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:

DNA damage response inhibitors in cancer therapy: lessons from the past, current status and future implications

Abstract

The DNA damage response (DDR) is a network of proteins that coordinate DNA repair and cell-cycle checkpoints to prevent damage being transmitted to daughter cells. DDR defects lead to genomic instability, which enables tumour development, but they also create vulnerabilities that can be used for cancer therapy. Historically, this vulnerability has been taken advantage of using DNA-damaging cytotoxic drugs and radiotherapy, which are more toxic to tumour cells than to normal tissues. However, the discovery of the unique sensitivity of tumours defective in the homologous recombination DNA repair pathway to PARP inhibition led to the approval of six PARP inhibitors worldwide and to a focus on making use of DDR defects through the development of other DDR-_targeting drugs. Here, we analyse the lessons learnt from PARP inhibitor development and how these may be applied to new _targets to maximize success. We explore why, despite so much research, no other DDR inhibitor class has been approved, and only a handful have advanced to later-stage clinical trials. We discuss why more reliable predictive biomarkers are needed, explore study design from past and current trials, and suggest alternative models for monotherapy and combination studies. _targeting multiple DDR pathways simultaneously and potential combinations with anti-angiogenic agents or immune checkpoint inhibitors are also discussed.

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

Access options

Buy this article

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

Fig. 1: _targeting the DDR response.
Fig. 2: Timeline of PARP inhibitor development and approvals.
Fig. 3: Comprehensive DDR inhibitor landscape per _target, development stage (preclinical or clinical) and status (active, not active or discontinued).

Similar content being viewed by others

References

  1. Hanahan, D. & Weinberg, R. A. Hallmarks of cancer: the next generation. Cell 144, 646–674 (2011). This key review updates Hanahan and Wieinberg’s original landmark paper of 2000 with additional focus on genetic instability as an enabling characteristic and drugs _targeting the hallmarks of cancer.

    PubMed  Google Scholar 

  2. Bartkova, J. et al. DNA damage response as a candidate anti-cancer barrier in early human tumorigenesis. Nature 434, 864–870 (2005).

    PubMed  Google Scholar 

  3. Bieging, K. T., Mello, S. S. & Attardi, L. D. Unravelling mechanisms of p53-mediated tumour suppression. Nat. Rev. Cancer 14, 359–370 (2014).

    PubMed  PubMed Central  Google Scholar 

  4. Boulton, S. J. Cellular functions of the BRCA tumour-suppressor proteins. Biochem. Soc. Trans. 34, 633–645 (2006).

    PubMed  Google Scholar 

  5. Sarni, D. & Kerem, B. Oncogene-induced replication stress drives genome instability and tumorigenesis. Int. J. Mol. Sci. 18, 1339 (2017).

    PubMed Central  Google Scholar 

  6. Jackson, S. P. & Bartek, J. The DNA-damage response in human biology and disease. Nature 22, 1071–1078 (2009). This is an excellent review of the DDR and its implications for human health that remains relevant today.

    Google Scholar 

  7. Groelly, F. J., Fawkes, M., Dagg, R. A., Blackford, A. N. & Tarsounas, M. _targeting DNA damage response pathways in cancer. Nat. Rev. Cancer 23, 78–94 (2023). This is an up-to-date review of the anticancer potential of _targeting the DDR.

    PubMed  Google Scholar 

  8. Colvin, M. & Chabner, B.A. in Cancer Chemotherapy: Principles and Practice (eds Chabner, B. A. & Collins, J. M.) 276–313 (Lippincott, 1990).

  9. Pegg, A. E. Mammalian O6-alkylguanine-DNA alkyltransferase: regulation and importance in response to alkylating carcinogenic and therapeutic agents. Cancer Res. 50, 6119–6129 (1990).

    PubMed  Google Scholar 

  10. Rabik, C. A., Njoku, M. C. & Dolan, M. E. Inactivation of O6-alkylguanine DNA alkyltransferase as a means to enhance chemotherapy. Cancer Treat. Rev. 32, 261–276 (2006).

    PubMed  Google Scholar 

  11. 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).

    PubMed  Google Scholar 

  12. Tawbi, H. A. et al. Inhibition of DNA repair with MGMT pseudosubstrates: phase I study of lomeguatrib in combination with dacarbazine in patients with advanced melanoma and other solid tumours. Br. J. Cancer 105, 773–777 (2011).

    PubMed  PubMed Central  Google Scholar 

  13. Yu, W., Zhang, L., Wei, Q. & Shao, A. O6-methylguanine-DNA methyltransferase (MGMT): challenges and new opportunities in glioma chemotherapy. Front. Oncol. 17, 1547 (2020).

    Google Scholar 

  14. Butler, M. et al. MGMT status as a clinical biomarker in glioblastoma. Trends Cancer 6, 380–391 (2020).

    PubMed  PubMed Central  Google Scholar 

  15. Purnell, M. R. & Whish, W. J. Novel inhibitors of poly(ADP-ribose) synthetase. Biochem. J. 185, 775–777 (1980).

    PubMed  PubMed Central  Google Scholar 

  16. Durkacz, B. W., Omidiji, O., Gray, D. A. & Shall, S. (ADP-ribose)n participates in DNA excision repair. Nature 283, 593–596 (1980). This study and that by Purnell & Whish (1980) show for the first time how the first PARPi, 3-aminobenzamide, potentiates the cytotoxicity of DNA methylating agents, suggesting its anticancer therapy potential.

    PubMed  Google Scholar 

  17. Ben-Hur, E. Involvement of poly(ADP-ribose) in the radiation response of mammalian cells. Int. J. Radiat. Biol. 46, 659–671 (1984).

    Google Scholar 

  18. Banasik, M., Komura, H., Shimoyama, M. & Ueda, K. Specific inhibitors of poly(ADP-ribose) synthetase and mono(ADP-ribosyl)transferase. J. Biol. Chem. 267, 1569–1575 (1992). This article describes the identification, through the classic ‘analogue by catalogue’ approach, of many potent PARPis, some of which have been further elaborated to give us some of today’s clinical drugs.

    PubMed  Google Scholar 

  19. Arundel-Suto, C. M., Scavone, S. V., Turner, W. R., Suto, M. J. & Sebolt-Leopold, J. S. Effects of PD128763, a new potent inhibitor of poly(ADP-ribose) polymerase, on X-ray induced cellular recovery processes in Chinese hamster V79 cells. Radiat. Res. 126, 367–371 (1991).

    PubMed  Google Scholar 

  20. Griffin, R. J. et al. Novel potent inhibitors of the DNA repair enzyme poly(ADP-ribose)polymerase (PARP). Anticancer Drug. Des. 10, 507–514 (1995).

    PubMed  Google Scholar 

  21. Bowman, K. J., Newell, D. R., Calvert, A. H. & Curtin, N. J. Differential effects of the poly (ADP-ribose) polymerase (PARP) inhibitor NU1025 on topoisomerase I and II inhibitor cytotoxicity in L1210 cells in vitro. Br. J. Cancer 84, 106–112 (2001).

    PubMed  PubMed Central  Google Scholar 

  22. Smith, L. M., Willmore, E., Austin, C. A. & Curtin, N. J. The novel poly(ADP-Ribose) polymerase inhibitor, AG14361, sensitizes cells to topoisomerase I poisons by increasing the persistence of DNA strand breaks. Clin. Cancer Res. 11, 8449–8457 (2005).

    PubMed  Google Scholar 

  23. Wang, Z. Q. et al. Mice lacking ADPRT and poly(ADP-ribosyl)ation develop normally but are susceptible to skin disease. Genes Dev. 9, 509–520 (1995).

    PubMed  Google Scholar 

  24. De Murcia, J. M. et al. Requirement of poly(ADP-ribose) polymerase in recovery from DNA damage in mice and in cells. Proc. Natl Acad. Sci. USA 94, 7303–7307 (1997). This reference and Wang et al. (1995) are the first reports to describe PARP1-knockout mice.

    PubMed  PubMed Central  Google Scholar 

  25. Ménissier de Murcia, J. et al. Functional interaction between PARP-1 and PARP-2 in chromosome stability and embryonic development in mouse. EMBO J. 22, 2255–2263 (2003).

    PubMed  PubMed Central  Google Scholar 

  26. Satoh, M. S. & Lindahl, T. Role of poly(ADP-ribose) formation in DNA repair. Nature 356, 356–358 (1992). This paper explores the role of PARP in DNA SSBR and is the first to describe the phenomenon in which inactive PARP (in the absence of substrate NAD+ or in the presence of an inhibitor) has a much greater impact on the persistence of DNA damage than the depletion of PARP. The authors propose that PARP binding to the DNA break obstructs the repair machinery, unless it can auto-polyADP-ribosylate and dissociate from the break.

    PubMed  Google Scholar 

  27. Murai, J. et al. Trapping of PARP1 and PARP2 by clinical PARP inhibitors. Cancer Res. 72, 5588–5599 (2012). This article is one of the most recent studies determining that the trapping potency — rather than catalytic inhibition potency — of clinically active PARPis correlates with their cytotoxicity.

    PubMed  PubMed Central  Google Scholar 

  28. Ferraris, D. V. Evolution of poly(ADP-ribose) polymerase-1 (PARP-1) inhibitors. From concept to clinic. J. Med. Chem. 53, 4561–4584 (2010).

    PubMed  Google Scholar 

  29. Thomas, H. D. et al. Preclinical selection of a novel poly(ADP-ribose) polymerase inhibitor for clinical trial. Mol. Cancer Ther. 6, 945–956 (2007).

    PubMed  Google Scholar 

  30. 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). This paper describes the first clinical trial with a PARPi that included pharmacodynamic as well as pharmacokinetic monitoring to achieve the levels of PARP inhibition predicted to be active from in vivo preclinical studies.

    PubMed  PubMed Central  Google Scholar 

  31. Lu, Y., Liu, Y., Pang, Y., Pacak, K. & Yang, C. Double-barreled gun: combination of PARP inhibitor with conventional chemotherapy. Pharmacol. Ther. 188, 168–175 (2018).

    PubMed  PubMed Central  Google Scholar 

  32. Curtin, N. J. & Szabo, C. Poly(ADP-ribose) polymerase inhibition: past, present and future. Nat. Rev. Drug Discov. 19, 711–736 (2020).

    PubMed  Google Scholar 

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

    PubMed  Google Scholar 

  34. Farmer, H. et al. _targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy. Nature 434, 917–921 (2005). This paper and Bryant et al. (2005) are the first to describe the synthetic lethality of PARPi in BRCA-mutant cells and tumours.

    PubMed  Google Scholar 

  35. 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). This paper describes the results of the first clinical trial of a PARPi in cancer patients with a BRCA mutation, which validates the preclinical data in the clinical setting.

    PubMed  Google Scholar 

  36. Lindahl, T. Instability and decay of the primary structure of DNA. Nature 362, 709–715 (1993).

    PubMed  Google Scholar 

  37. O’Shaughnessy, J. et al. Iniparib plus chemotherapy in metastatic triple-negative breast cancer. N. Engl. J. Med. 364, 205–214 (2011).

    PubMed  Google Scholar 

  38. O’Shaughnessy, J. et al. Phase III study of iniparib plus gemcitabine and carboplatin versus gemcitabine and carboplatin in patients with metastatic triple-negative breast cancer. J. Clin. Oncol. 32, 3840–3847 (2014).

    PubMed  Google Scholar 

  39. Liu, X. et al. Iniparib nonselectively modifies cysteine-containing proteins in tumor cells and is not a bona fide PARP inhibitor. Clin. Cancer Res. 18, 510–523 (2012).

    PubMed  Google Scholar 

  40. Curtin, N. PARP inhibitors for anticancer therapy. Biochem. Soc. Trans. 42, 82–88 (2014).

    PubMed  Google Scholar 

  41. Ihnen, M. et al. Therapeutic potential of the poly(ADP-ribose) Polymerase inhibitor rucaparib for the treatment of sporadic human ovarian cancer. Mol. Cancer Ther. 12, 1002–1015 (2013).

    PubMed  PubMed Central  Google Scholar 

  42. Evers, B. et al. Selective inhibition of BRCA2-deficient mammary tumor cell growth by AZD2281 and cisplatin. Clin. Cancer Res. 14, 3916–3925 (2008).

    PubMed  Google Scholar 

  43. Murai, J. et al. Rationale for poly(ADP-ribose)polymerase (PARP) inhibitors in combination therapy with camptothecins or temozolomide based on PARP trapping versus catalytic inhibition. J. Pharmacol. Exp. Ther. 349, 408–416 (2014).

    PubMed  PubMed Central  Google Scholar 

  44. Wilson, R. H. et al. A phase I study of intravenous and oral rucaparib in combination with chemotherapy in patients with advanced solid tumours. Br. J. Cancer 116, 884–892 (2017).

    PubMed  PubMed Central  Google Scholar 

  45. Kristeleit, R. et al. A phase I–II study of the oral PARP inhibitor rucaparib in patients with germline BRCA1/2-mutated ovarian carcinoma or other solid tumors. Clin. Cancer Res. 23, 4095–4106 (2017).

    PubMed  Google Scholar 

  46. Lee, J. M., Peer, C. J. & Yu, M. Sequence-specific pharmacokinetic and pharmacodynamic phase I/Ib study of olaparib tablets and carboplatin in women’s cancer. Clin. Cancer Res. 23, 1397–1406 (2017).

    PubMed  Google Scholar 

  47. Goldenberg, S. G., Cardillo, T. M., Govindan, S. V., Rossi, E. A. & Sharkey, R. M. Trop-2 is a novel _target for solid cancer therapy with sacituzumab govitecan (IMMU-132), an antibody-drug conjugate (ADC). Onco_target 6, 22496–22512 (2015).

    PubMed  PubMed Central  Google Scholar 

  48. Yap, T. A. et al. Phase Ib SEASTAR study: combining rucaparib and sacituzumab govitecan in patients with cancer with or without mutations in homologous recombination repair genes. JCO Precis. Oncol. 6, e2100456 (2022).

    PubMed  PubMed Central  Google Scholar 

  49. Aldivar, J. C., Cortez, D. & Cimprich, K. A. The essential kinase ATR: ensuring faithful duplication of a challenging genome. Nat. Rev. Mol. Cell Biol. 18, 622–636 (2017).

    Google Scholar 

  50. Kim, H. et al. Combining PARP with ATR inhibition overcomes PARP inhibitor and platinum resistance in ovarian cancer models. Nat. Commun. 11, 3726 (2020). This paper describes one of the first known mechanisms of PARPi resistance and how to overcome it in preclinical ovarian cancer models, leading to a new generation of clinical trials of PARPi combinations.

    PubMed  PubMed Central  Google Scholar 

  51. Wethington, S. L. et al. Combination ATR (ceralasertib) and PARP (olaparib) Inhibitor (CAPRI) trial in acquired PARP inhibitor-resistant homologous recombination-deficient ovarian cancer. Clin. Cancer Res. 29, 2800–2807 (2023).

    PubMed  Google Scholar 

  52. Yap, T. A. et al. STAR study NCI10329: Phase Ib Sequential Trial of Agents against DNA Repair (STAR) study to investigate the sequential combination of the poly (ADP-Ribose) polymerase inhibitor (PARPi) olaparib (ola) and WEE1 inhibitor (WEE1i) adavosertib (ada) in patients (pts) with DNA damage response (DDR)-aberrant advanced tumors, enriched for BRCA1/2 mutated and CCNE1 amplified cancers. Eur. J. Cancer 174 (Supplement 1), S7 (2022).

    Google Scholar 

  53. Hockings, H. & Miller, R. E. The role of PARP inhibitor combination therapy in ovarian cancer. Ther. Adv. Med. Oncol. 15, 17588359231173183 (2023).

    PubMed  PubMed Central  Google Scholar 

  54. Jiao, S. et al. PARP inhibitor upregulates PD-L1 expression and enhances cancer-associated immunosuppression. Clin. Cancer Res. 23, 3711–3720 (2017).

    PubMed  PubMed Central  Google Scholar 

  55. Higuchi, T. et al. CTLA-4 blockade synergizes therapeutically with PARP inhibition in BRCA1-deficient ovarian cancer. Cancer Immunol. Res. 3, 1257–1268 (2015).

    PubMed  PubMed Central  Google Scholar 

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

    PubMed  Google Scholar 

  57. Lim, J. J., Yang, K., Taylor-Harding, B., Ruprecht Wiedemeyer, W. & Buckanovich, R. J. VEGFR3 inhibition chemosensitizes ovarian cancer stem-like cells through down-regulation of BRCA1 and BRCA2. Neoplasia 16, 343–353.e2 (2014).

    PubMed  PubMed Central  Google Scholar 

  58. Hegan, D. C. et al. Inhibition of poly(ADP-ribose) polymerase down-regulates BRCA1 and RAD51 in a pathway mediated by E2F4 and p130. Proc. Natl Acad. Sci. USA 107, 2201–2206 (2010).

    PubMed  PubMed Central  Google Scholar 

  59. Kudo, M. Scientific rationale for combined immunotherapy with PD-1/PD-L1 antibodies and VEGF inhibitors in advanced hepatocellular carcinoma. Cancers 12, 1089 (2020).

    PubMed  PubMed Central  Google Scholar 

  60. Ray-Coquard, I. et al. Olaparib plus bevacizumab as first-line maintenance in ovarian cancer. PAOLA-1 Protocol. N. Engl. J. Med. 381, 2416–2428 (2019).

    PubMed  Google Scholar 

  61. Drew, Y. et al. Olaparib plus durvalumab, with or without bevacizumab, as treatment in PARP inhibitor-naïve platinum-sensitive relapsed ovarian cancer: a phase II multi-cohort study. Clin. Cancer Res. 30, 50–62 (2024).

    PubMed  Google Scholar 

  62. Harter, P. et al. Durvalumab with paclitaxel/carboplatin (PC) and bevacizumab (bev), followed by maintenance durvalumab, bev, and olaparib in patients (pts) with newly diagnosed advanced ovarian cancer (AOC) without a tumor BRCA1/2 mutation (non-tBRCAm): results from the randomized, placebo (pbo)-controlled phase III DUO-O trial. J. Clin. Oncol. 41, LBA5506 (2023).

    Google Scholar 

  63. Konstantinopoulos, P. A. et al. Single-arm phases 1 and 2 trial of niraparib in combination with pembrolizumab in patients with recurrent platinum-resistant ovarian carcinoma. JAMA Oncol. 5, 1141–1149 (2019).

    PubMed  PubMed Central  Google Scholar 

  64. Liu, J. et al. An open-label phase II study of dostarlimab (TSR-042), bevacizumab (bev), and niraparib combination in patients (pts) with platinum-resistant ovarian cancer (PROC): cohort A of the OPAL trial. Gynecol. Oncol. 162, S17–S18 (2021).

    Google Scholar 

  65. Clarke, N. W. et al. Abiraterone and olaparib for metastatic castration-resistant prostate cancer. NEJM Evid. 1, EVIDoa2200043 (2022).

    PubMed  Google Scholar 

  66. Agarwal, N. et al. Talazoparib plus enzalutamide in men with first-line metastatic castration-resistant prostate cancer (TALAPRO-2): a randomised, placebo-controlled, phase 3 trial. Lancet 402, 291–303 (2023).

    PubMed  Google Scholar 

  67. Illuzzi, G. et al. Preclinical characterization of AZD5305, a next-generation, highly selective PARP1 inhibitor and trapper. Clin. Cancer Res. 28, 4724–4736 (2022). This article reports the characterization of AZD5305, a highly selective PARP1 inhibitor and trapper, indicating its potential as a next-generation PARPi therapy with improved selectivity and efficacy for treating cancers with deficiencies in homologous recombination repair. AZD5305 selectively inhibits PARP1 over PARP2, which is suggested to result in reduced off-_target effects and enhanced therapeutic benefit.

    PubMed  PubMed Central  Google Scholar 

  68. Yap, T. A. et al. PETRA: First-in-human Phase 1/2a trial of the first-in-class new generation poly(ADP-ribose) polymerase-1 selective inhibitor (PARP1i) saruparib (AZD5305) in patients with advanced solid tumors with BRCA1/2, PALB2 or RAD51C/D mutations. In Proc. 115th Annual Meeting of the American Association for Cancer Research (AACR, 2024); https://doi.org/10.1158/1538-7445.AM2022-CT007.

  69. Dungey, F. A., Löser, D. A. & Chalmers, A. J. Replication-dependent radiosensitization of human glioma cells by inhibition of poly (ADP-ribose) polymerase: mechanisms and therapeutic potential. Int. J. Radiat. Oncol. Biol. Phys. 72, 1188–1197 (2008).

    PubMed  Google Scholar 

  70. Noël, G. et al. Radiosensitization by the poly (ADP-ribose) polymerase inhibitor 4-amino-1, 8-naphthalimide is specific of the S phase of the cell cycle and involves arrest of DNA synthesis. Mol. Cancer Ther. 5, 564–574 (2006).

    PubMed  Google Scholar 

  71. Chalmers, A., Johnston, P., Woodcock, M., Joiner, M. & Marples, B. PARP-1, PARP-2, and the cellular response to low doses of ionizing radiation. Int. J. Radiat. Oncol. Biol. Phys. 58, 410–419 (2004).

    PubMed  Google Scholar 

  72. McLaughlin, M. et al. Inflammatory microenvironment remodelling by tumour cells after radiotherapy. Nat. Rev. Cancer 20, 203–217 (2020).

    PubMed  Google Scholar 

  73. Pilger, D. et al. Interfaces between cellular responses to DNA damage and cancer immunotherapy. Genes Dev. 35, 602–618 (2021).

    PubMed  PubMed Central  Google Scholar 

  74. Barcellini, A. et al. PARP inhibitors in combination with radiotherapy: to do or not to do? Cancers 13, 5380 (2021). An excellent review of the challenges of combining PARPis with radiation and summary of recent studies.

    PubMed  PubMed Central  Google Scholar 

  75. Czito, B. G. et al. Safety and tolerability of veliparib combined with capecitabine plus radiotherapy in patients with locally advanced rectal cancer: a phase 1b study. Lancet Gastroenterol. Hepatol. 2, 418–426 (2017).

    PubMed  Google Scholar 

  76. 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 cancer. Gynecol. Oncol. 144, 486–490 (2017).

    PubMed  PubMed Central  Google Scholar 

  77. Jagsi, R. et al. Concurrent veliparib with chest wall and nodal radiotherapy in patients with inflammatory or locoregionally recurrent breast cancer: the TBCRC 024 phase I multicenter study. J. Clin. Oncol. 36, 1317–1322 (2018).

    PubMed  PubMed Central  Google Scholar 

  78. Karam, S. D. et al. Final report of a phase I trial of olaparib with cetuximab and radiation for heavy smoker patients with locally advanced head and neck cancer. Clin. Cancer Res. 24, 4949–4959 (2018).

    PubMed  PubMed Central  Google Scholar 

  79. Tuli, R. et al. A phase 1 study of veliparib, a PARP-1/2 inhibitor, with gemcitabine and radiotherapy in locally advanced pancreatic cancer. eBioMedicine 40, 375–381 (2019).

    PubMed  PubMed Central  Google Scholar 

  80. Cheung, Y. K. & Chappell, R. Sequential designs for phase I clinical trials with late onset toxicities. Biometrics 56, 1177–1182 (2000).

    PubMed  Google Scholar 

  81. Verhagen, C. V. et al. Extent of radiosensitization by the PARP inhibitor olaparib depends on its dose, the radiation dose and the integrity of the homologous recombination pathway of tumor cells. Radiother. Oncol. 116, 358–65 (2015).

    PubMed  Google Scholar 

  82. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/study/NCT04550104 (2024).

  83. Moore, K. et al. Maintenance olaparib in patients with newly diagnosed advanced ovarian cancer. N. Engl. J. Med. 379, 2495–2505 (2018).

    PubMed  Google Scholar 

  84. González-Martín, A. et al. Niraparib in patients with newly diagnosed advanced ovarian cancer. N. Engl. J. Med. 381, 2391–2402 (2019).

    PubMed  Google Scholar 

  85. Tew, W. P., Lacchetti, C., Kohn, E. C. & PARP Inhibitors in the Management of Ovarian Cancer Guideline Expert Panel, Poly(ADP-ribose) polymerase inhibitors in the management of ovarian cancer: ASCO guideline rapid recommendation update. J. Clin. Oncol. 40, 3878–3881 (2022).

    PubMed  Google Scholar 

  86. Mirza, M. R. et al. #161 Final overall survival and long-term safety in the ENGOT-OV16/NOVA phase 3 trial of niraparib in patients with recurrent ovarian cancer. Int. J. Gynecol. Cancer 33, A15–A16 (2023).

    Google Scholar 

  87. Oza, A. et al. 518O Overall survival results from ARIEL4: a phase III study assessing rucaparib vs chemotherapy in patients with advanced, relapsed ovarian carcinoma and a deleterious BRCA1/2 mutation. Ann. Oncol. 33, S780 (2022).

    Google Scholar 

  88. Drew, Y. et al. Phase 2 multicentre trial investigating intermittent and continuous dosing schedules of the poly(ADP-ribose) polymerase inhibitor rucaparib in germline BRCA mutation carriers with advanced ovarian and breast cancer. Br. J. Cancer 114, 723–730 (2016).

    PubMed  PubMed Central  Google Scholar 

  89. Mateo, J. et al. Olaparib in patients with metastatic castration-resistant prostate cancer with DNA repair gene aberrations (TOPARP-B): a multicentre, open-label, randomised, phase 2 trial. Lancet Oncol. 21, 162–174 (2020).

    PubMed  PubMed Central  Google Scholar 

  90. Swisher, E. M. et al. Molecular and clinical determinants of response and resistance to rucaparib for recurrent ovarian cancer treatment in ARIEL2 (Parts 1 and 2). Nat. Commun. 12, 2487 (2021).

    PubMed  PubMed Central  Google Scholar 

  91. Korsholm, L. M. et al. Combining homologous recombination-deficient testing and functional RAD51 analysis enhances the prediction of poly(ADP-ribose) polymerase inhibitor sensitivity. JCO Precis. Oncol. 8, e2300483 (2024).

    PubMed  PubMed Central  Google Scholar 

  92. Zimmermann, M. et al. CRISPR screens identify genomic ribonucleotides as a source of PARP-trapping lesions. Nature 559, 285–289 (2018).

    PubMed  PubMed Central  Google Scholar 

  93. Jones, S. et al. Frequent mutations of chromatin remodeling gene ARID1A in ovarian clear cell carcinoma. Science 330, 228–231 (2010).

    PubMed  PubMed Central  Google Scholar 

  94. Yakovlev, V. A., Sullivan, S. A., Fields, E. C. & Temkin, S. M. PARP inhibitors in the treatment of ARID1A mutant ovarian clear cell cancer: PI3K/Akt1-dependent mechanism of synthetic lethality. Front. Oncol. 13, 1124147 (2023).

    PubMed  PubMed Central  Google Scholar 

  95. Araujo, D. et al. Oncology phase I trial design and conduct: time for a change - MDICT Guidelines 2022. Ann. Oncol. 34, 48–60 (2023).

    PubMed  Google Scholar 

  96. Project Optimus — reforming the dose optimization and dose selection paradigm in oncology US Food & Drug Administration https://www.fda.gov/about-fda/oncology-center-excellence/project-optimus (accessed Jun. 06, 2022).

  97. Zhu, H. et al. _targeting p53–MDM2 interaction by small-molecule inhibitors: learning from MDM2 inhibitors in clinical trials. J. Hematol. Oncol. 15, 91 (2022).

    PubMed  PubMed Central  Google Scholar 

  98. Khurana, A. & Shafer, D. A. MDM2 antagonists as a novel treatment option for acute myeloid leukemia: perspectives on the therapeutic potential of idasanutlin (RG7388). Onco_targets Ther. 12, 2903–2910 (2019).

    Google Scholar 

  99. Moore, A. S., Blagg, J., Linardopoulos, S. & Pearson, A. D. J. Aurora kinase inhibitors: novel small molecules with promising activity in acute myeloid and Philadelphia-positive leukemias. Leukemia 24, 671–678 (2010).

    PubMed  Google Scholar 

  100. Ren, N. et al. Efficacy and safety of PARP inhibitor combination therapy in recurrent ovarian cancer: a systematic review and meta-analysis. Front. Oncol. 11, 638295 (2021).

    PubMed  PubMed Central  Google Scholar 

  101. Banerji, U. & Workman, P. Critical parameters in _targeted drug development: the pharmacological audit trail. Semin. Oncol. 4, 436–445 (2016).

    Google Scholar 

  102. da Costa, A. A. B. A., Chowdhury, D., Shapiro, G. I., D’Andrea, A. D. & Konstantinopoulos, P. A. _targeting replication stress in cancer therapy. Nat. Rev. Drug Discov. 22, 38–58 (2023).

    PubMed  Google Scholar 

  103. Brown, E. J. & Baltimore, D. ATR disruption leads to chromosomal fragmentation and early embryonic lethality. Genes Dev. 4, 397–402 (2000).

    Google Scholar 

  104. Yap, T. A. et al. Phase I trial of first-in-class ATR inhibitor M6620 (VX-970) as monotherapy or in combination with carboplatin in patients with advanced solid tumors. J. Clin. Oncol. 38, 3195–3204 (2020). This article reports the results from the phase I trial of the ATR inhibitor M6620 (VX-970), assessing its safety, tolerability and preliminary efficacy as monotherapy and in combination with carboplatin in patients with advanced-stage solid tumours. The study finds that M6620 is generally well tolerated and shows potential antitumour activity when combined with carboplatin.

    PubMed  PubMed Central  Google Scholar 

  105. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/study/NCT05450692 (2024).

  106. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/study/NCT03334617 (2024).

  107. Besse, B. et al. Biomarker-directed _targeted therapy plus durvalumab in advanced non-small-cell lung cancer: a phase 2 umbrella trial. Nat. Med. 30, 716–729 (2024). This article reports how the ATR inhibitor ceralasertib, when used in combination with durvalumab, can provide clinical benefit and has a manageable safety profile.

    PubMed  PubMed Central  Google Scholar 

  108. Dillon, M. T. et al. ATR inhibition potentiates the radiation-induced inflammatory tumor microenvironment. Clin. Cancer Res. 25, 3392–3403 (2019).

    PubMed  PubMed Central  Google Scholar 

  109. Jette, N. R. et al. Combined poly-ADP ribose polymerase and ataxia-telangiectasia mutated/Rad3-related inhibition _targets ataxia-telangiectasia mutated-deficient lung cancer cells. Br. J. Cancer 121, 600–610 (2019).

    PubMed  PubMed Central  Google Scholar 

  110. Lloyd, R. L. et al. Combined PARP and ATR inhibition potentiates genome instability and cell death in ATM-deficient cancer cells. Oncogene 39, 4869–4883 (2020).

    PubMed  PubMed Central  Google Scholar 

  111. Zimmermann, M. et al. Guiding ATR and PARP inhibitor combinations with chemogenomic screens. Cell Rep. 40, 111081 (2022).

    PubMed  Google Scholar 

  112. Ruiz, S. et al. A genome-wide CRISPR screen identifies CDC25A as a determinant of sensitivity to ATR inhibitors. Mol. Cell 62, 307–313 (2016).

    PubMed  PubMed Central  Google Scholar 

  113. Hustedt, N. et al. A consensus set of genetic vulnerabilities to ATR inhibition. Open Biol. 9, 190156 (2019).

    PubMed  PubMed Central  Google Scholar 

  114. Schleicher, E. M. et al. Dual genome-wide CRISPR knockout and CRISPR activation screens identify mechanisms that regulate the resistance to multiple ATR inhibitors. PLoS Genet. 16, e1009176 (2020).

    PubMed  PubMed Central  Google Scholar 

  115. Lloyd, R. L. et al. Loss of Cyclin C or CDK8 provides ATR inhibitor resistance by suppressing transcription-associated replication stress. Nucleic Acids Res. 49, 8665–8683 (2021).

    PubMed  PubMed Central  Google Scholar 

  116. Llorca-Cardenosa, M. J. et al. SMG8/SMG9 heterodimer loss modulates SMG1 kinase to drive ATR inhibitor resistance. Cancer Res. 82, 3962–3973 (2022).

    PubMed  PubMed Central  Google Scholar 

  117. O’Leary, P. C. et al. Resistance to ATR inhibitors is mediated by loss of the nonsense-mediated decay factor UPF2. Cancer Res. 82, 3950–3961 (2022).

    PubMed  PubMed Central  Google Scholar 

  118. Bradbury, A., Zenke, F. T., Curtin, N. J. & Drew, Y. The role of ATR inhibitors in ovarian cancer: investigating predictive biomarkers of response. Cells 11, 2361 (2022).

    PubMed  PubMed Central  Google Scholar 

  119. Yap, T. A. et al. First-in-human trial of the oral ataxia telangiectasia and RAD3-related (ATR) inhibitor BAY 1895344 in patients with advanced solid tumors. Cancer Discov. 11, 80–91 (2021).

    PubMed  Google Scholar 

  120. Aggarwal, R. et al. 512O Interim results from a phase II study of the ATR inhibitor ceralasertib in ARID1A-deficient and ARID1A-intact advanced solid tumor malignancies. Ann. Oncol. 32, S583 (2021).

    Google Scholar 

  121. Konstantinopoulos, P. A. et al. A Replication stress biomarker is associated with response to gemcitabine versus combined gemcitabine and ATR inhibitor therapy in ovarian cancer. Nat. Commun. 12, 5574 (2021).

    PubMed  PubMed Central  Google Scholar 

  122. Yap, T. A. et al. Camonsertib in DNA damage response-deficient advanced solid tumors: phase 1 trial results. Nat. Med. 29, 1400–1411 (2023).

    PubMed  PubMed Central  Google Scholar 

  123. Matheson, C. J., Backos, D. S. & Reigan, P. _targeting WEE1 kinase in cancer. Trends Pharmacol. Sci. 37, 872–881 (2016).

    PubMed  Google Scholar 

  124. Bukhari, A. B., Chan, G. K. & Gamper, A. M. _targeting the DNA damage response for cancer therapy by inhibiting the kinase Wee1. Front. Oncol. 12, 828684 (2022).

    PubMed  PubMed Central  Google Scholar 

  125. Tominaga, Y., Li, C., Wang, R. H. & Deng, C. X. Murine Wee1 plays a critical role in cell cycle regulation and pre-implantation stages of embryonic development. Int. J. Biol. Sci. 2, 161–170 (2006).

    PubMed  PubMed Central  Google Scholar 

  126. Kong, A. & Mehanna, H. WEE1 inhibitor: clinical development. Curr. Oncol. Rep. 23, 107 (2021).

    PubMed  PubMed Central  Google Scholar 

  127. Yang, X. et al. Development of cell permeable NanoBRET probes for the measurement of PLK1 _target engagement in live cells. Molecules 28, 2950 (2023).

    PubMed  PubMed Central  Google Scholar 

  128. Guler, S. et al. Selective Wee1 inhibitors led to antitumor activity in vitro and correlated with myelosuppression. ACS Med. Chem. Lett. 14, 566–576 (2023).

    PubMed  PubMed Central  Google Scholar 

  129. Kok, Y. P. et al. Overexpression of Cyclin E1 or Cdc25A leads to replication stress, mitotic aberrancies, and increased sensitivity to replication checkpoint inhibitors. Oncogenesis 9, 88 (2020).

    PubMed  PubMed Central  Google Scholar 

  130. Esguerra, Z. A., Watanabe, G., Okitsu, C. Y., Hsieh, C. L. & Lieber, M. R. DNA-PKcs chemical inhibition versus genetic mutation: Impact on the junctional repair steps of V(D)J recombination. Mol. Immunol. 120, 93–100 (2020).

    PubMed Central  Google Scholar 

  131. Zenke, F. T. et al. Pharmacologic inhibitor of DNA-PK, M3814, potentiates radiotherapy and regresses human tumors in mouse models. Mol. Cancer Ther. 19, 1091–1101 (2020). The authors of this paper report that the DNA-PK inhibitor M3814 strongly enhances the effects of radiotherapy, leading to tumour regression in mouse models of human cancer.

    PubMed  Google Scholar 

  132. Fok, J. H. L. et al. AZD7648 is a potent and selective DNA-PK inhibitor that enhances radiation, chemotherapy and olaparib activity. Nat. Commun. 10, 5065 (2019). AZD7648 is a DNA-PK inhibitor that enhances the efficacy of radiation therapy, chemotherapy and PARP inhibitor olaparib, highlighting its potential as a combination therapy in cancer treatment.

    PubMed  PubMed Central  Google Scholar 

  133. Matsumoto, Y. Development and evolution of DNA-dependent protein kinase inhibitors toward cancer therapy. Int. J. Mol. Sci. 23, 4264 (2022).

    PubMed  PubMed Central  Google Scholar 

  134. Patterson-Fortin, J. et al. _targeting DNA repair with combined inhibition of NHEJ and MMEJ induces synthetic lethality in TP53-mutant cancers. Cancer Res. 82, 3815–3829 (2022).

    PubMed  PubMed Central  Google Scholar 

  135. Karmokar, A. et al. Relevance of ATM status in driving sensitivity to DNA damage response inhibitors in patient-derived xenograft models. Cancers 15, 4195 (2023).

    PubMed  PubMed Central  Google Scholar 

  136. van Bussel, M. T. J. A first-in-man phase 1 study of the DNA-dependent protein kinase inhibitor peposertib (formerly M3814) in patients with advanced solid tumours. Br. J. Cancer 4, 728–735 (2021).

    Google Scholar 

  137. Kruchinin, A. A. & Makarova, A. V. Multifaceted nature of DNA polymerase θ. Int. J. Mol. Sci. 24, 3619 (2023).

    PubMed  PubMed Central  Google Scholar 

  138. Pismataro, M. C. et al. Small molecules _targeting DNA polymerase theta (POLθ) as promising synthetic lethal agents for precision cancer therapy. J. Med. Chem. 66, 6498–6522 (2023).

    PubMed  PubMed Central  Google Scholar 

  139. Shima, N., Munroe, R. J. & Schimenti, J. C. The mouse genomic instability mutation chaos1 is an allele of Polq that exhibits genetic interaction with Atm. Mol. Cell. Biol. 24, 10381–10389 (2004).

    PubMed  PubMed Central  Google Scholar 

  140. Zhou, J. et al. A first-in-class polymerase theta inhibitor selectively _targets homologous-recombination-deficient tumors. Nat. Cancer 6, 598–610 (2021).

    Google Scholar 

  141. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/study/NCT05687110 (2024).

  142. Zatreanu, D. et al. Polθ inhibitors elicit BRCA-gene synthetic lethality and _target PARP inhibitor resistance. Nat. Commun. 12, 3636 (2021). Zatreanu et al. show that Pol Θ inhibitors induce synthetic lethality in cancer cells with BRCA1 and BRCA2 gene mutations and _target cells resistant to PARPis, suggesting a promising therapeutic strategy for overcoming PARPi resistance in BRCA-mutant cancers.

    PubMed  PubMed Central  Google Scholar 

  143. Huang, P., Wang, Y., Zhang, P. & Li, Q. Ubiquitin-specific peptidase 1: assessing its role in cancer therapy. Clin. Exp. Med. 23, 2953–2966 (2023).

    PubMed  Google Scholar 

  144. Kim, J. M. Inactivation of murine Usp1 results in genomic instability and a Fanconi anemia phenotype. Dev. Cell 16, 314–320 (2009).

    PubMed  PubMed Central  Google Scholar 

  145. Lim, K. S. USP1 is required for replication fork protection in BRCA1-deficient tumors. Mol. Cell 72, 925–941.e4 (2018).

    PubMed  Google Scholar 

  146. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/study/NCT05240898 (2024).

  147. Morales-Juarez, D. A. & Jackson, S. P. Clinical prospects of WRN inhibition as a treatment for MSI tumours. NPJ Precis. Oncol. 6, 85 (2022).

    PubMed  PubMed Central  Google Scholar 

  148. Chan, E. M. et al. WRN helicase is a synthetic lethal _target in microsatellite unstable cancers. Nature 568, 551–556 (2019).

    PubMed  PubMed Central  Google Scholar 

  149. Lieb, S. et al. Werner syndrome helicase is a selective vulnerability of microsatellite instability-high tumor cells. eLife 8, e43333 (2019).

    PubMed  PubMed Central  Google Scholar 

  150. Kategaya, L., Perumal, S. K., Hager, J. H. & Belmont, L. D. Werner syndrome helicase is required for the survival of cancer cells with microsatellite instability. iScience 13, 488–497 (2019).

    PubMed  PubMed Central  Google Scholar 

  151. van Wietmarschen, N. et al. Repeat expansions confer WRN dependence in microsatellite-unstable cancers. Nature 586, 292–298 (2020).

    PubMed  PubMed Central  Google Scholar 

  152. Lebel, M. & Leder, P. A deletion within the murine Werner syndrome helicase induces sensitivity to inhibitors of topoisomerase and loss of cellular proliferative capacity. Proc. Natl Acad. Sci. USA 95, 13097–13102 (1998).

    PubMed  PubMed Central  Google Scholar 

  153. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/study/NCT05838768 (2024).

  154. Kaplan, A. R. & Glazer, P. M. Impact of hypoxia on DNA repair and genome integrity. Mutagenesis 35, 61–68 (2020).

    PubMed  Google Scholar 

  155. LaRose, M. et al. A phase I study of a combination of liposomal irinotecan and veliparib in solid tumors. Oncologist 28, 460–e298 (2023).

    PubMed  PubMed Central  Google Scholar 

  156. Berg, S. A. & Choudhury, A. D. Mutual ATRaction: assessing synergy of berzosertib with sacituzumab govitecan. Clin. Cancer Res. 29, 3557–3559 (2023).

    PubMed  Google Scholar 

  157. Carr, M. I. et al. DNA-PK inhibitor, M3814, as a new combination partner of Mylotarg in the treatment of acute myeloid leukemia. Front. Oncol. 10, 127 (2020).

    PubMed  PubMed Central  Google Scholar 

  158. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/study/NCT04039230 (2024).

  159. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/study/NCT04901702 (2024).

  160. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/study/NCT04516447 (2024).

  161. Wickstroem, K. et al. Preclinical combination studies of an FGFR2 _targeted thorium-227 conjugate and the ATR inhibitor BAY 1895344. Int. J. Radiat. Oncol. Biol. Phys. 105, 410–422 (2019).

    PubMed  Google Scholar 

  162. Pan, E. et al. A phase I study of combination olaparib and radium-223 in men with metastatic castration-resistant prostate cancer (mCRPC) with bone metastases (COMRADE). Mol. Cancer Ther. 22, 511–518 (2023).

    PubMed  PubMed Central  Google Scholar 

  163. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/study/NCT04071236 (2024).

  164. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/study/NCT04750954 (2024).

  165. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/study/NCT05868174 (2024).

  166. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/study/NCT04086485 (2024).

  167. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/study/NCT05687136 (2024).

  168. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/study/NCT05396833 (2024).

  169. Clark, C. A. & Yang, E. S. Therapeutic _targeting of DNA damage repair in the era of precision oncology and immune checkpoint inhibitors. J. Immunother. Precis. Oncol. 6, 31–49 (2022).

    PubMed  PubMed Central  Google Scholar 

  170. Chabanon et al. _targeting the DNA damage response in immuno-oncology: developments and opportunities. Nat. Rev. Cancer 21, 701–717 (2021).

    PubMed  Google Scholar 

  171. van Vugt, M. A. & Parkes, E. E. When breaks get hot: inflammatory signaling in BRCA1/2-mutant cancers. Trends Cancer 8, 174–189 (2022).

    PubMed  Google Scholar 

  172. Kornepati, A. V. R. et al. The complementarity of DDR, nucleic acids and anti-tumour immunity. Nature 619, 475–486 (2023).

    PubMed  Google Scholar 

  173. Kwon, M. et al. Phase II study of ceralasertib (AZD6738) in combination with durvalumab in patients with advanced gastric cancer. J. Immunother. Cancer 10, e005041 (2022).

    PubMed  PubMed Central  Google Scholar 

  174. Kim, R. et al. Phase II study of ceralasertib (AZD6738) in combination with durvalumab in patients with advanced/metastatic melanoma who have failed prior anti-PD-1 therapy. Ann. Oncol. 33, 193–203 (2022).

    PubMed  Google Scholar 

  175. Patterson-Fortin, J. et al. Polymerase θ inhibition activates the cGAS-STING pathway and cooperates with immune checkpoint blockade in models of BRCA-deficient cancer. Nat. Commun. 14, 1390 (2023); retraction 14, 8193 (2023).

    PubMed  PubMed Central  Google Scholar 

  176. Muller, P. Y., Milton, M., Lloyd, P., Sims, J. & Brennan, F. R. The minimum anticipated biological effect level (MABEL) for selection of first human dose in clinical trials with monoclonal antibodies. Curr. Opin. Biotechnol. 20, 722–729 (2009).

    PubMed  Google Scholar 

  177. 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).

    PubMed  Google Scholar 

  178. Suto, M. J., Turner, W. R., Arundel-Suto, C. M., Werbel, L. M. & Sebolt-Leopold, J. S. Dihydroisoquinolinones: the design and synthesis of a new series of potent inhibitors of poly(ADP-ribose) polymerase. Anticancer Drug Des. 6, 107–117 (1991).

    PubMed  Google Scholar 

  179. Boulton, S. et al. Potentiation of temozolomide-induced cytotoxicity: a comparative study of the biological effects of poly(ADP-ribose) polymerase inhibitors. Br. J. Cancer 72, 849–856 (1995).

    PubMed  PubMed Central  Google Scholar 

  180. Lindahl, T., Satoh, M. S., Poirier, G. G. & Klungland, A. Post-translational modification of poly(ADP-ribose) polymerase induced by DNA strand breaks. Trends Biochem. Sci. 20, 405–411 (1995).

    PubMed  Google Scholar 

  181. Shao, Z. et al. Inactive PARP1 causes embryonic lethality and genome instability in a dominant-negative manner. Proc. Natl Acad. Sci. USA 120, e2301972120 (2023).

    PubMed  PubMed Central  Google Scholar 

  182. Ström, C. E. et al. Poly (ADP-ribose) polymerase (PARP) is not involved in base excision repair but PARP inhibition traps a single-strand intermediate. Nucleic Acids Res. 39, 3166–3175 (2011).

    PubMed  Google Scholar 

  183. Shen, Y., Aoyagi-Scharber, M. & Wang, B. Trapping poly(ADP-ribose) polymerase. J. Pharmacol. Exp. Ther. 353, 446–457 (2015).

    PubMed  Google Scholar 

  184. Murai, J. & Pommier, Y. PARP trapping beyond homologous recombination and platinum sensitivity in cancers. Annu. Rev. Cancer Biol. 3, 131–150 (2019).

    Google Scholar 

  185. Pandey, N. & Black, B. E. Rapid detection and signaling of DNA damage by PARP-1. Trends Biochem. Sci. 46, 744–757 (2021).

    PubMed  PubMed Central  Google Scholar 

  186. Michelena, J. et al. Analysis of PARP inhibitor toxicity by multidimensional fluorescence microscopy reveals mechanisms of sensitivity and resistance. Nat. Commun. 9, 2678 (2018).

    PubMed  PubMed Central  Google Scholar 

  187. Zandarashvili, L. et al. Structural basis for allosteric PARP-1 retention on DNA breaks. Science 368, eaax6367 (2020).

    PubMed  PubMed Central  Google Scholar 

  188. Hopkins, T. A. et al. Mechanistic dissection of PARP1 trapping and the impact on in vivo tolerability and efficacy of PARP inhibitors. Mol. Cancer Res. 13, 1465–1477 (2015).

    PubMed  Google Scholar 

  189. Rudolph, J., Roberts, G. & Luger, K. Histone Parylation factor 1 contributes to the inhibition of PARP1 by cancer drugs. Nat. Commun. 12, 736 (2021). This paper and Hopkins et al. (2015) offer a comparison of the inhibitory potency and trapping ability of the clinically active PARPis using different biochemical methods.

    PubMed  PubMed Central  Google Scholar 

  190. Schultz, N., Lopez, E., Saleh-Gohari, N. & Helleday, T. Poly(ADP-ribose) polymerase (PARP-1) has a controlling role in homologous recombination. Nucleic Acids Res. 31, 4959–4964 (2003).

    PubMed  PubMed Central  Google Scholar 

  191. Barrows, L. R., Paxton, M. B., Kennedy, K. A. & Thompson, L. H. Characterization of revertants of the CHO EM9 mutant arising during DNA transfection. Carcinogenesis 12, 805–811 (1991).

    PubMed  Google Scholar 

  192. Saleh-Gohari, N. et al. Spontaneous homologous recombination is induced by collapsed replication forks that are caused by endogenous DNA single-strand breaks. Mol. Cell. Biol. 25, 7158–7169 (2005).

    PubMed  PubMed Central  Google Scholar 

  193. Peasland, A. et al. Identification and evaluation of a potent novel ATR inhibitor, NU6027, in breast and ovarian cancer cell lines. Br. J. Cancer 105, 372–381 (2011). This paper is the first to describe a small-molecule inhibitor of ATR that enhances the cytotoxicity of several DNA-damaging agents and increases PARPi cytotoxicity by inducing a HRD phenotype.

    PubMed  PubMed Central  Google Scholar 

  194. Demin, A. A. et al. XRCC1 prevents toxic PARP1 trapping during DNA base excision repair. Mol. Cell 81, 3018–3030.e5 (2021).

    PubMed  PubMed Central  Google Scholar 

  195. Croset, A. et al. Inhibition of DNA damage repair by artificial activation of PARP with siDNA. Nucleic Acids Res. 41, 7344–7355 (2013).

    PubMed  PubMed Central  Google Scholar 

  196. Wang, S. et al. Uncoupling of PARP1 trapping and inhibition using selective PARP1 degradation. Nat. Chem. Biol. 15, 1223–1231 (2019).

    PubMed  PubMed Central  Google Scholar 

  197. Derendorf H. & Schmidt S. Rowland and Tozer’s Clinical Pharmacokinetics and Pharmacodynamics: Concepts and Applications 5th edn (Wolters Kluwer, 2020).

  198. Kwon Y. Handbook of Essential Pharmacokinetics, Pharmacodynamics and Drug Metabolism for Industrial Scientists (Springer, 2013).

  199. Redon, C. E. et al. Histone γH2AX and poly(ADP-ribose) as clinical pharmacodynamic biomarkers. Clin. Cancer Res. 16, 4532–4542 (2010).

    PubMed  PubMed Central  Google Scholar 

  200. Noordermeer, S. M. & van Attikum, H. PARP inhibitor resistance: a tug-of-war in BRCA-mutated cells. Trends Cell Biol. 29, 820–834 (2019).

    PubMed  Google Scholar 

  201. Miller, R. E. et al. ESMO recommendations on predictive biomarker testing for homologous recombination deficiency and PARP inhibitor benefit in ovarian cancer. Ann. Oncol. 12, 1606–1622 (2020). This article is an excellent review of the currently available methods for detecting the HRD phenotype.

    Google Scholar 

  202. Mukhopadhyay, A. et al. Development of a functional assay for homologous recombination status in primary cultures of epithelial ovarian tumor and correlation with sensitivity to poly(ADP-ribose) polymerase inhibitors. Clin. Cancer Res. 16, 2344–2351 (2010). This paper is the first to determine that nearly 60% of ovarian cancers from patients not screened for family history or BRCA mutation are HRD.

    PubMed  Google Scholar 

  203. Pikkusaari, S. et al. Functional homologous recombination assay on FFPE specimens of advanced high-grade serous ovarian cancer predicts clinical outcomes. Clin. Cancer Res. 29, 3110–3123 (2023).

    PubMed  PubMed Central  Google Scholar 

  204. McDonald, E. S. et al. In vivo visualization of PARP inhibitor pharmacodynamics. JCI Insight 6, e146592 (2021).

    PubMed  PubMed Central  Google Scholar 

  205. Shuhendler, A. J. et al. [18 F]-SuPAR: a radiofluorinated probe for noninvasive imaging of DNA damage-dependent poly(ADP-ribose) polymerase activity. Bioconjugate Chem. 30, 1331–1342 (2019).

    Google Scholar 

  206. Knight, J. C., Koustoulidou, S. & Cornelissen, B. Imaging the DNA damage response with PET and SPECT. Eur. J. Nucl. Med. Mol. Imaging 44, 1065–1078 (2017).

    PubMed  PubMed Central  Google Scholar 

  207. Puentes, L. N., Makvandi, M. & Mach, R. H. Molecular imaging: PARP-1 and beyond. J. Nucl. Med. 62, 765–770 (2021).

    PubMed  Google Scholar 

  208. Makvandi, M. et al. A PET imaging agent for evaluating PARP-1 expression in ovarian cancer. J. Clin. Investig. 128, 2116–2126 (2018).

    PubMed  PubMed Central  Google Scholar 

  209. Pantel, A. R. et al. [18 F]FluorThanatrace ([18 F]FTT) PET imaging of PARP-inhibitor drug-_target engagement as a biomarker of response in ovarian cancer, a pilot study. Clin. Cancer Res. 29, 1515–1527 (2023).

    PubMed  Google Scholar 

  210. Lin, R., Zhou, Y., Yan, F., Li, D. & Yuan, Y. BOIN12: Bayesian optimal interval phase I/II trial design for utility-based dose finding in immunotherapy and _targeted therapies. JCO Precis. Oncol. 4, PO.20.00257 (2020).

    PubMed  PubMed Central  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Contributions

All authors researched data for article, contributed substantially to discussion of content, wrote the article and edited and reviewed the manuscript before submission.

Corresponding author

Correspondence to Nicola J Curtin.

Ethics declarations

Competing interests

Y.D. has participated in advisory boards for Clovis Oncology, AstraZeneca, Merck, Tesaro Inc (now GlaxoSmithKline), GlaxoSmithKline and Genmab. Y.D. has received research grant funding from Clovis Oncology and was involved in the preclinical and clinical development of rucaparib. Y.D. has received royalties for her involvement in the development of rucaparib. Y.D. has received research grant funding from AstraZeneca, Tesaro Inc and Roche. F.T.Z. was an employee at EMD Serono, Billerica, MA an affiliate of Merck KGaA, Darmstadt, Germany. Merck KGaA, Darmstadt, Germany, and/or its affiliates have certain rights in patents, patent applications pertaining to DNA Damage Response inhibitors, and F. Z. is named inventor on several patents. N.J.C. was involved in the development of rucaparib, receiving grant funding from Agouron Pharmaceuticals and Pfizer Oncology. She has received grant funding from BioMarin for translational studies with talazoparib. She was involved in the development of DNA-PK inhibitors and ATM inhibitors, receiving grant funding from Kudos (subsequently acquired by AstraZeneca). She was involved in the preclinical evaluation of ATR inhibitors, receiving grant funding from Vertex and Merck. She is inventor on several patents concerning PARP, DNA-PK, ATM and ATR inhibitors. She has received grant funding from Breakpoint Pharmaceuticals and has consulted for Abbvie and Tesaro on PARP inhibitors and Sierra Oncology on CHK1 inhibitors, she currently consults for Duke Street Bio. She diverts her royalty payments to charity.

Peer review

Peer review information

Nature Reviews Drug Discovery thanks the anonymous reviewers for their contribution to the peer review of this work.

Additional information

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

Related links

Beacon-Db: https://data.beacon-intelligence.com/

DepMap-ATR: https://depmap.org/portal/gene/ATR?tab=overview

DepMap-DNA-PK: https://depmap.org/portal/gene/PRKDC?tab=overview

DepMap-POLQ: https://depmap.org/portal/gene/POLQ?tab=overview

DepMap-USP1: https://depmap.org/portal/gene/USP1?tab=overview

DepMap-WEE1: https://depmap.org/portal/gene/WEE1?tab=overview

DepMap-WRN: https://depmap.org/portal/gene/WRN?tab=overview

Supplementary information

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Drew, Y., Zenke, F.T. & Curtin, N.J. DNA damage response inhibitors in cancer therapy: lessons from the past, current status and future implications. Nat Rev Drug Discov 24, 19–39 (2025). https://doi.org/10.1038/s41573-024-01060-w

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41573-024-01060-w

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer
  NODES
admin 1
Association 1
INTERN 1
Note 2
Project 2
twitter 1