1932

Abstract

Poly(ADP-ribose) polymerase inhibitors (PARPis) have recently been approved for the treatment of ovarian and breast cancers with mutations, as well as for maintenance therapies regardless of mutation for ovarian and primary peritoneal cancers that previously responded to platinum-based chemotherapy. The rationale of these indications is derived from the facts that cancer cells with mutations are defective in homologous recombination (HR), which confers synthetic lethality with PARPis, and that some of the sensitivity-determining factors for PARPis are shared with platinums. Although and are central for HR, more players within and beyond HR are emerging as response determinants to PARPis. Furthermore, there are similarities as well as differences in the DNA lesions and repair pathways induced by PARPis, platinums, and camptothecin topoisomerase 1 (TOP1) inhibitors. Here we review the sensitivity-determining factors for PARPis and the rationale for using PARPis as single agents and in combination therapy for cancers.

Loading

Article metrics loading...

/content/journals/10.1146/annurev-cancerbio-030518-055914
2019-03-04
2024-04-26
Loading full text...

Full text loading...

/deliver/fulltext/cancerbio/3/1/annurev-cancerbio-030518-055914.html?itemId=/content/journals/10.1146/annurev-cancerbio-030518-055914&mimeType=html&fmt=ahah

Literature Cited

  1. Al Abo M, Dejsuphong D, Hirota K, Yonetani Y, Yamazoe M et al. 2014. Compensatory functions and interdependency of the DNA-binding domain of BRCA2 with the BRCA1–PALB2–BRCA2 complex. Cancer Res 74:797–807
    [Google Scholar]
  2. Andres SN, Williams RS 2017. CtIP/Ctp1/Sae2, molecular form fit for function. DNA Repair 56:109–17
    [Google Scholar]
  3. Ashworth A. 2008. A synthetic lethal therapeutic approach: poly(ADP) ribose polymerase inhibitors for the treatment of cancers deficient in DNA double-strand break repair. J. Clin. Oncol. 26:3785–90
    [Google Scholar]
  4. Bajrami I, Frankum JR, Konde A, Miller RE, Rehman FL et al. 2014. Genome-wide profiling of genetic synthetic lethality identifies CDK12 as a novel determinant of PARP1/2 inhibitor sensitivity. Cancer Res 74:287–97
    [Google Scholar]
  5. Bajrami I, Kigozi A, Van Weverwijk A, Brough R, Frankum J et al. 2012. Synthetic lethality of PARP and NAMPT inhibition in triple-negative breast cancer cells. EMBO Mol. Med. 4:1087–96
    [Google Scholar]
  6. Barretina J, Caponigro G, Stransky N, Venkatesan K, Margolin AA et al. 2012. The Cancer Cell Line Encyclopedia enables predictive modelling of anticancer drug sensitivity. Nature 483:603–7
    [Google Scholar]
  7. Benjamin RC, Gill DM 1980. ADP-ribosylation in mammalian cell ghosts: dependence of poly(ADP-ribose) synthesis on strand breakage in DNA. J. Biol. Chem. 255:10493–501
    [Google Scholar]
  8. Bock FJ, Todorova TT, Chang P 2015. The promise of proteomics for the study of ADP-ribosylation. Mol. Cell 58:911–24
    [Google Scholar]
  9. Brill E, Yokoyama T, Nair J, Yu M, Ahn YR, Lee JM 2017. Prexasertib, a cell cycle checkpoint kinases 1 and 2 inhibitor, increases in vitro toxicity of PARP inhibition by preventing Rad51 foci formation in BRCA wild type high-grade serous ovarian cancer. Oncotarget 8:111026–40
    [Google Scholar]
  10. Bruno PM, Liu Y, Park GY, Murai J, Koch CE et al. 2017. A subset of platinum-containing chemotherapeutic agents kills cells by inducing ribosome biogenesis stress. Nat. Med. 23:461–71
    [Google Scholar]
  11. Bryant HE, Helleday T 2006. Inhibition of poly (ADP-ribose) polymerase activates ATM which is required for subsequent homologous recombination repair. Nucleic Acids Res 34:1685–91
    [Google Scholar]
  12. Bryant HE, Petermann E, Schultz N, Jemth AS, Loseva O et al. 2009. PARP is activated at stalled forks to mediate Mre11-dependent replication restart and recombination. EMBO J 28:2601–15
    [Google Scholar]
  13. Bryant HE, Schultz N, Thomas HD, Parker KM, Flower D et al. 2005. Specific killing of BRCA2-deficient tumours with inhibitors of poly(ADP-ribose) polymerase. Nature 434:913–17
    [Google Scholar]
  14. Buis J, Stoneham T, Spehalski E, Ferguson DO 2012. Mre11 regulates CtIP-dependent double-strand break repair by interaction with CDK2. Nat. Struct. Mol. Biol. 19:246–52
    [Google Scholar]
  15. Bunting SF, Callen E, Wong N, Chen HT, Polato F et al. 2010. 53BP1 inhibits homologous recombination in Brca1-deficient cells by blocking resection of DNA breaks. Cell 141:243–54
    [Google Scholar]
  16. Ceccaldi R, Liu JC, Amunugama R, Hajdu I, Primack B et al. 2015.a Homologous-recombination-deficient tumours are dependent on Polθ-mediated repair. Nature 518:258–62
    [Google Scholar]
  17. Ceccaldi R, O'Connor KW, Mouw KW, Li AY, Matulonis UA et al. 2015.b A unique subset of epithelial ovarian cancers with platinum sensitivity and PARP inhibitor resistance. Cancer Res 75:628–34
    [Google Scholar]
  18. Cejka P. 2015. DNA end resection: Nucleases team up with the right partners to initiate homologous recombination. J. Biol. Chem. 290:22931–38
    [Google Scholar]
  19. Chand SN, Zarei M, Schiewer MJ, Kamath AR, Romeo C et al. 2017. Posttranscriptional regulation of PARG mRNA by HuR facilitates DNA repair and resistance to PARP inhibitors. Cancer Res 77:5011–25
    [Google Scholar]
  20. Chen WH, Song SS, Qi MH, Huan XJ, Wang YQ et al. 2017. Discovery of potent 2,4-difluoro-linker poly(ADP-ribose) polymerase 1 inhibitors with enhanced water solubility and in vivo anticancer efficacy. Acta Pharmacol. Sin. 38:1521–32
    [Google Scholar]
  21. Chen X, Huan X, Liu Q, Wang Y, He Q et al. 2018. Design and synthesis of 2-(4,5,6,7-tetrahydrothienopyridin-2-yl)-benzoimidazole carboxamides as novel orally efficacious poly(ADP-ribose)polymerase (PARP) inhibitors. Eur. J. Med. Chem. 145:389–403
    [Google Scholar]
  22. Chernikova SB, Game JC, Brown JM 2012. Inhibiting homologous recombination for cancer therapy. Cancer Biol. Ther. 13:61–68
    [Google Scholar]
  23. Chun J, Buechelmaier ES, Powell SN 2013. Rad51 paralog complexes BCDX2 and CX3 act at different stages in the BRCA1-BRCA2-dependent homologous recombination pathway. Mol. Cell. Biol. 33:387–95
    [Google Scholar]
  24. Cistulli C, Lavrik OI, Prasad R, Hou E, Wilson SH 2004. AP endonuclease and poly(ADP-ribose) polymerase-1 interact with the same base excision repair intermediate. DNA Repair 3:581–91
    [Google Scholar]
  25. Cliby WA, Roberts CJ, Cimprich KA, Stringer CM, Lamb JR et al. 1998. Overexpression of a kinase-inactive ATR protein causes sensitivity to DNA-damaging agents and defects in cell cycle checkpoints. EMBO J 17:159–69
    [Google Scholar]
  26. Coleman RL, Oza AM, Lorusso D, Aghajanian C, Oaknin A et al. 2017. Rucaparib maintenance treatment for recurrent ovarian carcinoma after response to platinum therapy (ARIEL3): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 390:1949–61
    [Google Scholar]
  27. Curtin NJ, Szabo C 2013. Therapeutic applications of PARP inhibitors: anticancer therapy and beyond. Mol. Aspects Med. 34:61217–56
    [Google Scholar]
  28. Das BB, Huang SY, Murai J, Rehman I, Ame JC et al. 2014. PARP1–TDP1 coupling for the repair of topoisomerase I–induced DNA damage. Nucleic Acids Res 42:4435–49
    [Google Scholar]
  29. Davidson D, Wang Y, Aloyz R, Panasci L 2013. The PARP inhibitor ABT-888 synergizes irinotecan treatment of colon cancer cell lines. Investig. New Drugs 31:461–68
    [Google Scholar]
  30. Dehe PM, Gaillard PH 2017. Control of structure-specific endonucleases to maintain genome stability. Nat. Rev. Mol. Cell Biol. 18:315–30
    [Google Scholar]
  31. Ding X, Ray Chaudhuri A, Callen E, Pang Y, Biswas K et al. 2016. Synthetic viability by BRCA2 and PARP1/ARTD1 deficiencies. Nat. Commun. 7:12425
    [Google Scholar]
  32. Durkacz BW, Omidiji O, Gray DA, Shall S 1980. (ADP-ribose)n participates in DNA excision repair. Nature 283:593–96
    [Google Scholar]
  33. Farmer H, McCabe N, Lord CJ, Tutt AN, Johnson DA et al. 2005. Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy. Nature 434:917–21
    [Google Scholar]
  34. Feng X, Koh DW 2013. Inhibition of poly(ADP-ribose) polymerase-1 or poly(ADPribose) glycohydrolase individually, but not in combination, leads to improved chemotherapeutic efficacy in HeLa cells. Int. J. Oncol. 42:749–56
    [Google Scholar]
  35. Fong PC, Boss DS, Yap TA, Tutt A, Wu P et al. 2009. Inhibition of poly(ADP-ribose) polymerase in tumors from BRCA mutation carriers. N. Engl. J. Med. 361:123–34
    [Google Scholar]
  36. Friedel AM, Pike BL, Gasser SM 2009. ATR/Mec1: coordinating fork stability and repair. Curr. Opin. Cell Biol. 21:237–44
    [Google Scholar]
  37. Gardner EE, Lok BH, Schneeberger VE, Desmeules P, Miles LA et al. 2017. Chemosensitive relapse in small cell lung cancer proceeds through an EZH2-SLFN11 axis. Cancer Cell 31:286–99
    [Google Scholar]
  38. Gibson BA, Kraus WL 2012. New insights into the molecular and cellular functions of poly(ADP-ribose) and PARPs. Nat. Rev. Mol. Cell Biol. 13:411–24
    [Google Scholar]
  39. Goetz JD, Motycka TA, Han M, Jasin M, Tomkinson AE 2005. Reduced repair of DNA double-strand breaks by homologous recombination in a DNA ligase I-deficient human cell line. DNA Repair 4:649–54
    [Google Scholar]
  40. Hazra TK, Das A, Das S, Choudhury S, Kow YW, Roy R 2007. Oxidative DNA damage repair in mammalian cells: a new perspective. DNA Repair 6:470–80
    [Google Scholar]
  41. Hellebrand H, Sutter C, Honisch E, Gross E, Wappenschmidt B et al. 2011. Germline mutations in the PALB2 gene are population specific and occur with low frequencies in familial breast cancer. Hum. Mutat. 32:E2176–88
    [Google Scholar]
  42. Helleday T. 2011. The underlying mechanism for the PARP and BRCA synthetic lethality: clearing up the misunderstandings. Mol. Oncol. 5:387–93
    [Google Scholar]
  43. Helleday T. 2016. PARP inhibitor receives FDA breakthrough therapy designation in castration resistant prostate cancer: beyond germline BRCA mutations. Ann. Oncol. 27:755–57
    [Google Scholar]
  44. Higuchi T, Flies DB, Marjon NA, Mantia-Smaldone G, Ronner L et al. 2015. CTLA-4 blockade synergizes therapeutically with PARP inhibition in BRCA1-deficient ovarian cancer. Cancer Immunol. Res. 3:1257–68
    [Google Scholar]
  45. Hoa NN, Akagawa R, Yamasaki T, Hirota K, Sasa K et al. 2015. Relative contribution of four nucleases, CtIP, Dna2, Exo1 and Mre11, to the initial step of DNA double-strand break repair by homologous recombination in both the chicken DT40 and human TK6 cell lines. Genes Cells 20:1059–76
    [Google Scholar]
  46. Hochegger H, Dejsuphong D, Fukushima T, Morrison C, Sonoda E et al. 2006. Parp-1 protects homologous recombination from interference by Ku and Ligase IV in vertebrate cells. EMBO J 25:1305–14
    [Google Scholar]
  47. Horton JK, Stefanick DF, Prasad R, Gassman NR, Kedar PS, Wilson SH 2014. Base excision repair defects invoke hypersensitivity to PARP inhibition. Mol. Cancer Res. 12:1128–39
    [Google Scholar]
  48. Horton JK, Wilson SH 2013. Strategic combination of DNA-damaging agent and PARP inhibitor results in enhanced cytotoxicity. Front. Oncol. 3:257
    [Google Scholar]
  49. Jaspers JE, Kersbergen A, Boon U, Sol W, van Deemter L et al. 2013. Loss of 53BP1 causes PARP inhibitor resistance in Brca1-mutated mouse mammary tumors. Cancer Discov 3:68–81
    [Google Scholar]
  50. Ji J, Kinders RJ, Zhang Y, Rubinstein L, Kummar S et al. 2011. Modeling pharmacodynamic response to the poly(ADP-Ribose) polymerase inhibitor ABT-888 in human peripheral blood mononuclear cells. PLOS ONE 6:e26152
    [Google Scholar]
  51. Jiao S, Xia W, Yamaguchi H, Wei Y, Chen MK et al. 2017. PARP inhibitor upregulates PD-L1 expression and enhances cancer-associated immunosuppression. Clin. Cancer Res. 23:3711–20
    [Google Scholar]
  52. Josse R, Martin SE, Guha R, Ormanoglu P, Pfister TD et al. 2014. ATR inhibitors VE-821 and VX-970 sensitize cancer cells to topoisomerase I inhibitors by disabling DNA replication initiation and fork elongation responses. Cancer Res 74:6968–79
    [Google Scholar]
  53. Karakashev S, Zhu H, Yokoyama Y, Zhao B, Fatkhutdinov N et al. 2017. BET bromodomain inhibition synergizes with PARP inhibitor in epithelial ovarian cancer. Cell Rep 21:3398–405
    [Google Scholar]
  54. Karnitz LM, Zou L 2015. Molecular pathways: targeting ATR in cancer therapy. Clin. Cancer Res. 21:4780–85
    [Google Scholar]
  55. Kedar PS, Stefanick DF, Horton JK, Wilson SH 2012. Increased PARP-1 association with DNA in alkylation damaged, PARP-inhibited mouse fibroblasts. Mol. Cancer Res. 10:360–68
    [Google Scholar]
  56. Kelland L. 2007. The resurgence of platinum-based cancer chemotherapy. Nat. Rev. Cancer 7:573–84
    [Google Scholar]
  57. Kennedy RD, D'Andrea AD 2006. DNA repair pathways in clinical practice: lessons from pediatric cancer susceptibility syndromes. J. Clin. Oncol. 24:3799–808
    [Google Scholar]
  58. Kim MY, Mauro S, Gevry N, Lis JT, Kraus WL 2004. NAD+-dependent modulation of chromatin structure and transcription by nucleosome binding properties of PARP-1. Cell 119:803–14
    [Google Scholar]
  59. Konstantinopoulos PA, Ceccaldi R, Shapiro GI, D'Andrea AD 2015. Homologous recombination deficiency: exploiting the fundamental vulnerability of ovarian cancer. Cancer Discov 5:1137–54
    [Google Scholar]
  60. Kummar S, Chen A, Ji J, Zhang Y, Reid JM et al. 2011. Phase I study of PARP inhibitor ABT-888 in combination with topotecan in adults with refractory solid tumors and lymphomas. Cancer Res 71:5626–34
    [Google Scholar]
  61. Kummar S, Chen A, Parchment RE, Kinders RJ, Ji J et al. 2012. Advances in using PARP inhibitors to treat cancer. BMC Med 10:25
    [Google Scholar]
  62. Ledermann J, Harter P, Gourley C, Friedlander M, Vergote I et al. 2012. Olaparib maintenance therapy in platinum-sensitive relapsed ovarian cancer. N. Engl. J. Med. 366:1382–92
    [Google Scholar]
  63. Lee JM, Nair J, Zimmer A, Lipkowitz S, Annunziata CM et al. 2018. Prexasertib, a cell cycle checkpoint kinase 1 and 2 inhibitor, in BRCA wild-type recurrent high-grade serous ovarian cancer: a first-in-class proof-of-concept phase 2 study. Lancet Oncol 19:207–15
    [Google Scholar]
  64. Leidecker O, Bonfiglio JJ, Colby T, Zhang Q, Atanassov I et al. 2016. Serine is a new target residue for endogenous ADP-ribosylation on histones. Nat. Chem. Biol. 12:998–1000
    [Google Scholar]
  65. Li M, Yu X 2015. The role of poly(ADP-ribosyl)ation in DNA damage response and cancer chemotherapy. Oncogene 34:3349–56
    [Google Scholar]
  66. Lok BH, Gardner EE, Schneeberger VE, Ni A, Desmeules P et al. 2017. PARP inhibitor activity correlates with SLFN11 expression and demonstrates synergy with temozolomide in small cell lung cancer. Clin. Cancer Res. 23:523–35
    [Google Scholar]
  67. Lord CJ, Ashworth A 2012. The DNA damage response and cancer therapy. Nature 481:287–94
    [Google Scholar]
  68. Lord CJ, Ashworth A 2017. PARP inhibitors: synthetic lethality in the clinic. Science 355:1152–58
    [Google Scholar]
  69. Maede Y, Shimizu H, Fukushima T, Kogame T, Nakamura T et al. 2014. Differential and common DNA repair pathways for topoisomerase I- and II-targeted drugs in a genetic DT40 repair cell screen panel. Mol. Cancer Ther. 13:214–20
    [Google Scholar]
  70. Martinez-Zamudio R, Ha HC 2012. Histone ADP-ribosylation facilitates gene transcription by directly remodeling nucleosomes. Mol. Cell. Biol. 32:2490–502
    [Google Scholar]
  71. McCabe N, Turner NC, Lord CJ, Kluzek K, Bialkowska A et al. 2006. Deficiency in the repair of DNA damage by homologous recombination and sensitivity to poly(ADP-ribose) polymerase inhibition. Cancer Res 66:8109–15
    [Google Scholar]
  72. Ménissier-de Murcia J, Ricoul M, Tartier L, Niedergang C, Huber A et al. 2003. Functional interaction between PARP-1 and PARP-2 in chromosome stability and embryonic development in mouse. EMBO J 22:2255–63
    [Google Scholar]
  73. Messner S, Hottiger MO 2011. Histone ADP-ribosylation in DNA repair, replication and transcription. Trends Cell Biol 21:534–42
    [Google Scholar]
  74. Michl J, Zimmer J, Tarsounas M 2016. Interplay between Fanconi anemia and homologous recombination pathways in genome integrity. EMBO J 35:909–23
    [Google Scholar]
  75. Mirza MR, Monk BJ, Herrstedt J, Oza AM, Mahner S et al. 2016. Niraparib maintenance therapy in platinum-sensitive, recurrent ovarian cancer. N. Engl. J. Med. 375:2154–64
    [Google Scholar]
  76. Mouw KW, Goldberg MS, Konstantinopoulos PA, D'Andrea AD 2017. DNA damage and repair biomarkers of immunotherapy response. Cancer Discov 7:675–93
    [Google Scholar]
  77. Murai J. 2017. Targeting DNA repair and replication stress in the treatment of ovarian cancer. Int. J. Clin. Oncol. 22:619–28
    [Google Scholar]
  78. Murai J, Feng Y, Yu GK, Ru Y, Tang SW et al. 2016. Resistance to PARP inhibitors by SLFN11 inactivation can be overcome by ATR inhibition. Oncotarget 7:76534–50
    [Google Scholar]
  79. Murai J, Huang SY, Das BB, Renaud A, Zhang Y et al. 2012. Trapping of PARP1 and PARP2 by clinical PARP inhibitors. Cancer Res 72:5588–99
    [Google Scholar]
  80. Murai J, Huang SY, Renaud A, Zhang Y, Ji J et al. 2014.a Stereospecific PARP trapping by BMN 673 and comparison with olaparib and rucaparib. Mol. Cancer Ther. 13:433–43
    [Google Scholar]
  81. Murai J, Marchand C, Shahane SA, Sun H, Huang R et al. 2014.b Identification of novel PARP inhibitors using a cell-based TDP1 inhibitory assay in a quantitative high-throughput screening platform. DNA Repair 21:177–82
    [Google Scholar]
  82. Murai J, Pommier Y 2015. Classification of PARP inhibitors based on PARP trapping and catalytic inhibition, and rationale for combination with topoisomerase I inhibitors and alkylating agents. PARP Inhibitors for Cancer Therapy NJ Curtin, RA Sharma 261–74 Cham, Switz.: Humana
    [Google Scholar]
  83. Murai J, Tang SW, Leo E, Baechler SA, Redon CE et al. 2018. SLFN11 blocks stressed replication forks independently of ATR. Mol. Cell 69:371–84.e6
    [Google Scholar]
  84. Murai J, Zhang Y, Morris J, Ji J, Takeda S et al. 2014.c 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. 349:408–16
    [Google Scholar]
  85. Musella A, Bardhi E, Marchetti C, Vertechy L, Santangelo G et al. 2018. Rucaparib: an emerging PARP inhibitor for treatment of recurrent ovarian cancer. Cancer Treat. Rev. 66:7–14
    [Google Scholar]
  86. Nam EA, Cortez D 2011. ATR signalling: more than meeting at the fork. Biochem. J. 436:527–36
    [Google Scholar]
  87. Nogales V, Reinhold WC, Varma S, Martinez-Cardus A, Moutinho C et al. 2016. Epigenetic inactivation of the putative DNA/RNA helicase SLFN11 in human cancer confers resistance to platinum drugs. Oncotarget 7:3084–97
    [Google Scholar]
  88. O'Sullivan CC, Moon DH, Kohn EC, Lee JM 2014. Beyond breast and ovarian cancers: PARP inhibitors for BRCA mutation-associated and BRCA-like solid tumors. Front. Oncol. 4:42
    [Google Scholar]
  89. Papeo G, Posteri H, Borghi D, Busel AA, Caprera F et al. 2015. Discovery of 2-[1-(4,4-difluorocyclohexyl)piperidin-4-yl]-6-fluoro-3-oxo-2,3-dihydro-1H-isoind ole-4-carboxamide (NMS-P118): a potent, orally available, and highly selective PARP-1 inhibitor for cancer therapy. J. Med. Chem. 58:6875–98
    [Google Scholar]
  90. Patel MR, Bhatt A, Steffen JD, Chergui A, Murai J et al. 2014. Discovery and structure-activity relationship of novel 2,3-dihydrobenzofuran-7-carboxamide and 2,3-dihydrobenzofuran-3(2H)-one-7-carboxamide derivatives as poly(ADP-ribose)polymerase-1 inhibitors. J. Med. Chem. 57:5579–601
    [Google Scholar]
  91. Pietrobon V, Freon K, Hardy J, Costes A, Iraqui I et al. 2014. The chromatin assembly factor 1 promotes Rad51-dependent template switches at replication forks by counteracting D-loop disassembly by the RecQ-type helicase Rqh1. PLOS Biol 12:e1001968
    [Google Scholar]
  92. Pommier Y 2012. DNA Topoisomerases and Cancer New York: Humana
  93. Pommier Y. 2013. Drugging topoisomerases: lessons and challenges. ACS Chem. Biol. 8:82–95
    [Google Scholar]
  94. Pommier Y, Huang SY, Gao R, Das BB, Murai J, Marchand C 2014. Tyrosyl-DNA-phosphodiesterases (TDP1 and TDP2). DNA Repair 19:114–29
    [Google Scholar]
  95. Pommier Y, O'Connor MJ, de Bono J 2016.a Laying a trap to kill cancer cells: PARP inhibitors and their mechanisms of action. Sci. Transl. Med. 8:362ps17
    [Google Scholar]
  96. Pommier Y, Sun Y, Huang SN, Nitiss JL 2016.b Roles of eukaryotic topoisomerases in transcription, replication and genomic stability. Nat. Rev. Mol. Cell Biol. 17:703–21
    [Google Scholar]
  97. Prakash R, Zhang Y, Feng W, Jasin M 2015. Homologous recombination and human health: the roles of BRCA1, BRCA2, and associated proteins. Cold Spring Harb. Perspect. Biol. 7:a016600
    [Google Scholar]
  98. Rajapakse VN, Luna A, Yamade M, Loman L, Varma S et al. 2018. CellMinerCDB for integrative cross-database genomics and pharmacogenomics analyses of cancer cell lines. iScience 10:247–247
    [Google Scholar]
  99. Ray Chaudhuri A, Callen E, Ding X, Gogola E, Duarte AA et al. 2016. Replication fork stability confers chemoresistance in BRCA-deficient cells. Nature 535:382–87 Erratum. 2016 Nature 539:456
    [Google Scholar]
  100. Ray Chaudhuri A, Hashimoto Y, Herrador R, Neelsen KJ, Fachinetti D et al. 2012. Topoisomerase I poisoning results in PARP-mediated replication fork reversal. Nat. Struct. Mol. Biol. 19:417–23
    [Google Scholar]
  101. Regairaz M, Zhang Y-W, Fu H, Agama KK, Tata N et al. 2011. Mus81-mediated DNA cleavage resolves replication forks stalled by topoisomerase I–DNA complexes. J. Cell Biol. 195:739–49
    [Google Scholar]
  102. Robson M, Im SA, Senkus E, Xu BH, Domchek SM et al. 2017. Olaparib for metastatic breast cancer in patients with a germline BRCA mutation. N. Engl. J. Med. 377:523–33
    [Google Scholar]
  103. Rondinelli B, Gogola E, Yucel H, Duarte AA, van de Ven M et al. 2017. EZH2 promotes degradation of stalled replication forks by recruiting MUS81 through histone H3 trimethylation. Nat. Cell Biol. 19:1371–78
    [Google Scholar]
  104. Rottenberg S, Jaspers JE, Kersbergen A, van der Burg E, Nygren AO et al. 2008. High sensitivity of BRCA1-deficient mammary tumors to the PARP inhibitor AZD2281 alone and in combination with platinum drugs. PNAS 105:17079–84
    [Google Scholar]
  105. Rouleau M, Patel A, Hendzel MJ, Kaufmann SH, Poirier GG 2010. PARP inhibition: PARP1 and beyond. Nat. Rev. Cancer 10:293–301
    [Google Scholar]
  106. Sato K, Shimomuki M, Katsuki Y, Takahashi D, Kobayashi W et al. 2016. FANCI-FANCD2 stabilizes the RAD51-DNA complex by binding RAD51 and protects the 5′-DNA end. Nucleic Acids Res 44:10758–71
    [Google Scholar]
  107. Schlacher K, Wu H, Jasin M 2012. A distinct replication fork protection pathway connects Fanconi anemia tumor suppressors to RAD51-BRCA1/2. Cancer Cell 22:106–16
    [Google Scholar]
  108. Schmitt A, Knittel G, Welcker D, Yang TP, George J et al. 2017. ATM deficiency is associated with sensitivity to PARP1- and ATR inhibitors in lung adenocarcinoma. Cancer Res 77:3040–56
    [Google Scholar]
  109. Seiler JA, Conti C, Syed A, Aladjem MI, Pommier Y 2007. The intra-S-phase checkpoint affects both DNA replication initiation and elongation: single-cell and -DNA fiber analyses. Mol. Cell. Biol. 27:5806–18
    [Google Scholar]
  110. Shelton JW, Waxweiler TV, Landry J, Gao H, Xu Y et al. 2013. In vitro and in vivo enhancement of chemoradiation using the oral PARP inhibitor ABT-888 in colorectal cancer cells. Int. J. Radiat. Oncol. Biol. Phys. 86:469–76
    [Google Scholar]
  111. Smith J, Tho LM, Xu N, Gillespie DA 2010. The ATM-Chk2 and ATR-Chk1 pathways in DNA damage signaling and cancer. Adv Cancer Res 108:73–112
    [Google Scholar]
  112. Sørensen CS, Beck H, Nähse-Kumpf V, Syljuåsen RG 2011. Faithful DNA replication requires regulation of CDK activity by checkpoint kinases. InTechOpen http://doi.org/10.5772/18616
    [Crossref]
  113. Stewart CA, Tong P, Cardnell RJ, Sen T, Li L et al. 2017. Dynamic variations in epithelial-to-mesenchymal transition (EMT), ATM, and SLFN11 govern response to PARP inhibitors and cisplatin in small cell lung cancer. Oncotarget 8:1728575–87
    [Google Scholar]
  114. Sun C, Yin J, Fang Y, Chen J, Jeong KJ et al. 2018. BRD4 Inhibition is synthetic lethal with PARP inhibitors through the induction of homologous recombination deficiency. Cancer Cell 33:401–16.e8
    [Google Scholar]
  115. Tahara M, Inoue T, Sato F, Miyakura Y, Horie H et al. 2014. The use of olaparib (AZD2281) potentiates SN-38 cytotoxicity in colon cancer cells by indirect inhibition of Rad51-mediated repair of DNA double-strand breaks. Mol. Cancer Ther. 13:1170–80
    [Google Scholar]
  116. Tang SW, Thomas A, Murai J, Trepel JB, Bates SE et al. 2018. Overcoming resistance to DNA-targeted agents by epigenetic activation of Schlafen 11 (SLFN11) expression with class I histone deacetylase inhibitors. Clin. Cancer Res. 24:81944–53
    [Google Scholar]
  117. Tang Y, Hamed HA, Poklepovic A, Dai Y, Grant S, Dent P 2012. Poly(ADP-ribose) polymerase 1 modulates the lethality of CHK1 inhibitors in mammary tumors. Mol. Pharmacol. 82:322–32
    [Google Scholar]
  118. Tentori L, Leonetti C, Scarsella M, Muzi A, Mazzon E et al. 2006. Inhibition of poly(ADP-ribose) polymerase prevents irinotecan-induced intestinal damage and enhances irinotecan/temozolomide efficacy against colon carcinoma. FASEB J 20:1709–11
    [Google Scholar]
  119. Wahlberg E, Karlberg T, Kouznetsova E, Markova N, Macchiarulo A et al. 2012. Family-wide chemical profiling and structural analysis of PARP and tankyrase inhibitors. Nat. Biotechnol. 30:283–88
    [Google Scholar]
  120. Wright RH, Castellano G, Bonet J, Le Dily F, Font-Mateu J et al. 2012. CDK2-dependent activation of PARP-1 is required for hormonal gene regulation in breast cancer cells. Genes Dev 26:1972–83
    [Google Scholar]
  121. Wright RH, Lioutas A, Le Dily F, Soronellas D, Pohl A et al. 2016. ADP-ribose-derived nuclear ATP synthesis by NUDIX5 is required for chromatin remodeling. Science 352:1221–25
    [Google Scholar]
  122. Wyatt HD, West SC 2014. Holliday junction resolvases. Cold Spring Harb. Perspect. Biol. 6:a023192
    [Google Scholar]
  123. Xu G, Chapman JR, Brandsma I, Yuan J, Mistrik M et al. 2015. REV7 counteracts DNA double-strand break resection and affects PARP inhibition. Nature 521:541–44
    [Google Scholar]
  124. Yang L, Zhang Y, Shan W, Hu Z, Yuan J et al. 2017. Repression of BET activity sensitizes homologous recombination-proficient cancers to PARP inhibition. Sci. Transl. Med. 9:eaal1645
    [Google Scholar]
  125. Yang W, Soares J, Greninger P, Edelman EJ, Lightfoot H et al. 2013. Genomics of drug sensitivity in cancer (GDSC): a resource for therapeutic biomarker discovery in cancer cells. Nucleic Acids Res 41:D955–61
    [Google Scholar]
  126. Zeman MK, Cimprich KA 2014. Causes and consequences of replication stress. Nat. Cell Biol. 16:2–9
    [Google Scholar]
  127. Zhang YW, Regairaz M, Seiler JA, Agama KK, Doroshow JH, Pommier Y 2011. Poly(ADP-ribose) polymerase and XPF-ERCC1 participate in distinct pathways for the repair of topoisomerase I-induced DNA damage in mammalian cells. Nucleic Acids Res 39:3607–20
    [Google Scholar]
  128. Zoppoli G, Regairaz M, Leo E, Reinhold WC, Varma S et al. 2012. Putative DNA/RNA helicase Schlafen-11 (SLFN11) sensitizes cancer cells to DNA-damaging agents. PNAS 109:15030–35
    [Google Scholar]
/content/journals/10.1146/annurev-cancerbio-030518-055914
Loading
/content/journals/10.1146/annurev-cancerbio-030518-055914
Loading

Data & Media loading...

  • Article Type: Review Article
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error