Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

State-of-the-art strategies for targeting the DNA damage response in cancer

Abstract

Genomic instability is a key hallmark of cancer that arises owing to defects in the DNA damage response (DDR) and/or increased replication stress. These alterations promote the clonal evolution of cancer cells via the accumulation of driver aberrations, including gene copy-number changes, rearrangements and mutations; however, these same defects also create vulnerabilities that are relatively specific to cancer cells, which could potentially be exploited to increase the therapeutic index of anticancer treatments and thereby improve patient outcomes. The discovery that BRCA-mutant cancer cells are exquisitely sensitive to inhibition of poly(ADP-ribose) polymerase has ushered in a new era of research on biomarker-driven synthetic lethal treatment strategies for different cancers. The therapeutic landscape of antitumour agents targeting the DDR has rapidly expanded to include inhibitors of other key mediators of DNA repair and replication, such as ATM, ATR, CHK1 and CHK2, DNA-PK and WEE1. Efforts to optimize these therapies are ongoing across a range of cancers, involving the development of predictive biomarker assays of responsiveness (beyond BRCA mutations), assessment of the mechanisms underlying intrinsic and acquired resistance, and evaluation of rational, tolerable combinations with standard-of-care treatments (such as chemotherapeutics and radiation), novel molecularly targeted agents and immune-checkpoint inhibitors. In this Review, we discuss the current status of anticancer therapies targeting the DDR.

Key points

  • The DNA damage response (DDR) involves a complex network of genes responsible for sensing and responding to specific types of DNA damage, encompassing specific machineries mediating DNA repair, cell cycle regulation, replication stress responses and apoptosis.

  • Defects in the DDR give rise to genomic instability in cells, aiding in cancer initiation and progression via mutation accumulation, but also providing targetable vulnerabilities relatively specific to cancer cells that can be exploited for clinical benefit with the use of DDR inhibitors.

  • Targeting BRCA1/2-deficient cancers using poly(ADP-ribose) polymerase (PARP) inhibitors is the archetype of synthetic lethality, but now the therapeutic landscape of DDR inhibitors is rapidly expanding; bridging preclinical data on each of these agents to the clinical setting is vital to inform appropriate biomarkers and timing for their use.

  • Preclinical and clinical data on DDR inhibitors indicate that biomarkers of response and resistance extend beyond BRCA1/2 to provide a more inclusive and functionally informed approach to patient selection.

  • Preclinical and clinical research with PARP inhibition has revealed multiple resistance mechanisms across a variety of cancer subtypes, highlighting the need for functional biomarkers and sequential or combination treatment strategies.

  • While impressive clinical responses can be seen rarely with the use of single-agent DDR inhibitors, a multitude of biologically informed combination treatment strategies using a backbone of DDR inhibitors are under development to extend their use to larger populations, while minimizing overlapping toxicities.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: DNA damage response pathways being targeted in the clinic.
Fig. 2: Timeline of key events leading to FDA approvals of PARP inhibitors in cancer medicine.
Fig. 3: Mechanisms of resistance to PARP inhibitors.
Fig. 4: Biomarker-driven combination strategies to augment PARP inhibitor responses.

Similar content being viewed by others

References

  1. Roos, W. P., Thomas, A. D. & Kaina, B. DNA damage and the balance between survival and death in cancer biology. Nat. Rev. Cancer 16, 20–33 (2016).

    CAS  PubMed  Google Scholar 

  2. Xu, Y. DNA damage: a trigger of innate immunity but a requirement for adaptive immune homeostasis. Nat. Rev. Immunol. 6, 261–270 (2006).

    CAS  PubMed  Google Scholar 

  3. Hanahan, D. & Weinberg, R. A. Hallmarks of cancer: the next generation. Cell 144, 646–674 (2011).

    CAS  PubMed  Google Scholar 

  4. Tubbs, A. & Nussenzweig, A. Endogenous DNA damage as a source of genomic instability in cancer. Cell 168, 644–656 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Parkes, E. E. et al. Activation of STING-dependent innate immune signaling by S-phase-specific DNA damage in breast cancer. J. Natl. Cancer Inst. 109, djw199 (2017).

    Google Scholar 

  6. Ferguson, L. R. et al. Genomic instability in human cancer: Molecular insights and opportunities for therapeutic attack and prevention through diet and nutrition. Semin. Cancer Biol. 35 (Suppl.), 5–24 (2015).

    Google Scholar 

  7. Lord, C. J. & Ashworth, A. PARP inhibitors: synthetic lethality in the clinic. Science 355, 1152–1158 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Fong, P. C. et al. Poly(ADP)-ribose polymerase inhibition: frequent durable responses in BRCA carrier ovarian cancer correlating with platinum-free interval. J. Clin. Oncol. 28, 2512–2519 (2010).

    CAS  PubMed  Google Scholar 

  9. Fong, P. C. et al. Inhibition of poly(ADP-ribose) polymerase in tumors from BRCA mutation carriers. N. Engl. J. Med. 361, 123–134 (2009).

    CAS  PubMed  Google Scholar 

  10. Bryant, H. E. et al. Specific killing of BRCA2-deficient tumours with inhibitors of poly(ADP-ribose) polymerase. Nature 434, 913–917 (2005).

    CAS  PubMed  Google Scholar 

  11. Kaufman, B. et al. Olaparib monotherapy in patients with advanced cancer and a germline BRCA1/2 mutation. J. Clin. Oncol. 33, 244–250 (2015).

    CAS  PubMed  Google Scholar 

  12. Ashworth, A. & Lord, C. J. Synthetic lethal therapies for cancer: what’s next after PARP inhibitors? Nat. Rev. Clin. Oncol. 15, 564–576 (2018).

    CAS  PubMed  Google Scholar 

  13. Murai, J. et al. Trapping of PARP1 and PARP2 by clinical PARP inhibitors. Cancer Res. 72, 5588–5599 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Satoh, M. S. & Lindahl, T. Role of poly(ADP-ribose) formation in DNA repair. Nature 356, 356–358 (1992).

    CAS  PubMed  Google Scholar 

  15. Kedar, P. S., Stefanick, D. F., Horton, J. K. & Wilson, S. H. Increased PARP-1 association with DNA in alkylation damaged, PARP-inhibited mouse fibroblasts. Mol. Cancer Res. 10, 360–368 (2012).

    CAS  PubMed  Google Scholar 

  16. Carney, B. et al. Target engagement imaging of PARP inhibitors in small-cell lung cancer. Nat. Commun. 9, 176 (2018).

    PubMed  PubMed Central  Google Scholar 

  17. Brown, J. S., Kaye, S. B. & Yap, T. A. PARP inhibitors: the race is on. Br. J. Cancer 114, 713–715 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Swisher, E. M. et al. Rucaparib in relapsed, platinum-sensitive high-grade ovarian carcinoma (ARIEL2 Part 1): an international, multicentre, open-label, phase 2 trial. Lancet Oncol. 18, 75–87 (2017).

    CAS  PubMed  Google Scholar 

  19. Gelmon, K. A. et al. Olaparib in patients with recurrent high-grade serous or poorly differentiated ovarian carcinoma or triple-negative breast cancer: a phase 2, multicentre, open-label, non-randomised study. Lancet Oncol. 12, 852–861 (2011).

    CAS  PubMed  Google Scholar 

  20. Kaye, S. B. et al. Phase II, open-label, randomized, multicenter study comparing the efficacy and safety of olaparib, a poly (ADP-ribose) polymerase inhibitor, and pegylated liposomal doxorubicin in patients with BRCA1 or BRCA2 mutations and recurrent ovarian cancer. J. Clin. Oncol. 30, 372–379 (2012).

    CAS  PubMed  Google Scholar 

  21. Oza, A. M. et al. Antitumor activity and safety of the PARP inhibitor rucaparib in patients with high-grade ovarian carcinoma and a germline or somatic BRCA1 or BRCA2 mutation: integrated analysis of data from Study 10 and ARIEL2. Gynecol. Oncol. 147, 267–275 (2017).

    CAS  PubMed  Google Scholar 

  22. Sandhu, S. K. et al. The poly(ADP-ribose) polymerase inhibitor niraparib (MK4827) in BRCA mutation carriers and patients with sporadic cancer: a phase 1 dose-escalation trial. Lancet Oncol. 14, 882–892 (2013).

    CAS  PubMed  Google Scholar 

  23. de Bono, J. et al. Phase I, dose-escalation, two-part trial of the PARP inhibitor talazoparib in patients with advanced germline BRCA1/2 mutations and selected sporadic cancers. Cancer Discov. 7, 620–629 (2017).

    PubMed  PubMed Central  Google Scholar 

  24. Litton, J. K. et al. Talazoparib in patients with advanced breast cancer and a germline BRCA mutation. N. Engl. J. Med. 379, 753–763 (2018).

    CAS  PubMed  Google Scholar 

  25. Hong, R. et al. 53BP1 depletion causes PARP inhibitor resistance in ATM-deficient breast cancer cells. BMC Cancer 16, 725 (2016).

    PubMed  PubMed Central  Google Scholar 

  26. Jaspers, J. E. et al. Loss of 53BP1 causes PARP inhibitor resistance in Brca1-mutated mouse mammary tumors. Cancer Discov. 3, 68–81 (2013).

    CAS  PubMed  Google Scholar 

  27. Sonnenblick, A., de Azambuja, E., Azim, H. A. & Piccart, M. An update on PARP inhibitors — moving to the adjuvant setting. Nat. Rev. Clin. Oncol. 12, 27–41 (2015).

    CAS  PubMed  Google Scholar 

  28. Moore, K. N., Mirza, M. R. & Matulonis, U. A. The poly (ADP ribose) polymerase inhibitor niraparib: management of toxicities. Gynecol. Oncol. 149, 214–220 (2018).

    CAS  PubMed  Google Scholar 

  29. Kim, G. et al. FDA Approval Summary: Olaparib monotherapy in patients with deleterious germline BRCA-mutated advanced ovarian cancer treated with three or more lines of chemotherapy. Clin. Cancer Res. 21, 4257–4261 (2015).

    CAS  PubMed  Google Scholar 

  30. Balasubramaniam, S. et al. FDA Approval Summary: Rucaparib for the treatment of patients with deleterious BRCA mutation-associated advanced ovarian cancer. Clin. Cancer Res. 23, 7165–7170 (2017).

    CAS  PubMed  Google Scholar 

  31. Robson, M. et al. Olaparib for metastatic breast cancer in patients with a germline BRCA mutation. N. Engl. J. Med. 377, 523–533 (2017).

    CAS  PubMed  Google Scholar 

  32. National Comprehensive Cancer Network. NCCN guidelines. NCCN https://www.nccn.org/professionals/physician_gls/pdf/ovarian.pdf (2017).

  33. Ledermann, J. et al. Olaparib maintenance therapy in platinum-sensitive relapsed ovarian cancer. N. Engl. J. Med. 366, 1382–1392 (2012).

    CAS  PubMed  Google Scholar 

  34. Ledermann, J. et al. Olaparib maintenance therapy in patients with platinum-sensitive relapsed serous ovarian cancer: a preplanned retrospective analysis of outcomes by BRCA status in a randomised phase 2 trial. Lancet Oncol. 15, 852–861 (2014).

    CAS  PubMed  Google Scholar 

  35. Ledermann, J. A. et al. Overall survival in patients with platinum-sensitive recurrent serous ovarian cancer receiving olaparib maintenance monotherapy: an updated analysis from a randomised, placebo-controlled, double-blind, phase 2 trial. Lancet Oncol. 17, 1579–1589 (2016).

    CAS  PubMed  Google Scholar 

  36. Pujade-Lauraine, E. et al. Olaparib tablets as maintenance therapy in patients with platinum-sensitive, relapsed ovarian cancer and a BRCA1/2 mutation (SOLO2/ENGOT-Ov21): a double-blind, randomised, placebo-controlled, phase 3 trial. Lancet Oncol. 18, 1274–1284 (2017).

    CAS  PubMed  Google Scholar 

  37. Mirza, M. R. et al. Niraparib maintenance therapy in platinum-sensitive, recurrent ovarian cancer. N. Engl. J. Med. 375, 2154–2164 (2016).

    CAS  PubMed  Google Scholar 

  38. Coleman, R. L. et al. Rucaparib maintenance treatment for recurrent ovarian carcinoma after response to platinum therapy (ARIEL3): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 390, 1949–1961 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Kristeleit, R. S. et al. Clinical activity of the poly(ADP-ribose) polymerase (PARP) inhibitor rucaparib in patients (pts) with high-grade ovarian carcinoma (HGOC) and a BRCA mutation (BRCAmut): Analysis of pooled data from Study 10 (parts 1, 2a, and 3) and ARIEL2 (parts 1 and 2). Ann. Oncol. 27, 8560–8560 (2016).

    Google Scholar 

  40. Wooster, R. et al. Localization of a breast cancer susceptibility gene, BRCA2, to chromosome 13q12-13. Science 265, 2088–2090 (1994).

    CAS  PubMed  Google Scholar 

  41. Miki, Y. et al. A strong candidate for the breast and ovarian cancer susceptibility gene BRCA1. Science 266, 66–71 (1994).

    CAS  PubMed  Google Scholar 

  42. King, M.-C., Marks, J. H. & Mandell, J. B., New York Breast Cancer Study Group. Breast and ovarian cancer risks due to inherited mutations in BRCA1 and BRCA2. Science 302, 643–646 (2003).

    CAS  PubMed  Google Scholar 

  43. Mandelker, D. et al. Mutation detection in patients with advanced cancer by universal sequencing of cancer-related genes in tumor and normal DNA versus guideline-based germline testing. JAMA 318, 825–835 (2017).

    PubMed  PubMed Central  Google Scholar 

  44. Pritchard, C. C. et al. Inherited DNA-repair gene mutations in men with metastatic prostate cancer. N. Engl. J. Med. 375, 443–453 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Schrader, K. A. et al. Germline variants in targeted tumor sequencing using matched normal DNA. JAMA Oncol. 2, 104–111 (2016).

    PubMed  PubMed Central  Google Scholar 

  46. Meric-Bernstam, F. et al. Incidental germline variants in 1000 advanced cancers on a prospective somatic genomic profiling protocol. Ann. Oncol. 27, 795–800 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Shindo, K. et al. Deleterious germline mutations in patients with apparently sporadic pancreatic adenocarcinoma. J. Clin. Oncol. 35, 3382–3390 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Ettl, J. et al. Quality of life with talazoparib versus physician’s choice of chemotherapy in patients with advanced breast cancer and germline BRCA1/2 mutation: patient-reported outcomes from the EMBRACA phase III trial. Ann. Oncol. https://doi.org/10.1093/annonc/mdy257 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  49. Rugo, H. S. et al. Adaptive randomization of veliparib-carboplatin treatment in breast cancer. N. Engl. J. Med. 375, 23–34 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Loibl, S. et al. Addition of the PARP inhibitor veliparib plus carboplatin or carboplatin alone to standard neoadjuvant chemotherapy in triple-negative breast cancer (BrighTNess): a randomised, phase 3 trial. Lancet Oncol. 19, 497–509 (2018).

    CAS  PubMed  Google Scholar 

  51. Litton, J. K., Scoggins, M., Ramirez, D. & Arun, B. A pilot study of neoadjuvant talazoparib for early-stage breast cancer patients with a BRCA mutation. Ann. Oncol. 27, 43–67 (2016).

    Google Scholar 

  52. Litton, J. K. et al. Neoadjuvant talazoparib (TALA) for operable breast cancer patients with a BRCA mutation (BRCA+). J. Clin. Oncol. 36, 508–508 (2018).

    Google Scholar 

  53. Mateo, J. et al. DNA-repair defects and olaparib in metastatic prostate cancer. N. Engl. J. Med. 373, 1697–1708 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Goodall, J. et al. Circulating cell-free DNA to guide prostate cancer treatment with PARP inhibition. Cancer Discov. 7, 1006–1017 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Bendell, J. et al. Phase I study of olaparib plus gemcitabine in patients with advanced solid tumours and comparison with gemcitabine alone in patients with locally advanced/metastatic pancreatic cancer. Ann. Oncol. 26, 804–811 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Kim, H. S. et al. Concordance of ATM (ataxia telangiectasia mutated) immunohistochemistry between biopsy or metastatic tumor samples and primary tumors in gastric cancer patients. Pathobiol. J. Immunopathol. Mol. Cell. Biol. 80, 127–137 (2013).

    CAS  Google Scholar 

  57. Kubota, E. et al. Low ATM protein expression and depletion of p53 correlates with olaparib sensitivity in gastric cancer cell lines. Cell Cycle 13, 2129–2137 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Bang, Y.-J. et al. Randomized, double-blind phase II trial with prospective classification by ATM protein level to evaluate the efficacy and tolerability of olaparib plus paclitaxel in patients with recurrent or metastatic gastric cancer. J. Clin. Oncol. 33, 3858–3865 (2015).

    CAS  PubMed  Google Scholar 

  59. Bang, Y.-J. et al. Olaparib in combination with paclitaxel in patients with advanced gastric cancer who have progressed following first-line therapy (GOLD): a double-blind, randomised, placebo-controlled, phase 3 trial. Lancet Oncol. 18, 1637–1651 (2017).

    CAS  PubMed  Google Scholar 

  60. Smyth, E. Missing a GOLDen opportunity in gastric cancer. Lancet Oncol. 18, 1561–1563 (2017).

    PubMed  Google Scholar 

  61. Mo, M., Yang, J., Zhu, X. & Zhu, J. Use of olaparib in patients with advanced gastric cancer. Lancet Oncol. 19, e75 (2018).

    PubMed  Google Scholar 

  62. Ballinger, M. L. et al. Monogenic and polygenic determinants of sarcoma risk: an international genetic study. Lancet Oncol. 17, 1261–1271 (2016).

    PubMed  Google Scholar 

  63. Felsenstein, K. M. & Theodorescu, D. Precision medicine for urothelial bladder cancer: update on tumour genomics and immunotherapy. Nat. Rev. Urol. 15, 92–111 (2017).

    PubMed  Google Scholar 

  64. Daly, M. B. et al. NCCN Guidelines Insights: Genetic/familial high-risk assessment: breast and ovarian, version 2.2017. J. Natl Compr. Cancer Netw. 15, 9–20 (2017).

    CAS  Google Scholar 

  65. Knijnenburg, T. A. et al. Genomic and molecular landscape of DNA damage repair deficiency across The Cancer Genome Atlas. Cell Rep. 23, 239–254.e6 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Quinn, J. A. et al. Phase II trial of temozolomide plus o6-benzylguanine in adults with recurrent, temozolomide-resistant malignant glioma. J. Clin. Oncol. 27, 1262–1267 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Rajan, A. et al. A phase I combination study of olaparib with cisplatin and gemcitabine in adults with solid tumors. Clin. Cancer Res. 18, 2344–2351 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Dréan, A., Lord, C. J. & Ashworth, A. PARP inhibitor combination therapy. Crit. Rev. Oncol. Hematol. 108, 73–85 (2016).

    PubMed  Google Scholar 

  69. Turner, N., Tutt, A. & Ashworth, A. Hallmarks of ‘BRCAness’ in sporadic cancers. Nat. Rev. Cancer 4, 814–819 (2004).

    CAS  PubMed  Google Scholar 

  70. Metzger, M. J., Stoddard, B. L. & Monnat, R. J. PARP-mediated repair, homologous recombination, and back-up non-homologous end joining-like repair of single-strand nicks. DNA Repair 12, 529–534 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Sen, T., Gay, C. M. & Byers, L. A. Targeting DNA damage repair in small cell lung cancer and the biomarker landscape. Transl Lung Cancer Res. 7, 50–68 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Abkevich, V. et al. Patterns of genomic loss of heterozygosity predict homologous recombination repair defects in epithelial ovarian cancer. Br. J. Cancer 107, 1776–1782 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Watkins, J. A., Irshad, S., Grigoriadis, A. & Tutt, A. N. J. Genomic scars as biomarkers of homologous recombination deficiency and drug response in breast and ovarian cancers. Breast Cancer Res. 16, 211 (2014).

    PubMed  PubMed Central  Google Scholar 

  74. Wang, Z. C. et al. Profiles of genomic instability in high-grade serous ovarian cancer predict treatment outcome. Clin. Cancer Res. 18, 5806–5815 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. González Martín, A. Progress in PARP inhibitors beyond BRCA mutant recurrent ovarian cancer? Lancet Oncol. 18, 8–9 (2017).

    PubMed  Google Scholar 

  76. Garsed, D. W. et al. Homologous recombination DNA repair pathway disruption and retinoblastoma protein loss are associated with exceptional survival in high-grade serous ovarian cancer. Clin. Cancer Res. 24, 569–580 (2018).

    CAS  PubMed  Google Scholar 

  77. Peng, G. & Mills, G. B. Surviving ovarian cancer: an affair between defective DNA repair and RB1. Clin. Cancer Res. 24, 508–510 (2018).

    CAS  PubMed  Google Scholar 

  78. Alexandrov, L. B. et al. Signatures of mutational processes in human cancer. Nature 500, 415–421 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Davies, H. et al. HRDetect is a predictor of BRCA1 and BRCA2 deficiency based on mutational signatures. Nat. Med. 23, 517–525 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  80. Polak, P. et al. A mutational signature reveals alterations underlying deficient homologous recombination repair in breast cancer. Nat. Genet. 49, 1476–1486 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Lord, C. J. & Ashworth, A. BRCAness revisited. Nat. Rev. Cancer 16, 110–120 (2016).

    CAS  PubMed  Google Scholar 

  82. Gilardini Montani, M. S. et al. ATM-depletion in breast cancer cells confers sensitivity to PARP inhibition. J. Exp. Clin. Cancer Res. CR 32, 95 (2013).

    PubMed  Google Scholar 

  83. Wu, C. et al. Targeting AURKA-CDC25C axis to induce synthetic lethality in ARID1A-deficient colorectal cancer cells. Nat. Commun. 9, 3212 (2018).

    PubMed  PubMed Central  Google Scholar 

  84. Shen, J. et al. ARID1A deficiency impairs the DNA damage checkpoint and sensitizes cells to PARP inhibitors. Cancer Discov. 5, 752–767 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Yu, H. et al. Tumor suppressor and deubiquitinase BAP1 promotes DNA double-strand break repair. Proc. Natl Acad. Sci. USA 111, 285–290 (2014).

    CAS  PubMed  Google Scholar 

  86. Parrotta, R. et al. A novel BRCA1-associated protein-1 isoform affects response of mesothelioma cells to drugs impairing BRCA1-mediated DNA repair. J. Thorac. Oncol. 12, 1309–1319 (2017).

    PubMed  Google Scholar 

  87. Bajrami, I. et al. Genome-wide profiling of genetic synthetic lethality identifies CDK12 as a novel determinant of PARP1/2 inhibitor sensitivity. Cancer Res. 74, 287–297 (2014).

    CAS  PubMed  Google Scholar 

  88. Wu, Y.-M. et al. Inactivation of CDK12 delineates a distinct immunogenic class of advanced prostate cancer. Cell 173, 1770–1782 (2018).

    CAS  PubMed  Google Scholar 

  89. Yi, S. et al. Functional variomics and network perturbation: connecting genotype to phenotype in cancer. Nat. Rev. Genet. 18, 395–410 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  90. Peng, G. et al. Genome-wide transcriptome profiling of homologous recombination DNA repair. Nat. Commun. 5, 3361 (2014).

    PubMed  Google Scholar 

  91. Konstantinopoulos, P. A. et al. Gene expression profile of BRCAness that correlates with responsiveness to chemotherapy and with outcome in patients with epithelial ovarian cancer. J. Clin. Oncol. 28, 3555–3561 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  92. Severson, T. M. et al. The BRCA1ness signature is associated significantly with response to PARP inhibitor treatment versus control in the I-SPY 2 randomized neoadjuvant setting. Breast Cancer Res. 19, 99 (2017).

    PubMed  PubMed Central  Google Scholar 

  93. Wolf, D. M. et al. DNA repair deficiency biomarkers and the 70-gene ultra-high risk signature as predictors of veliparib/carboplatin response in the I-SPY 2 breast cancer trial. NPJ Breast Cancer 3, 31 (2017).

    PubMed  PubMed Central  Google Scholar 

  94. Weinstein, J. N. & Lorenzi, P. L. Cancer: discrepancies in drug sensitivity. Nature 504, 381–383 (2013).

    CAS  PubMed  Google Scholar 

  95. Sulkowski, P. L. et al. 2-Hydroxyglutarate produced by neomorphic IDH mutations suppresses homologous recombination and induces PARP inhibitor sensitivity. Sci. Transl Med. 9, eaal2463 (2017).

    PubMed  PubMed Central  Google Scholar 

  96. Pietanza, M. C. et al. Randomized, double-blind, phase II study of temozolomide in combination with either veliparib or placebo in patients with relapsed-sensitive or refractory small-cell lung cancer. J. Clin. Oncol. 36, 2386–2394 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  97. Allison Stewart, C. et al. Dynamic variations in epithelial-to-mesenchymal transition (EMT), ATM, and SLFN11 govern response to PARP inhibitors and cisplatin in small cell lung cancer. Oncotarget 8, 28575–28587 (2017).

    CAS  PubMed  Google Scholar 

  98. Byers, L. A. et al. Proteomic profiling identifies dysregulated pathways in small cell lung cancer and novel therapeutic targets including PARP1. Cancer Discov. 2, 798–811 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  99. Bélanger, F. et al. Replication protein a availability during DNA replication stress is a major determinant of cisplatin resistance in ovarian cancer cells. Cancer Res. https://doi.org/10.1158/0008-5472.CAN-18-0618 (2018).

    Article  PubMed  Google Scholar 

  100. Lu, Y. et al. Chemosensitivity of IDH1-mutated gliomas due to an impairment in PARP1-mediated DNA repair. Cancer Res. 77, 1709–1718 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Aparicio, A. M. et al. Combined tumor suppressor defects characterize clinically defined aggressive variant prostate cancers. Clin. Cancer Res. 22, 1520–1530 (2016).

    CAS  PubMed  Google Scholar 

  102. Aparicio, A. M. et al. Platinum-based chemotherapy for variant castrate-resistant prostate cancer. Clin. Cancer Res. 19, 3621–3630 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  103. McGrail, D. J. et al. Improved prediction of PARP inhibitor response and identification of synergizing agents through use of a novel gene expression signature generation algorithm. NPJ Syst. Biol. Appl. 3, 8 (2017).

    PubMed  PubMed Central  Google Scholar 

  104. Lheureux, S. et al. Long-term responders on olaparib maintenance in high-grade serous ovarian cancer: clinical and molecular characterization. Clin. Cancer Res. 23, 4086–4094 (2017).

    CAS  PubMed  Google Scholar 

  105. Kondrashova, O. et al. Secondary somatic mutations restoring RAD51C and RAD51D associated with acquired resistance to the PARP inhibitor rucaparib in high-grade ovarian carcinoma. Cancer Discov. 7, 984–998 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  106. Quigley, D. et al. Analysis of circulating cell-free DNA identifies multiclonal heterogeneity of BRCA2 reversion mutations associated with resistance to PARP inhibitors. Cancer Discov. 7, 999–1005 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  107. Patch, A.-M. et al. Whole-genome characterization of chemoresistant ovarian cancer. Nature 521, 489–494 (2015).

    CAS  PubMed  Google Scholar 

  108. Domchek, S. M. Reversion mutations with clinical use of PARP inhibitors: many genes, many versions. Cancer Discov. 7, 937–939 (2017).

    CAS  PubMed  Google Scholar 

  109. Johnson, N. et al. Stabilization of mutant BRCA1 protein confers PARP inhibitor and platinum resistance. Proc. Natl Acad. Sci. USA 110, 17041–17046 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  110. Ter Brugge, P. et al. Mechanisms of therapy resistance in patient-derived xenograft models of BRCA1-deficient breast cancer. J. Natl. Cancer Inst. 108, djw148 (2016).

    Google Scholar 

  111. Fojo, T. & Bates, S. Mechanisms of resistance to PARP inhibitors — three and counting. Cancer Discov. 3, 20–23 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  112. Ray Chaudhuri, A. et al. Replication fork stability confers chemoresistance in BRCA-deficient cells. Nature 535, 382–387 (2016).

    PubMed  Google Scholar 

  113. Lee, S.-B. et al. Tousled-like kinases stabilize replication forks and show synthetic lethality with checkpoint and PARP inhibitors. Sci. Adv. 4, eaat4985 (2018).

    PubMed  PubMed Central  Google Scholar 

  114. Murai, J. et al. Resistance to PARP inhibitors by SLFN11 inactivation can be overcome by ATR inhibition. Oncotarget 7, 76534–76550 (2016).

    PubMed  PubMed Central  Google Scholar 

  115. Lok, B. H. et al. PARP inhibitor activity correlates with SLFN11 expression and demonstrates synergy with temozolomide in small cell lung cancer. Clin. Cancer Res. 23, 523–535 (2017).

    CAS  PubMed  Google Scholar 

  116. Murai, J. et al. SLFN11 Blocks Stressed Replication Forks Independently of ATR. Mol. Cell 69, 371–384.e6 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  117. Henneman, L. et al. Selective resistance to the PARP inhibitor olaparib in a mouse model for BRCA1-deficient metaplastic breast cancer. Proc. Natl Acad. Sci. USA 112, 8409–8414 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  118. Gogola, E. et al. Selective loss of PARG restores PARylation and counteracts PARP inhibitor-mediated synthetic lethality. Cancer Cell 33, 1078–1093.e12 (2018).

    CAS  PubMed  Google Scholar 

  119. Pettitt, S. J. et al. Genome-wide and high-density CRISPR-Cas9 screens identify point mutations in PARP1 causing PARP inhibitor resistance. Nat. Commun. 9, 1849 (2018).

    PubMed  PubMed Central  Google Scholar 

  120. Yazinski, S. A. et al. ATR inhibition disrupts rewired homologous recombination and fork protection pathways in PARP inhibitor-resistant BRCA-deficient cancer cells. Genes Dev. 31, 318–332 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  121. Bitler, B. G., Watson, Z. L., Wheeler, L. J. & Behbakht, K. PARP inhibitors: Clinical utility and possibilities of overcoming resistance. Gynecol. Oncol. 147, 695–704 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  122. Cardnell, R. J. et al. Proteomic markers of DNA repair and PI3K pathway activation predict response to the PARP inhibitor BMN 673 in small cell lung cancer. Clin. Cancer Res. 19, 6322–6328 (2013).

    CAS  PubMed  Google Scholar 

  123. Mo, W. et al. mTOR inhibitors suppress homologous recombination repair and synergize with PARP inhibitors via regulating SUV39H1 in BRCA-proficient triple-negative breast cancer. Clin. Cancer Res. 22, 1699–1712 (2016).

    CAS  PubMed  Google Scholar 

  124. Sun, C. et al. Rational combination therapy with PARP and MEK inhibitors capitalizes on therapeutic liabilities in RAS mutant cancers. Sci. Transl Med. 9, eaal5148 (2017).

    PubMed  PubMed Central  Google Scholar 

  125. Li, L. et al. Androgen receptor inhibitor-induced ‘BRCAness’ and PARP inhibition are synthetically lethal for castration-resistant prostate cancer. Sci. Signal. 10, eaam7479 (2017).

    PubMed  PubMed Central  Google Scholar 

  126. Sundar, R., Brown, J., Ingles Russo, A. & Yap, T. A. Targeting ATR in cancer medicine. Curr. Probl. Cancer 41, 302–315 (2017).

    PubMed  Google Scholar 

  127. O’Carrigan, B. et al. Phase I trial of a first-in-class ATR inhibitor VX-970 as monotherapy (mono) or in combination (combo) with carboplatin (CP) incorporating pharmacodynamics (PD) studies [abstract]. J. Clin. Oncol. 34, 2504 (2016).

    Google Scholar 

  128. Yap, T. A. et al. Phase I trial of first-in-class ataxia telangiectasia-mutated and Rad3-related (ATR) inhibitor VX-970 as monotherapy (mono) or in combination with carboplatin (CP) in advanced cancer patients (pts) with preliminary evidence of target modulation and antitumor activity [abstract]. Mol. Cancer Ther. 14, R14 (2015).

    Google Scholar 

  129. Shapiro, G. et al. Phase 1 trial of first-in-class ATR inhibitor VX-970 in combination with cisplatin (Cis) in patients (pts) with advanced solid tumors (NCT02157792) [abstract]. Cancer Res. 76, CT012 (2016).

    Google Scholar 

  130. Plummer, E. R. et al. Phase I trial of first-in-class ATR inhibitor VX-970 in combination with gemcitabine (Gem) in advanced solid tumors (NCT02157792) [abstract]. J. Clin. Oncol. 34, 2513 (2016).

    Google Scholar 

  131. Dillon, M. T. et al. A Phase I dose-escalation study of ATR inhibitor monotherapy with AZD6738 in advanced solid tumors (PATRIOT Part A) [abstract]. Cancer Res. 77, CT084 (2017).

    Google Scholar 

  132. Yap, T. A. et al. Phase I modular study of AZD6738, a novel oral, potent and selective ataxia telangiectasia Rad3-related (ATR) inhibitor in combination (combo) with carboplatin, olaparib or durvalumab in patients (pts) with advanced cancers [abstract 1LBA]. Eur. J. Cancer 69, S2 (2016).

    Google Scholar 

  133. Middleton, F. K. et al. Common cancer-associated imbalances in the DNA damage response confer sensitivity to single agent ATR inhibition. Oncotarget 6, 32396–32409 (2015).

    PubMed  PubMed Central  Google Scholar 

  134. Mohni, K. N. et al. A synthetic lethal screen identifies DNA repair pathways that sensitize cancer cells to combined ATR inhibition and cisplatin treatments. PLOS ONE 10, e0125482 (2015).

    PubMed  PubMed Central  Google Scholar 

  135. Kim, H. et al. Targeting the ATR/CHK1 axis with PARP inhibition results in tumor regression in BRCA-mutant ovarian cancer models. Clin. Cancer Res. 23, 3097–3108 (2017).

    CAS  PubMed  Google Scholar 

  136. Reaper, P. M. et al. Selective killing of ATM- or p53-deficient cancer cells through inhibition of ATR. Nat. Chem. Biol. 7, 428–430 (2011).

    CAS  PubMed  Google Scholar 

  137. Williamson, C. T. et al. ATR inhibitors as a synthetic lethal therapy for tumours deficient in ARID1A. Nat. Commun. 7, 13837 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  138. VandenBussche, C. J. et al. Alternative lengthening of telomeres and ATRX/DAXX loss can be reliably detected in FNAs of pancreatic neuroendocrine tumors. Cancer 125, 544–551 (2017).

    CAS  Google Scholar 

  139. Cesare, A. J. & Reddel, R. R. Alternative lengthening of telomeres: models, mechanisms and implications. Nat. Rev. Genet. 11, 319–330 (2010).

    CAS  PubMed  Google Scholar 

  140. Flynn, R. L. et al. Alternative lengthening of telomeres renders cancer cells hypersensitive to ATR inhibitors. Science 347, 273–277 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  141. Deeg, K. I., Chung, I., Bauer, C. & Rippe, K. Cancer cells with alternative lengthening of telomeres do not display a general hypersensitivity to ATR inhibition. Front. Oncol. 6, 186 (2016).

    PubMed  PubMed Central  Google Scholar 

  142. Kim, H.-J. et al. Anti-tumor activity of the ATR inhibitor AZD6738 in HER2 positive breast cancer cells. Int. J. Cancer 140, 109–119 (2017).

    CAS  PubMed  Google Scholar 

  143. Yan, Y. et al. A novel function of HER2/Neu in the activation of G2/M checkpoint in response to γ-irradiation. Oncogene 34, 2215–2226 (2015).

    CAS  PubMed  Google Scholar 

  144. Weber, A. M. & Ryan, A. J. ATM and ATR as therapeutic targets in cancer. Pharmacol. Ther. 149, 124–138 (2015).

    CAS  PubMed  Google Scholar 

  145. Brown, J. S., O’Carrigan, B., Jackson, S. P. & Yap, T. A. Targeting DNA repair in cancer: beyond PARP inhibitors. Cancer Discov. 7, 20–37 (2017).

    CAS  PubMed  Google Scholar 

  146. Willers, H., Dahm-Daphi, J. & Powell, S. N. Repair of radiation damage to DNA. Br. J. Cancer 90, 1297–1301 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  147. Mortensen, D. S. et al. Optimization of a series of triazole containing mammalian target of rapamycin (mTOR) kinase inhibitors and the discovery of CC-115. J. Med. Chem. 58, 5599–5608 (2015).

    CAS  PubMed  Google Scholar 

  148. Munster, P. N. et al. Phase I trial of a dual TOR kinase and DNA-PK inhibitor (CC-115) in advanced solid and hematologic cancers. J. Clin. Oncol. 34, 2505 (2016).

    Google Scholar 

  149. Van Triest, B. et al. A phase Ia/Ib trial of the DNA-dependent protein kinase inhibitor (DNA-PKi) M3814 in combination with radiotherapy in patients with advanced solid tumors. J. Clin. Oncol. 35, e14048 (2016).

    Google Scholar 

  150. Matthews, T. P., Jones, A. M. & Collins, I. Structure-based design, discovery and development of checkpoint kinase inhibitors as potential anticancer therapies. Expert Opin. Drug Discov. 8, 621–640 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  151. Welch, S. et al. UCN-01 in combination with topotecan in patients with advanced recurrent ovarian cancer: a study of the Princess Margaret Hospital Phase II consortium. Gynecol. Oncol. 106, 305–310 (2007).

    CAS  PubMed  Google Scholar 

  152. Ma, C. X. et al. A phase II study of UCN-01 in combination with irinotecan in patients with metastatic triple negative breast cancer. Breast Cancer Res. Treat. 137, 483–492 (2013).

    CAS  PubMed  Google Scholar 

  153. Sausville, E. et al. Phase I dose-escalation study of AZD7762, a checkpoint kinase inhibitor, in combination with gemcitabine in US patients with advanced solid tumors. Cancer Chemother. Pharmacol. 73, 539–549 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  154. Seto, T. et al. Phase I, dose-escalation study of AZD7762 alone and in combination with gemcitabine in Japanese patients with advanced solid tumours. Cancer Chemother. Pharmacol. 72, 619–627 (2013).

    CAS  PubMed  Google Scholar 

  155. Ho, A. L. et al. Phase I, open-label, dose-escalation study of AZD7762 in combination with irinotecan (irino) in patients (pts) with advanced solid tumors. J. Clin. Oncol. 29, 3033–3033 (2011).

    Google Scholar 

  156. Al-Ahmadie, H. et al. Synthetic lethality in ATM-deficient RAD50-mutant tumors underlies outlier response to cancer therapy. Cancer Discov. 4, 1014–1021 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  157. Wehler, T. et al. A randomized, phase 2 evaluation of the CHK1 inhibitor, LY2603618, administered in combination with pemetrexed and cisplatin in patients with advanced nonsquamous non-small cell lung cancer. Lung Cancer 108, 212–216 (2017).

    PubMed  Google Scholar 

  158. Laquente, B. et al. A phase II study to evaluate LY2603618 in combination with gemcitabine in pancreatic cancer patients. BMC Cancer 17, 137 (2017).

    PubMed  PubMed Central  Google Scholar 

  159. Daud, A. I. et al. Phase I dose-escalation trial of checkpoint kinase 1 inhibitor MK-8776 as monotherapy and in combination with gemcitabine in patients with advanced solid tumors. J. Clin. Oncol. 33, 1060–1066 (2015).

    CAS  PubMed  Google Scholar 

  160. Karp, J. E. et al. Phase I and pharmacologic trial of cytosine arabinoside with the selective checkpoint 1 inhibitor Sch 900776 in refractory acute leukemias. Clin. Cancer Res. 18, 6723–6731 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  161. Hong, D. et al. Phase I study of LY2606368, a checkpoint kinase 1 inhibitor, in patients with advanced cancer. J. Clin. Oncol. 34, 1764–1771 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  162. Hong, D. S. et al. Evaluation of prexasertib, a checkpoint kinase 1 inhibitor, in a phase Ib study of patients with squamous cell carcinoma. Clin. Cancer Res. 24, 3263–3272 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  163. Lee, J. M. 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 (gBRCAm+ OvCa). Ann. Oncol. 27, 8550 (2016).

    Google Scholar 

  164. Hirai, H. et al. Small-molecule inhibition of Wee1 kinase by MK-1775 selectively sensitizes p53-deficient tumor cells to DNA-damaging agents. Mol. Cancer Ther. 8, 2992–3000 (2009).

    CAS  PubMed  Google Scholar 

  165. Aarts, M. et al. Forced mitotic entry of S-phase cells as a therapeutic strategy induced by inhibition of WEE1. Cancer Discov. 2, 524–539 (2012).

    CAS  PubMed  Google Scholar 

  166. Bridges, K. A. et al. MK-1775, a novel Wee1 kinase inhibitor, radiosensitizes p53-defective human tumor cells. Clin. Cancer Res. 17, 5638–5648 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  167. Pfister, S. X. et al. Inhibiting WEE1 selectively kills histone H3K36me3-deficient cancers by dNTP starvation. Cancer Cell 28, 557–568 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  168. Pfister, S. X. et al. SETD2-dependent histone H3K36 trimethylation is required for homologous recombination repair and genome stability. Cell Rep. 7, 2006–2018 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  169. Guertin, A. D. et al. Preclinical evaluation of the WEE1 inhibitor MK-1775 as single-agent anticancer therapy. Mol. Cancer Ther. 12, 1442–1452 (2013).

    CAS  PubMed  Google Scholar 

  170. Mizuarai, S. et al. Discovery of gene expression-based pharmacodynamic biomarker for a p53 context-specific anti-tumor drug Wee1 inhibitor. Mol. Cancer 8, 34 (2009).

    PubMed  PubMed Central  Google Scholar 

  171. Leijen, S. et al. Phase I study evaluating WEE1 inhibitor AZD1775 as monotherapy and in combination with gemcitabine, cisplatin, or carboplatin in patients with advanced solid tumors. J. Clin. Oncol. 34, 4371–4380 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  172. Do, K. et al. Phase I study of single-agent AZD1775 (MK-1775), a Wee1 kinase inhibitor, in patients with refractory solid tumors. J. Clin. Oncol. 33, 3409–3415 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  173. Leijen, S. et al. Phase II study of WEE1 inhibitor AZD1775 plus carboplatin in patients with TP53-mutated ovarian cancer refractory or resistant to first-line therapy within 3 months. J. Clin. Oncol. 34, 4354–4361 (2016).

    CAS  PubMed  Google Scholar 

  174. Oza, A. M. et al. An international, biomarker-directed, randomized, phase II trial of AZD1775 plus paclitaxel and carboplatin (P/C) for the treatment of women with platinum-sensitive, TP53-mutant ovarian cancer. J. Clin. Oncol. 33, 5506–5506 (2015).

    Google Scholar 

  175. Chera, B. S. et al. Phase Ib trial of dose-escalating AZD1775 in combination with concurrent radiation and cisplatin for intermediate and high risk head and neck squamous cell carcinoma. J. Clin. Oncol. 34, TPS6106 (2016).

    Google Scholar 

  176. Lal, S. et al. WEE1 inhibition in pancreatic cancer cells is dependent on DNA repair status in a context dependent manner. Sci. Rep. 6, 33323 (2016).

    Google Scholar 

  177. Chabot, P. et al. Veliparib in combination with whole-brain radiation therapy for patients with brain metastases from non-small cell lung cancer: results of a randomized, global, placebo-controlled study. J. Neurooncol. 131, 105–115 (2017).

    CAS  PubMed  Google Scholar 

  178. Reiss, K. A. et al. A final report of a phase I study of veliparib (ABT-888) in combination with low-dose fractionated whole abdominal radiation therapy (LDFWAR) in patients with advanced solid malignancies and peritoneal carcinomatosis with a dose escalation in ovarian and fallopian tube cancers. Gynecol. Oncol. 144, 486–490 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  179. Blumenthal, D. T. et al. A Phase III study of radiation therapy (RT) and O6-benzylguanine + BCNU versus RT and BCNU alone and methylation status in newly diagnosed glioblastoma and gliosarcoma: Southwest Oncology Group (SWOG) study S0001. Int. J. Clin. Oncol. 20, 650–658 (2015).

    CAS  PubMed  Google Scholar 

  180. Bhattacharya, S. et al. RAD51 interconnects between DNA replication, DNA repair and immunity. Nucleic Acids Res. 45, 4590–4605 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  181. Karnak, D. et al. Combined inhibition of Wee1 and PARP1/2 for radiosensitization in pancreatic cancer. Clin. Cancer Res. 20, 5085–5096 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  182. Karakashev, S. et al. BET Bromodomain inhibition synergizes with PARP inhibitor in epithelial ovarian cancer. Cell Rep. 21, 3398–3405 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  183. Yang, L. et al. Repression of BET activity sensitizes homologous recombination-proficient cancers to PARP inhibition. Sci. Transl Med. 9, eaal1645 (2017).

    PubMed  PubMed Central  Google Scholar 

  184. Sun, C. et al. BRD4 inhibition is synthetic lethal with PARP inhibitors through the induction of homologous recombination deficiency. Cancer Cell 33, 401–416.e8 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  185. O’Connor, M. J. Targeting the DNA damage response in cancer. Mol. Cell 60, 547–560 (2015).

    PubMed  Google Scholar 

  186. Ceccaldi, R. et al. Homologous-recombination-deficient tumours are dependent on Polθ-mediated repair. Nature 518, 258–262 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  187. Ibrahim, Y. H. et al. PI3K inhibition impairs BRCA1/2 expression and sensitizes BRCA-proficient triple-negative breast cancer to PARP inhibition. Cancer Discov. 2, 1036–1047 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  188. Liu, J. F. et al. Combination cediranib and olaparib versus olaparib alone for women with recurrent platinum-sensitive ovarian cancer: a randomised phase 2 study. Lancet Oncol. 15, 1207–1214 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  189. Juvekar, A. et al. Combining a PI3K inhibitor with a PARP inhibitor provides an effective therapy for BRCA1-related breast cancer. Cancer Discov. 2, 1048–1063 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  190. Carey, J. P. et al. Synthetic lethality of PARP inhibitors in combination with MYC blockade is independent of BRCA status in triple negative breast cancer. Cancer Res. 78, 742–757 (2017).

    PubMed  PubMed Central  Google Scholar 

  191. Min, A. et al. Histone deacetylase inhibitor, suberoylanilide hydroxamic acid (SAHA), enhances anti-tumor effects of the poly (ADP-ribose) polymerase (PARP) inhibitor olaparib in triple-negative breast cancer cells. Breast Cancer Res. 17, 33 (2015).

    PubMed  PubMed Central  Google Scholar 

  192. Yamaguchi, H. et al. EZH2 contributes to the response to PARP inhibitors through its PARP-mediated poly-ADP ribosylation in breast cancer. Oncogene 37, 208–217 (2018).

    CAS  PubMed  Google Scholar 

  193. Chao, O. S. & Goodman, O. B. Synergistic loss of prostate cancer cell viability by coinhibition of HDAC and PARP. Mol. Cancer Res. 12, 1755–1766 (2014).

    CAS  PubMed  Google Scholar 

  194. Muvarak, N. E. et al. Enhancing the cytotoxic effects of PARP inhibitors with DNA demethylating agents — a potential therapy for cancer. Cancer Cell 30, 637–650 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  195. Tentori, L. et al. Poly(ADP-ribose) polymerase (PARP) inhibition or PARP-1 gene deletion reduces angiogenesis. Eur. J. Cancer 43, 2124–2133 (2007).

    CAS  PubMed  Google Scholar 

  196. Borst, G. R. et al. Neoadjuvant olaparib targets hypoxia to improve radioresponse in a homologous recombination-proficient breast cancer model. Oncotarget 8, 87638–87646 (2017).

    PubMed  PubMed Central  Google Scholar 

  197. Ali, M. et al. Vasoactivity of AG014699, a clinically active small molecule inhibitor of poly(ADP-ribose) polymerase: a contributory factor to chemopotentiation in vivo? Clin. Cancer Res. 15, 6106–6112 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  198. Bindra, R. S. et al. Down-regulation of Rad51 and decreased homologous recombination in hypoxic cancer cells. Mol. Cell. Biol. 24, 8504–8518 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  199. Dean, E. et al. Phase I study to assess the safety and tolerability of olaparib in combination with bevacizumab in patients with advanced solid tumours. Br. J. Cancer 106, 468–474 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  200. Liu, J. F. et al. A Phase 1 trial of the poly(ADP-ribose) polymerase inhibitor olaparib (AZD2281) in combination with the anti-angiogenic cediranib (AZD2171) in recurrent epithelial ovarian or triple-negative breast cancer. Eur. J. Cancer 49, 2972–2978 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  201. Liu, J. F. et al. Overall survival and updated progression-free survival results from a randomized phase 2 trial comparing the combination of olaparib and cediranib against olaparib alone in recurrent platinum-sensitive ovarian cancer. J. Clin. Oncol. 35, 5535–5535 (2017).

    Google Scholar 

  202. Philip, C.-A. et al. Inhibition of PI3K-AKT-mTOR pathway sensitizes endometrial cancer cell lines to PARP inhibitors. BMC Cancer 17, 638 (2017).

    PubMed  PubMed Central  Google Scholar 

  203. Wang, D. et al. Effective use of PI3K inhibitor BKM120 and PARP inhibitor Olaparib to treat PIK3CA mutant ovarian cancer. Oncotarget 7, 13153–13166 (2016).

    PubMed  PubMed Central  Google Scholar 

  204. Matulonis, U. A. et al. Phase I dose escalation study of the PI3kinase pathway inhibitor BKM120 and the oral poly (ADP ribose) polymerase (PARP) inhibitor olaparib for the treatment of high-grade serous ovarian and breast cancer. Ann. Oncol. 28, 512–518 (2017).

    CAS  PubMed  Google Scholar 

  205. Matulonis, U. A. et al. Phase I of oral BKM120 or BYL719 and olaparib for high-grade serous ovarian cancer or triple-negative breast cancer: final results of the BKM120 plus olaparib cohort [abstract]. Cancer Res. 75, CT324 (2015).

    Google Scholar 

  206. Konstantinopoulos, P. A. et al. Phase I study of the alpha specific PI3-Kinase inhibitor BYL719 and the poly (ADP-Ribose) polymerase (PARP) inhibitor olaparib in recurrent ovarian and breast cancer: analysis of the dose escalation and ovarian cancer expansion cohort [abstract]. Cancer Res. 77, CT008 (2017).

    Google Scholar 

  207. Michalarea, V. et al. Phase I trial combining the PARP inhibitor olaparib (Ola) and AKT inhibitor AZD5363 (AZD) in germline (g)BRCA and non-BRCA mutant (m) advanced cancer patients (pts) incorporating noninvasive monitoring of cancer mutations [abstract]. Cancer Res. 76, CT010 (2016).

    Google Scholar 

  208. Du, Y. et al. Blocking c-Met-mediated PARP1 phosphorylation enhances anti-tumor effects of PARP inhibitors. Nat. Med. 22, 194–201 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  209. Cottini, F. et al. Synthetic lethal approaches exploiting DNA damage in aggressive myeloma. Cancer Discov. 5, 972–987 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  210. Shapiro, G. I. et al. Phase 1 dose-escalation study of the CDK inhibitor dinaciclib in combination with the PARP inhibitor veliparib in patients with advanced solid tumors [abstract]. Cancer Res. 77, CT047 (2016).

    Google Scholar 

  211. Karanika, S. et al. Targeting DNA damage response in prostate cancer by inhibiting androgen receptor-CDC6-ATR-Chk1 signaling. Cell Rep. 18, 1970–1981 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  212. Asim, M. et al. Synthetic lethality between androgen receptor signalling and the PARP pathway in prostate cancer. Nat. Commun. 8, 374 (2017).

    PubMed  PubMed Central  Google Scholar 

  213. Clarke, N. et al. Olaparib combined with abiraterone in patients with metastatic castration-resistant prostate cancer: a randomised, double-blind, placebo-controlled, phase 2 trial. Lancet Oncol. 19, 975–986 (2018).

    CAS  PubMed  Google Scholar 

  214. Le, D. T. et al. PD-1 blockade in tumors with mismatch-repair deficiency. N. Engl. J. Med. 372, 2509–2520 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  215. Le, D. T. et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science 357, 409–413 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  216. Germano, G. et al. Inactivation of DNA repair triggers neoantigen generation and impairs tumour growth. Nature 552, 116–120 (2017).

    CAS  PubMed  Google Scholar 

  217. Brown, J. S., Sundar, R. & Lopez, J. Combining DNA damaging therapeutics with immunotherapy: more haste, less speed. Br. J. Cancer 118, 312–324 (2018).

    CAS  PubMed  Google Scholar 

  218. Härtlova, A. et al. DNA damage primes the type I interferon system via the cytosolic DNA sensor STING to promote anti-microbial innate immunity. Immunity 42, 332–343 (2015).

    PubMed  Google Scholar 

  219. Jiao, S. et al. PARP Inhibitor Upregulates PD-L1 Expression and Enhances Cancer-Associated Immunosuppression. Clin. Cancer Res. 23, 3711–3720 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  220. Sato, H. et al. DNA double-strand break repair pathway regulates PD-L1 expression in cancer cells. Nat. Commun. 8, 1751 (2017).

    PubMed  PubMed Central  Google Scholar 

  221. Turajlic, S. et al. Insertion-and-deletion-derived tumour-specific neoantigens and the immunogenic phenotype: a pan-cancer analysis. Lancet Oncol. 18, 1009–1021 (2017).

    CAS  PubMed  Google Scholar 

  222. McGrail, D. J. et al. Multi-omics analysis reveals neoantigen-independent immune cell infiltration in copy-number driven cancers. Nat. Commun. 9, 1317 (2018).

    PubMed  PubMed Central  Google Scholar 

  223. Lee, J.-M. et al. Safety and clinical activity of the programmed death-ligand 1 inhibitor durvalumab in combination with poly (ADP-ribose) polymerase inhibitor olaparib or vascular endothelial growth factor receptor 1–3 inhibitor cediranib in women’s cancers: a dose-escalation, phase i study. J. Clin. Oncol. 35, 2193–2202 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  224. Domchek, S. et al. An open-label, multitumor, phase II basket study of olaparib and durvalumab (MEDIOLA): results in germline BRCA-mutated (gBRCAm) HER2-negative metastatic breast cancer (MBC) [abstract]. Cancer Res. 78, PD6-11 (2018).

    Google Scholar 

  225. Karzai, F. et al. A phase 2 study of olaparib and durvalumab in metastatic castrate-resistant prostate cancer (mCRPC) in an unselected population. J. Clin. Oncol. 36, 163 (2018).

    Google Scholar 

  226. Powles, T. et al. BISCAY, a phase Ib, biomarker-directed multidrug umbrella study in patients with metastatic bladder cancer. J. Clin. Oncol. 34, TPS4577 (2017).

    Google Scholar 

  227. Dobzhansky, T. Genetics of natural populations; recombination and variability in populations of Drosophila pseudoobscura. Genetics 31, 269–290 (1946).

    CAS  PubMed  PubMed Central  Google Scholar 

  228. Chambon, P., Weill, J. D. & Mandel, P. Nicotinamide mononucleotide activation of new DNA-dependent polyadenylic acid synthesizing nuclear enzyme. Biochem. Biophys. Res. Commun. 11, 39–43 (1963).

    CAS  PubMed  Google Scholar 

  229. Chambon, P., Weill, J. D., Doly, J., Strosser, M. & Mandell, P. On the formation of a novel adelylic compound by enzymatic extracts of liver nuclei. Biochem. Biophys. Res. Commun. 25, 638–643 (1966).

    CAS  Google Scholar 

  230. Yamada, M., Miwa, M. & Sugimura, T. Studies on poly (adenosine diphosphate-ribose). X. Properties of a partially purified poly (adenosine diphosphate-ribose) polymerase. Arch. Biochem. Biophys. 146, 579–586 (1971).

    CAS  PubMed  Google Scholar 

  231. Durkacz, B. W., Omidiji, O., Gray, D. A. & Shall, S. (ADP-ribose)n participates in DNA excision repair. Nature 283, 593 (1980).

    CAS  PubMed  Google Scholar 

  232. Hall, J. M. et al. Linkage of early-onset familial breast cancer to chromosome 17q21. Science 250, 1684–1689 (1990).

    CAS  PubMed  Google Scholar 

  233. Wooster, R. et al. Identification of the breast cancer susceptibility gene BRCA2. Nature 378, 789–792 (1995).

    CAS  PubMed  Google Scholar 

  234. White, A. W. et al. Resistance-modifying agents. 9. Synthesis and biological properties of benzimidazole inhibitors of the DNA repair enzyme poly(ADP-ribose) polymerase. J. Med. Chem. 43, 4084–4097 (2000).

    CAS  PubMed  Google Scholar 

  235. Farmer, H. et al. Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy. Nature 434, 917–921 (2005).

    CAS  PubMed  Google Scholar 

  236. Menear, K. A. et al. 4-[3-(4-cyclopropanecarbonylpiperazine-1-carbonyl)-4-fluorobenzyl]-2H-phthalazin-1-one: a novel bioavailable inhibitor of poly(ADP-ribose) polymerase-1. J. Med. Chem. 51, 6581–6591 (2008).

    CAS  PubMed  Google Scholar 

  237. Plummer, R. et al. Phase I study of the poly(ADP-ribose) polymerase inhibitor, AG014699, in combination with temozolomide in patients with advanced solid tumors. Clin. Cancer Res. 14, 7917–7923 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  238. Jones, P. et al. Discovery of 2-{4-[(3S)-piperidin-3-yl]phenyl}-2H-indazole-7-carboxamide (MK-4827): a novel oral poly(ADP-ribose)polymerase (PARP) inhibitor efficacious in BRCA-1 and -2 mutant tumors. J. Med. Chem. 52, 7170–7185 (2009).

    CAS  PubMed  Google Scholar 

  239. Sanofi-aventis. Sanofi-aventis reports top-line results from phase III study with iniparib (BSI-201) in metastatic triple-negative breast cancer. Sanofi http://www.news.sanofi.us/press-releases?item=118547 (2011).

  240. Patel, A. G., De Lorenzo, S. B., Flatten, K. S., Poirier, G. G. & Kaufmann, S. H. Failure of iniparib to inhibit poly(ADP-Ribose) polymerase in vitro. Clin. Cancer Res. 18, 1655–1662 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  241. Supreme Court of the United States. Association for Molecular Pathology et al. petitioners v. Myriad Genetics, INC. et al. Supreme Court of the United States https://www.supremecourt.gov/opinions/12pdf/12-398_1b7d.pdf (2013).

Download references

Acknowledgements

The University of Texas MD Anderson Cancer Center (Houston, TX, USA) is supported by the NIH Cancer Center Support Grant CA016672.

Reviewer information

Nature Reviews Clinical Oncology thanks T. Laetsch and the other anonymous reviewer(s) for their contribution to the peer review of this work.

Author information

Authors and Affiliations

Authors

Contributions

P.G.P., C.T. and T.A.Y. made substantial contributions to researching data for this article and discussions of content. All authors contributed to writing the article and reviewing/editing of the manuscript before submission.

Corresponding author

Correspondence to Timothy A. Yap.

Ethics declarations

Competing interests

G.B.M. has received research support from AstraZeneca, Pfizer and Tesaro, and has been a consultant for AstraZeneca, Pfizer and Tesaro. T.A.Y. has received research support from AstraZeneca, Bayer, Pfizer, Tesaro and Vertex Pharmaceuticals; has served on advisory boards of Aduro, Almac, AstraZeneca, Atrin, Bristol-Myers Squibb, Clovis, EMD Serono, Ignyta, Jansen, Merck, Pfizer, Roche, Seattle Genetics and Vertex Pharmaceuticals; and has received speaker bureau from AstraZeneca, Merck and Tesaro. The other authors declare no competing interests.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Related links

ClinicalTrials.gov database: https://clinicaltrials.gov/ct2/home

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Pilié, P.G., Tang, C., Mills, G.B. et al. State-of-the-art strategies for targeting the DNA damage response in cancer. Nat Rev Clin Oncol 16, 81–104 (2019). https://doi.org/10.1038/s41571-018-0114-z

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41571-018-0114-z

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing