumm E, et al.Preclinical analyses and phase I evaluation of LY2603618 administered incombination with pemetrexed and cisplatin in patients with advancedcancer. Invest New Drugs 2014;32:955–68.

19. Daud AI, Ashworth MT, Strosberg J, Goldman JW, Mendelson D, SpringettG, et al. Phase I dose-escalation trial of checkpoint kinase 1 inhibitor MK-8776 as monotherapy and in combination with gemcitabine in patientswith advanced solid tumors. J Clin Oncol 2015;33:1060–6.

20. Doi T, Yoshino T, Shitara K, Matsubara N, Fuse N, Naito Y, et al. Phase Istudy of LY2603618, a CHK1 inhibitor, in combination with gemcita-bine in Japanese patients with solid tumors. Anticancer Drugs 2015;26:1043–53.

21. Infante JR, Hollebecque A, Postel-Vinay S, Bauer TM, Blackwood E,Evangelista M, et al. Phase I study of GDC-0425, a checkpoint kinase1 inhibitor, in combination with gemcitabine in patients with refrac-tory solid tumors [abstract]. In: Proceedings of the 106th AnnualMeeting of the American Association for Cancer Research; 2015Apr 18–22; Philadelphia, PA. Philadelphia (PA): AACR; 2015.Abstract nr CT139.

22. Karp JE, Thomas BM, Greer JM, Sorge C, Gore SD, Pratz KW, et al. Phase Iand pharmacologic trial of cytosine arabinoside with the selective check-point 1 inhibitor Sch 900776 in refractory acute leukemias. ClinCancer Res2012;18:6723–31.

23. Sausville E, Lorusso P, Carducci M, Carter J, Quinn MF, Malburg L, et al.Phase I dose-escalation study of AZD7762, a checkpoint kinase inhibitor,in combination with gemcitabine in US patients with advanced solidtumors. Cancer Chemother Pharmacol 2014;73:539–49.

24. Seto T, Esaki T, Hirai F, Arita S, Nosaki K, Makiyama A, et al. Phase I, dose-escalation study of AZD7762 alone and in combination with gemcitabinein Japanese patients with advanced solid tumours. Cancer ChemotherPharmacol 2013;72:619–27.

25. Weiss GJ, Donehower RC, Iyengar T, Ramanathan RK, Lewandowski K,Westin E, et al. Phase I dose-escalation study to examine the safety andtolerability of LY2603618, a checkpoint 1 kinase inhibitor, administered 1day after pemetrexed 500 mg/m(2) every 21 days in patients with cancer.Invest New Drugs 2013;31:136–44.

26. Scagliotti G, Kang JH, Smith D, Rosenberg R, Park K, Kim SW, et al. Phase IIevaluation of LY2603618, a first-generation CHK1 inhibitor, in combina-tion with pemetrexed in patients with advanced or metastatic non-smallcell lung cancer. Invest New Drugs 2016;34:625–35.

27. Webster JA, Tibes R, Blackford A, Morris L, Patnaik MM, Wang L, et al.Randomized phase II trial of timed sequential cytosine arabinoside withand without the CHK1 inhibitor MK-8876 in adults with relapsed andrefractory acute myelogenous leukemia. Blood 2015;126:2563.

28. Do K, Wilsker D, Ji J, Zlott J, Freshwater T, Kinders RJ, et al. Phase I study ofsingle-agent AZD1775 (MK-1775), a Wee1 kinase inhibitor, in patientswith refractory solid tumors. J Clin Oncol 2015;33:3409–15.

29. HongD, Infante J, JankuF, Jones S,Nguyen LM, BurrisH, et al. Phase I studyof LY2606368, a checkpoint kinase 1 inhibitor, in patients with advancedcancer. J Clin Oncol 2016;34:1764–71.

30. Zabludoff SD, Deng C, Grondine MR, Sheehy AM, Ashwell S, Caleb BL,et al. AZD7762, a novel checkpoint kinase inhibitor, drives checkpointabrogation and potentiates DNA-targeted therapies. Mol Cancer Ther2008;7:2955–66.

31. Hirai H, Iwasawa Y, Okada M, Arai T, Nishibata T, Kobayashi M, et al.Small-molecule inhibition of Wee1 kinase by MK-1775 selectively sensi-tizes p53-deficient tumor cells to DNA-damaging agents. Mol Cancer Ther2009;8:2992–3000.

32. King C, Diaz H, Barnard D, Barda D, Clawson D, Blosser W, et al.Characterization and preclinical development of LY2603618: a selectiveand potent Chk1 inhibitor. Invest New Drugs 2014;32:213–26.

33. Rajeshkumar NV, De Oliveira E, Ottenhof N, Watters J, Brooks D, DemuthT, et al. MK-1775, a potent Wee1 inhibitor, synergizes with gemcitabine toachieve tumor regressions, selectively in p53-deficient pancreatic cancerxenografts. Clin Cancer Res 2011;17:2799–806.

Lin et al.

Clin Cancer Res; 23(13) July 1, 2017 Clinical Cancer Research3238

on January 14, 2021. © 2017 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst March 22, 2017; DOI: 10.1158/1078-0432.CCR-16-0083

Page 8: Achieving Precision Death with Cell-Cycle Inhibitors that ......inevitable for cell-cycle inhibitors. Hence, in the 1990s, cancer drug discovery shifted focus from cell-cycle targets

34. Reaper PM,GriffithsMR, Long JM,Charrier JD,Maccormick S,CharltonPA,et al. Selective killing of ATM- or p53-deficient cancer cells throughinhibition of ATR. Nat Chem Biol 2011;7:428–30.

35. Toledo LI, Murga M, Zur R, Soria R, Rodriguez A, Martinez S, et al. Acell-based screen identifies ATR inhibitors with synthetic lethal proper-ties for cancer-associated mutations. Nat Struct Mol Biol 2011;18:721–7.

36. Leijen S, van Geel RM, Sonke GS, de Jong D, Rosenberg EH, Marchetti S,et al. Phase II study of WEE1 inhibitor AZD1775 plus carboplatin inpatients with TP53-mutated ovarian cancer refractory or resistant tofirst-line therapy within 3 months. J Clin Oncol 2016;34:4354–61.

37. Oza AM,Weberpals JI, ProvencherDM,Grischke E-M,HallM,UyarD, et al.An international, biomarker-directed, randomized, phase II trial ofAZD1775 plus paclitaxel and carboplatin (P/C) for the treatment ofwomenwith platinum-sensitive, TP53-mutant ovarian cancer. J ClinOncol33, 2015 (suppl; abstr 5506).

38. Leijen S, van Geel RM, Pavlick AC, Tibes R, Rosen L, Razak AR, et al. Phase Istudy evaluating WEE1 inhibitor AZD1775 as monotherapy and in com-bination with gemcitabine, cisplatin, or carboplatin in patients withadvanced solid tumors. J Clin Oncol 2016;34:4371–80.

39. Lord CJ, Ashworth A. BRCAness revisited. Nat Rev Cancer 2016;16:110–20.

40. McCabe N, Turner NC, Lord CJ, Kluzek K, Bialkowska A, Swift S, et al.Deficiency in the repair of DNA damage by homologous recombinationand sensitivity to poly(ADP-ribose) polymerase inhibition. Cancer Res2006;66:8109–15.

41. Bryant HE, Schultz N, Thomas HD, Parker KM, Flower D, Lopez E, et al.Specific killing of BRCA2-deficient tumours with inhibitors of poly(ADP-ribose) polymerase. Nature 2005;434:913–7.

42. Farmer H, McCabe N, Lord CJ, Tutt AN, Johnson DA, Richardson TB, et al.Targeting the DNA repair defect in BRCA mutant cells as a therapeuticstrategy. Nature 2005;434:917–21.

43. Dobbelstein M, Sorensen CS. Exploiting replicative stress to treat cancer.Nat Rev Drug Discov 2015;14:405–23.

44. LeconaE, Fernandez-CapetilloO. Replication stress and cancer: it takes twoto tango. Exp Cell Res 2014;329:26–34.

45. Hills SA, Diffley JF. DNA replication and oncogene-induced replicativestress. Curr Biol 2014;24:R435–44.

46. Gaillard H, Garcia-Muse T, Aguilera A. Replication stress and cancer. NatRev Cancer 2015;15:276–89.

47. Zeman MK, Cimprich KA. Causes and consequences of replication stress.Nat Cell Biol 2014;16:2–9.

48. Mazouzi A, Velimezi G, Loizou JI. DNA replication stress: causes, resolu-tion and disease. Exp Cell Res 2014;329:85–93.

49. Murga M, Campaner S, Lopez-Contreras AJ, Toledo LI, Soria R, MontanaMF, et al. Exploiting oncogene-induced replicative stress for the selectivekilling of Myc-driven tumors. Nat Struct Mol Biol 2011;18:1331–5.

50. Maya-Mendoza A, Petermann E, Gillespie DA, Caldecott KW, Jackson DA.Chk1 regulates the density of active replication origins during the verte-brate S phase. EMBO J 2007;26:2719–31.

51. Petermann E, Woodcock M, Helleday T. Chk1 promotes replication forkprogression by controlling replication initiation. Proc Natl Acad Sci U S A2010;107:16090–5.

52. Cole KA,Huggins J, LaquagliaM,HuldermanCE, RussellMR, Bosse K, et al.RNAi screen of the protein kinome identifies checkpoint kinase 1 (CHK1)as a therapeutic target in neuroblastoma. Proc Natl Acad Sci U S A2011;108:3336–41.

53. David L, Fernandez-Vidal A, Bertoli S, Grgurevic S, Lepage B, Deshaies D,et al. CHK1 as a therapeutic target to bypass chemoresistance in AML. SciSignal 2016;9:ra90.

54. Derenzini E, Agostinelli C, Imbrogno E, Iacobucci I, Casadei B, Brighenti E,et al. Constitutive activation of the DNA damage response pathway as anovel therapeutic target in diffuse large B-cell lymphoma. Oncotarget2015;6:6553–69.

55. Ghelli Luserna Di Rora A, Iacobucci I, Imbrogno E, Papayannidis C,Derenzini E, Ferrari A, et al. Prexasertib, a Chk1/Chk2 inhibitor, increasesthe effectiveness of conventional therapy in B-/T- cell progenitor acutelymphoblastic leukemia. Oncotarget 2016;7:53377–91.

56. May C, Beckmann R, Blosser W, Dowless M, VanWye A, Burke T, et al.Targeting checkpoint kinase 1 (CHK1) with the small molecule inhibitorLY2606368mesylate monohydrate in models of high-risk pediatric cancer

yields significant antitumor effects [abstract]. In: Proceedings of the 107thAnnualMeeting of theAmericanAssociation forCancer Research; 2016Apr16-20; NewOrleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14Suppl):Abstract nr 2458.

57. Mohni KN, Kavanaugh GM, Cortez D. ATR pathway inhibition is synthet-ically lethal in cancer cells with ERCC1 deficiency. Cancer Res 2014;74:2835–45.

58. Morgado-Palacin I, Day A, Murga M, Lafarga V, Anton ME, Tubbs A, et al.Targeting the kinase activities of ATR andATMexhibits antitumoral activityin mouse models of MLL-rearranged AML. Sci Signal 2016;9:ra91.

59. Nieto-Soler M,Morgado-Palacin I, Lafarga V, Lecona E, MurgaM, Callen E,et al. Efficacy of ATR inhibitors as single agents in Ewing sarcoma.Oncotarget 2016;7:58759–67.

60. Pfister SX, Markkanen E, Jiang Y, Sarkar S, Woodcock M, Orlando G, et al.Inhibiting WEE1 selectively kills histone H3K36me3-deficient cancers bydNTP starvation. Cancer Cell 2015;28:557–68.

61. Restelli V, Chila R, Lupi M, Rinaldi A, Kwee I, Bertoni F, et al. Character-ization of a mantle cell lymphoma cell line resistant to the Chk1 inhibitorPF-00477736. Oncotarget 2015;6:37229–40.

62. Saini P, Li Y, Dobbelstein M. Wee1 is required to sustain ATR/Chk1signaling upon replicative stress. Oncotarget 2015;6:13072–87.

63. Sarmento LM, Povoa V, Nascimento R, Real G, Antunes I, Martins LR, et al.CHK1 overexpression in T-cell acute lymphoblastic leukemia is essentialfor proliferation and survival by preventing excessive replication stress.Oncogene 2015;34:2978–90.

64. Kwok M, Davies N, Agathanggelou A, Smith E, Oldreive C, Petermann E,et al. ATR inhibition induces synthetic lethality and overcomes chemore-sistance in TP53- or ATM-defective chronic lymphocytic leukemia cells.Blood 2016;127:582–95.

65. Yap TA, Luken MJdM, O'Carrigan B, Roda D, Papadatos-Pastos D, LorenteD, et al. Phase I trial offirst-in-class ataxia telangiectasia-mutated andRad3-related (ATR) inhibitor VX-970 as monotherapy (mono) or in combina-tion with carboplatin (CP) in advanced cancer patients (pts) with prelim-inary evidence of target modulation and antitumor activity. Mol CancerTher 2015;14(12 Suppl 2):PR14.

66. Gerald WL, Haber DA. The EWS-WT1 gene fusion in desmoplastic smallround cell tumor. Semin Cancer Biol 2005;15:197–205.

67. Shern JF, Chen L, Chmielecki J, Wei JS, Patidar R, Rosenberg M, et al.Comprehensive genomic analysis of rhabdomyosarcoma reveals a land-scape of alterations affecting a common genetic axis in fusion-positive andfusion-negative tumors. Cancer Discov 2014;4:216–31.

68. Harris PS, Venkataraman S, Alimova I, Birks DK, Balakrishnan I, CristianoB, et al. Integrated genomic analysis identifies the mitotic checkpointkinaseWEE1 as a novel therapeutic target inmedulloblastoma.Mol Cancer2014;13:72.

69. MoreiraD, Venkataraman S, Balakrishnan I, Prince E, ForemanN, VibhakarR. MB-71 The role of wee1 in Myc-driven medulloblastoma. Neuro Oncol2016;18(suppl_3):iii113.

70. Kahen E, Yu D, Harrison DJ, Clark J, Hingorani P, Cubitt CL, et al.Identification of clinically achievable combination therapies in childhoodrhabdomyosarcoma. Cancer Chemother Pharmacol 2016;78:313–23.

71. Mueller S, Hashizume R, Yang X, Kolkowitz I, Olow AK, Phillips J, et al.Targeting Wee1 for the treatment of pediatric high-grade gliomas. NeuroOncol 2014;16:352–60.

72. Carrassa L, Chila R, Lupi M, Ricci F, Celenza C, Mazzoletti M, et al.Combined inhibition of Chk1 and Wee1: in vitro synergistic effect trans-lates to tumor growth inhibition invivo. Cell Cycle 2012;11:2507–17.

73. Guertin AD, Martin MM, Roberts B, Hurd M, Qu X, Miselis NR, et al.Unique functions of CHK1 and WEE1 underlie synergistic anti-tumoractivity upon pharmacologic inhibition. Cancer Cell Int 2012;12:45.

74. Sanjiv K, Hagenkort A, Calderon-Montano JM, Koolmeister T, Reaper PM,Mortusewicz O, et al. Cancer-specific synthetic lethality between ATR andCHK1 kinase activities. Cell Rep 2016;14:298–309.

75. Kennedy RD, D'Andrea AD. The Fanconi Anemia/BRCA pathway: newfaces in the crowd. Genes Dev 2005;19:2925–40.

76. Chen CC, Kennedy RD, Sidi S, Look AT, D'Andrea A. CHK1 inhibition as astrategy for targeting Fanconi Anemia (FA) DNA repair pathway deficienttumors. Mol Cancer 2009;8:24.

77. Olaussen KA, Dunant A, Fouret P, Brambilla E, Andre F, Haddad V, et al.DNA repair by ERCC1 in non-small-cell lung cancer and cisplatin-basedadjuvant chemotherapy. N Engl J Med 2006;355:983–91.

Selective Targeting of Checkpoint Kinase Inhibitors

www.aacrjournals.org Clin Cancer Res; 23(13) July 1, 2017 3239

on January 14, 2021. © 2017 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst March 22, 2017; DOI: 10.1158/1078-0432.CCR-16-0083

Page 9: Achieving Precision Death with Cell-Cycle Inhibitors that ......inevitable for cell-cycle inhibitors. Hence, in the 1990s, cancer drug discovery shifted focus from cell-cycle targets

78. Sultana R, Abdel-Fatah T, Perry C, Moseley P, Albarakti N, Mohan V, et al.Ataxia telangiectasia mutated and Rad3 related (ATR) protein kinaseinhibition is synthetically lethal in XRCC1 deficient ovarian cancer cells.PLoS One 2013;8:e57098.

79. Sorensen CS, Hansen LT, Dziegielewski J, Syljuasen RG, Lundin C, Bartek J,et al. The cell-cycle checkpoint kinase Chk1 is required for mammalianhomologous recombination repair. Nat Cell Biol 2005;7:195–201.

80. Booth L, Cruickshanks N, Ridder T, Dai Y, Grant S, Dent P. PARP and CHKinhibitors interact to cause DNA damage and cell death in mammarycarcinoma cells. Cancer Biol Ther 2013;14:458–65.

81. Kim H, George E, Ragland RL, Rafail S, Zhang R, Krepler C, et al. Targetingthe ATR/CHK1 axis with PARP inhibition results in tumor regression inBRCAmutant ovarian cancer models. Clin Cancer Res 2016 Dec 19. [Epubahead of print].

82. Konstantinopoulos PA, Ceccaldi R, Shapiro GI, D'Andrea AD. Homolo-gous recombination deficiency: exploiting the fundamental vulnerabilityof ovarian cancer. Cancer Discov 2015;5:1137–54.

83. Bowtell DD, Bohm S, Ahmed AA, Aspuria PJ, Bast RCJr, Beral V,et al. Rethinking ovarian cancer II: reducing mortality fromhigh-grade serous ovarian cancer. Nat Rev Cancer 2015;15:668–79.

84. EtemadmoghadamD,Weir BA, Au-YeungG, Alsop K,Mitchell G, George J,et al. Synthetic lethality between CCNE1 amplification and loss of BRCA1.Proc Natl Acad Sci U S A 2013;110:19489–94.

85. Jones RM, Mortusewicz O, Afzal I, Lorvellec M, Garcia P, HelledayT, et al. Increased replication initiation and conflicts with tran-scription underlie Cyclin E-induced replication stress. Oncogene2013;32:3744–53.

86. Lee J-M, Karzai FH, Zimmer A, Annunziata CM, Lipkowitz S, Parker B, et al.A phase II study of the cell cycle checkpoint kinases 1 and 2 inhibitor(LY2606368; Prexasertib monomesylate monohydrate) in sporadic high-grade serous ovarian cancer (HGSOC) and germline BRCA mutation-associated ovarian cancer (gBRCAmþ OvCa). Ann Oncol 2016;27Suppl6:855O.

Clin Cancer Res; 23(13) July 1, 2017 Clinical Cancer Research3240

Lin et al.

on January 14, 2021. © 2017 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst March 22, 2017; DOI: 10.1158/1078-0432.CCR-16-0083

Page 10: Achieving Precision Death with Cell-Cycle Inhibitors that ......inevitable for cell-cycle inhibitors. Hence, in the 1990s, cancer drug discovery shifted focus from cell-cycle targets

2017;23:3232-3240. Published OnlineFirst March 22, 2017.Clin Cancer Res   Aimee Bence Lin, Samuel C. McNeely and Richard P. Beckmann  DNA Replication and RepairAchieving Precision Death with Cell-Cycle Inhibitors that Target

  Updated version

  10.1158/1078-0432.CCR-16-0083doi:

Access the most recent version of this article at:

   

   

  Cited articles

  http://clincancerres.aacrjournals.org/content/23/13/3232.full#ref-list-1

This article cites 82 articles, 21 of which you can access for free at:

  Citing articles

  http://clincancerres.aacrjournals.org/content/23/13/3232.full#related-urls

This article has been cited by 6 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://clincancerres.aacrjournals.org/content/23/13/3232To request permission to re-use all or part of this article, use this link

on January 14, 2021. © 2017 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst March 22, 2017; DOI: 10.1158/1078-0432.CCR-16-0083