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.

  • Analysis
  • Published:

Therapeutic opportunities within the DNA damage response

Key Points

  • The purpose of this Analysis article is to comprehensively overview the DNA damage response (DDR) pathways, assess current drug development activities in this area, examine the emerging understanding of how defects in genome stability mechanisms contribute to cancer and, by utilizing systematic bioinformatics and chemogenomic approaches, highlight the considerable opportunities these pathways offer as targets for new cancer treatments.

  • We first provide a brief review of the different types of DNA damage and the DDR pathways and mechanisms that have evolved to repair them, how defects in these systems promote cancer, how some existing therapies operate via DNA damage, and the emerging concept of pharmacological synthetic lethality that takes advantage of cancer-associated DDR defects.

  • We then describe our assembly of an expert-curated set of 450 proteins involved in the DDR and in closely associated processes such as chromatin remodelling and chromosome segregation. We present their assignment to specific DDR pathways and the functional interactions within and between pathways. We classify these proteins into functional classes and discuss the current knowledge of their involvement in cancer.

  • We then undertake a systematic bioinformatics analysis of the DDR proteins in 15 different cancers, using data from The Cancer Genome Atlas. We analyse protein coding mutations, copy-number variation and overexpression and underexpression. We find that DDR processes that are commonly mutated differ markedly between different cancer types, but note that every DDR process is functionally impaired to some extent in one or more cancer type, providing many opportunities for drug therapy but with substantial challenges for optimal targeting.

  • We explore the concept of synthetic lethality and document current examples, including pharmacological inhibition of poly(ADP-ribose) polymerase 1 (PARP1) in homologous recombination-deficient cancers. We use data from whole-genome negative genetic analysis of yeast to identify further opportunities for synthetic lethal relationships with the curated DDR proteins that may be amenable to drug discovery.

  • Next we document the limited set of drug discovery projects that are currently targeting DDR proteins and discuss the application of druggability analysis to expand the therapeutic opportunities among these proteins.

  • Finally, we use a combination of approaches to identify a number of classes among the DDR proteins — particularly DNA helicases and scaffold proteins — that should have particular promise as cancer drug targets. We also highlight potentially druggable targets for each of the DDR pathways.

Abstract

The DNA damage response (DDR) is essential for maintaining the genomic integrity of the cell, and its disruption is one of the hallmarks of cancer. Classically, defects in the DDR have been exploited therapeutically in the treatment of cancer with radiation therapies or genotoxic chemotherapies. More recently, protein components of the DDR systems have been identified as promising avenues for targeted cancer therapeutics. Here, we present an in-depth analysis of the function, role in cancer and therapeutic potential of 450 expert-curated human DDR genes. We discuss the DDR drugs that have been approved by the US Food and Drug Administration (FDA) or that are under clinical investigation. We examine large-scale genomic and expression data for 15 cancers to identify deregulated components of the DDR, and we apply systematic computational analysis to identify DDR proteins that are amenable to modulation by small molecules, highlighting potential novel therapeutic targets.

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

Figure 1: DNA damage proteins with known small-molecule modulators.
Figure 2: A network view of the DNA damage response.
Figure 3: Functional annotation of the DNA damage response pathways.
Figure 4: Pathway-based disruption diagrams for individual cancer types.
Figure 5: Predicted human synthetic lethality or sensitivity within the DNA damage response.

Similar content being viewed by others

References

  1. Friedberg, E. C. et al. DNA repair: from molecular mechanism to human disease. DNA Repair 5, 986–996 (2006).

    Article  CAS  PubMed  Google Scholar 

  2. Harper, J. W. & Elledge, S. J. The DNA damage response: ten years after. Mol. Cell 28, 739–745 (2007).

    Article  CAS  PubMed  Google Scholar 

  3. Vogelstein, B. et al. Cancer genome landscapes. Science 339, 1546–1558 (2013). This paper describes mutational patterns in different cancer types and the 20:20 rule for discriminating between tumour suppressors and oncogenes.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Forbes, S. A. et al. COSMIC: mining complete cancer genomes in the Catalogue of Somatic Mutations in Cancer. Nucleic Acids Res. 39, D945–D950 (2011). This paper descibes the COSMIC database, which is a repository of publically reported somatic mutations.

    Article  CAS  PubMed  Google Scholar 

  5. Chang, K. et al. The Cancer Genome Atlas Pan-Cancer analysis project. Nature Genet. 45, 1113–1120 (2013).

    Article  CAS  Google Scholar 

  6. Barber, L. J. et al. Secondary mutations in BRCA2 associated with clinical resistance to a PARP inhibitor. J. Pathol. 229, 422–429 (2013).

    Article  CAS  PubMed  Google Scholar 

  7. Mereniuk, T. R. et al. Synthetic lethal targeting of PTEN-deficient cancer cells using selective disruption of polynucleotide kinase/phosphatase. Mol. Cancer Ther. 12, 2135–2144 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Brough, R. et al. APRIN is a cell cycle specific BRCA2-interacting protein required for genome integrity and a predictor of outcome after chemotherapy in breast cancer. EMBO J. 31, 1160–1176 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Bartkova, J. et al. Oncogene-induced senescence is part of the tumorigenesis barrier imposed by DNA damage checkpoints. Nature 444, 633–637 (2006).

    Article  CAS  PubMed  Google Scholar 

  10. Halazonetis, T. D., Gorgoulis, V. G. & Bartek, J. An oncogene-induced DNA damage model for cancer development. Science 319, 1352–1355 (2008).

    Article  CAS  PubMed  Google Scholar 

  11. Burrell, R. A., McGranahan, N., Bartek, J. & Swanton, C. The causes and consequences of genetic heterogeneity in cancer evolution. Nature 501, 338–345 (2013). This paper reviews how genomic instability leads to an increased mutation rate, tumour evolution and tumour heterogeneity.

    Article  CAS  PubMed  Google Scholar 

  12. Cahill, D. P., Kinzler, K. W., Vogelstein, B. & Lengauer, C. Genetic instability and darwinian selection in tumours. Trends Cell Biol. 9, M57–M60 (1999).

    Article  CAS  PubMed  Google Scholar 

  13. Lord, C. J. & Ashworth, A. The DNA damage response and cancer therapy. Nature 481, 287–294 (2012).

    Article  CAS  PubMed  Google Scholar 

  14. Bouwman, P. & Jonkers, J. The effects of deregulated DNA damage signalling on cancer chemotherapy response and resistance. Nature Rev. Cancer 12, 587–598 (2012).

    Article  CAS  Google Scholar 

  15. Curtin, N. J. DNA repair dysregulation from cancer driver to therapeutic target. Nature Rev. Cancer 12, 801–817 (2012).

    Article  CAS  Google Scholar 

  16. Helleday, T., Petermann, E., Lundin, C., Hodgson, B. & Sharma, R. A. DNA repair pathways as targets for cancer therapy. Nature Rev. Cancer 8, 193–204 (2008).

    Article  CAS  Google Scholar 

  17. Zhang, J., Stevens, M. F. & Bradshaw, T. D. Temozolomide: mechanisms of action, repair and resistance. Curr. Mol. Pharmacol. 5, 102–114 (2012).

    Article  CAS  PubMed  Google Scholar 

  18. Galluzzi, L. et al. Molecular mechanisms of cisplatin resistance. Oncogene 31, 1869–1883 (2012).

    Article  CAS  PubMed  Google Scholar 

  19. Liu, L. V. et al. Definition of the intermediates and mechanism of the anticancer drug bleomycin using nuclear resonance vibrational spectroscopy and related methods. Proc. Natl Acad. Sci. USA 107, 22419–22424 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Brough, R., Frankum, J. R., Costa-Cabral, S., Lord, C. J. & Ashworth, A. Searching for synthetic lethality in cancer. Curr. Opin. Genet. Dev. 21, 34–41 (2011).

    Article  CAS  PubMed  Google Scholar 

  21. Kaelin, W. G. Jr. The concept of synthetic lethality in the context of anticancer therapy. Nature Rev. Cancer 5, 689–698 (2005). References 20 and 21 describe the concept of synthetic lethalityand its utility in identifying novel drug targets.

    Article  CAS  Google Scholar 

  22. TCGA. Integrated genomic analyses of ovarian carcinoma. Nature 474, 609–615 (2011).

  23. Lord, C. J., Tutt, A. N. & Ashworth, A. Synthetic lethality and cancer therapy: lessons learned from the development of PARP inhibitors. Annu. Rev. Med. http://dx.doi.org/10.1146/annurev-med-050913-022545 (2014).

  24. Janne, P. A., Gray, N. & Settleman, J. Factors underlying sensitivity of cancers to small-molecule kinase inhibitors. Nature Rev. Drug Discov. 8, 709–723 (2009).

    Article  CAS  Google Scholar 

  25. Futreal, P. A. et al. A census of human cancer genes. Nature Rev. Cancer 4, 177–183 (2004).

    Article  CAS  Google Scholar 

  26. Greenman, C. et al. Patterns of somatic mutation in human cancer genomes. Nature 446, 153–158 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Kanehisa, M. et al. Data, information, knowledge and principle: back to metabolism in KEGG. Nucleic Acids Res. 42, D199–D205 (2014).

    Article  CAS  PubMed  Google Scholar 

  28. Croft, D. et al. Reactome: a database of reactions, pathways and biological processes. Nucleic Acids Res. 39, D691–D697 (2011).

    Article  CAS  PubMed  Google Scholar 

  29. Franceschini, A. et al. STRING v9.1: protein-protein interaction networks, with increased coverage and integration. Nucleic Acids Res. 41, D808–D815 (2013).

    Article  CAS  PubMed  Google Scholar 

  30. Ashburner, M. et al. Gene ontology: tool for the unification of biology. The Gene Ontology Consortium. Nature Genet. 25, 25–29 (2000).

    Article  CAS  PubMed  Google Scholar 

  31. Nakazawa, Y. et al. Mutations in UVSSA cause UV-sensitive syndrome and impair RNA polymerase IIo processing in transcription-coupled nucleotide-excision repair. Nature Genet. 44, 586–592 (2012).

    Article  CAS  PubMed  Google Scholar 

  32. Buisson, R. et al. Breast cancer proteins PALB2 and BRCA2 stimulate polymerase eta in recombination-associated DNA synthesis at blocked replication forks. Cell Rep. 6, 553–564 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Overington, J. P., Al-Lazikani, B. & Hopkins, A. L. How many drug targets are there? Nature Rev. Drug Discov. 5, 993–996 (2006).

    Article  CAS  Google Scholar 

  34. Patel, M. N., Halling-Brown, M. D., Tym, J. E., Workman, P. & Al-Lazikani, B. Objective assessment of cancer genes for drug discovery. Nature Rev. Drug Discov. 12, 35–50 (2013).

    Article  CAS  Google Scholar 

  35. Workman, P., Al-Lazikani, B. & Clarke, P. A. Genome-based cancer therapeutics: targets, kinase drug resistance and future strategies for precision oncology. Curr. Opin. Pharmacol. 13, 486–496 (2013).

    Article  CAS  PubMed  Google Scholar 

  36. Antoniou, A. C. et al. The BOADICEA model of genetic susceptibility to breast and ovarian cancers: updates and extensions. Br. J. Cancer 98, 1457–1466 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Kanchi, K. L. et al. Integrated analysis of germline and somatic variants in ovarian cancer. Nature Commun. 5, 3156 (2014).

    Article  CAS  Google Scholar 

  38. Bancroft, E. K. et al. Targeted prostate cancer screening in BRCA1 and BRCA2 mutation carriers: results from the initial screening round of the IMPACT study. Eur. Urol. 66, 489–499 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Tischkowitz, M. et al. Analysis of the gene coding for the BRCA2-interacting protein PALB2 in hereditary prostate cancer. Prostate 68, 675–678 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Rustgi, A. K. Familial pancreatic cancer: genetic advances. Genes Dev. 28, 1–7 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Howlett, N. G. et al. Biallelic inactivation of BRCA2 in Fanconi anemia. Science 297, 606–609 (2002).

    Article  CAS  PubMed  Google Scholar 

  42. McKusick, V. A. A. Catalog of Human Genes and Genetic Disorders (John Hopkins Univ. Press, 1998).

  43. Richardson, C. J. et al. MoKCa database—mutations of kinases in cancer. Nucleic Acids Res. 37, D824–D831 (2009).

    Article  CAS  PubMed  Google Scholar 

  44. Espinosa, O., Mitsopoulos, K., Hakas, J., Pearl, F. & Zvelebil, M. Deriving a mutation index of carcinogenicity using protein structure and protein interfaces. PLoS ONE 9, e84598 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Khoo, K. H., Verma, C. S. & Lane, D. P. Drugging the p53 pathway: understanding the route to clinical efficacy. Nature Rev. Drug Discov. 13, 217–236 (2014). This review describes the different therapeutic approaches for restoring function to p53.

    Article  CAS  Google Scholar 

  46. Weinstein, J. N. et al. The Cancer Genome Atlas Pan-Cancer analysis project. Nature Genet. 45, 1113–1120 (2013).

    Article  CAS  PubMed  Google Scholar 

  47. Collins, F. S. & Barker, A. D. Mapping the cancer genome. Pinpointing the genes involved in cancer will help chart a new course across the complex landscape of human malignancies. Sci. Am. 296, 50–57 (2007).

    Article  CAS  PubMed  Google Scholar 

  48. Kuhn, E. et al. Identification of molecular pathway aberrations in uterine serous carcinoma by genome-wide analyses. J. Natl Cancer Inst. 104, 1503–1513 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Alexandrov, L. B. et al. Signatures of mutational processes in human cancer. Nature 500, 415–421 (2013). This paper describes patterns of mutations termed 'mutational signatures'. The authors describe the aetiology of these signatures and how they vary between cancer type.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Pfeifer, G. P. et al. Tobacco smoke carcinogens, DNA damage and p53 mutations in smoking-associated cancers. Oncogene 21, 7435–7451 (2002).

    Article  CAS  PubMed  Google Scholar 

  51. Alexandrov, L. B. & Stratton, M. R. Mutational signatures: the patterns of somatic mutations hidden in cancer genomes. Curr. Opin. Genet. Dev. 24, 52–60 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. TCGA. Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature 455, 1061–1068 (2008).

  53. Parsons, D. W. et al. An integrated genomic analysis of human glioblastoma multiforme. Science 321, 1807–1812 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Lindstrom, M. S. NPM1/B23: a multifunctional chaperone in ribosome biogenesis and chromatin remodeling. Biochem. Res. Int. 2011, 195209 (2011).

    Article  CAS  PubMed  Google Scholar 

  55. Nikolaev, S. I. et al. A single-nucleotide substitution mutator phenotype revealed by exome sequencing of human colon adenomas. Cancer Res. 72, 6279–6289 (2012).

    Article  CAS  PubMed  Google Scholar 

  56. Hoe, K. K., Verma, C. S. & Lane, D. P. Drugging the p53 pathway: understanding the route to clinical efficacy. Nature Rev. Drug Discov. 13, 217–236 (2014).

    Article  CAS  Google Scholar 

  57. Riffell, J. L., Lord, C. J. & Ashworth, A. Tankyrase-targeted therapeutics: expanding opportunities in the PARP family. Nature Rev. Drug Discov. 11, 923–936 (2012).

    Article  CAS  Google Scholar 

  58. Martin, S. A. et al. DNA polymerases as potential therapeutic targets for cancers deficient in the DNA mismatch repair proteins MSH2 or MLH1. Cancer Cell 17, 235–248 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. van Pel, D. M., Stirling, P. C., Minaker, S. W., Sipahimalani, P. & Hieter, P. Saccharomyces cerevisiae genetics predicts candidate therapeutic genetic interactions at the mammalian replication fork. G3 (Bethesda) 3, 273–282 (2013).

    Article  CAS  Google Scholar 

  60. Chatr-Aryamontri, A. et al. The BioGRID interaction database: 2013 update. Nucleic Acids Res. 41, D816–D823 (2013).

    Article  CAS  PubMed  Google Scholar 

  61. Paul, J. M., Templeton, S. D., Baharani, A., Freywald, A. & Vizeacoumar, F. J. Building high-resolution synthetic lethal networks: a 'Google map' of the cancer cell. Trends Mol. Med. 20, 704–715 (2014).

    Article  CAS  PubMed  Google Scholar 

  62. Franchitto, A. et al. The mammalian mismatch repair protein MSH2 is required for correct MRE11 and RAD51 relocalization and for efficient cell cycle arrest induced by ionizing radiation in G2 phase. Oncogene 22, 2110–2120 (2003).

    Article  CAS  PubMed  Google Scholar 

  63. Bulusu, K. C., Tym, J. E., Coker, E. A. & Schierz, A. C. & Al-Lazikani, B. CanSAR: updated cancer research and drug discovery knowledgebase. Nucleic Acids Res. 42, D1040–D1047 (2014).

    Article  CAS  PubMed  Google Scholar 

  64. Gaulton, A. et al. ChEMBL: a large-scale bioactivity database for drug discovery. Nucleic Acids Res. 40, D1100–D1107 (2012).

    Article  CAS  PubMed  Google Scholar 

  65. Lipinski, C. A. Drug-like properties and the causes of poor solubility and poor permeability. J. Pharmacol. Toxicol. Methods 44, 235–249 (2000). This paper describes the properties required for a molecule to be a good drug. It presents the rule of five.

    Article  CAS  PubMed  Google Scholar 

  66. Fauman, E. B., Rai, B. K. & Huang, E. S. Structure-based druggability assessment—identifying suitable targets for small molecule therapeutics. Curr. Opin. Chem. Biol. 15, 463–468 (2011).

    Article  CAS  PubMed  Google Scholar 

  67. Mehra, R., Serebriiskii, I. G., Burtness, B., Astsaturov, I. & Golemis, E. A. Aurora kinases in head and neck cancer. Lancet Oncol. 14, e425–e435 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Weiss, L. & Efferth, T. Polo-like kinase 1 as target for cancer therapy. Exp. Hematol. Oncol. 1, 38 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Hornbeck, P. V. et al. PhosphoSitePlus: a comprehensive resource for investigating the structure and function of experimentally determined post-translational modifications in man and mouse. Nucleic Acids Res. 40, D261–D270 (2012).

    Article  CAS  PubMed  Google Scholar 

  70. Landre, V., Rotblat, B., Melino, S., Bernassola, F. & Melino, G. Screening for E3-Ubiquitin ligase inhibitors: challenges and opportunities. Oncotarget 5, 7988–8013 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  71. Jacq, X., Kemp, M., Martin, N. M. & Jackson, S. P. Deubiquitylating enzymes and DNA damage response pathways. Cell Biochem. Biophys. 67, 25–43 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Lim, K. H. & Baek, K. H. Deubiquitinating enzymes as therapeutic targets in cancer. Curr. Pharm. Des. 19, 4039–4052 (2013).

    Article  CAS  PubMed  Google Scholar 

  73. Liang, Q. et al. A selective USP1-UAF1 inhibitor links deubiquitination to DNA damage responses. Nature Chem. Biol. 10, 298–304 (2014).

    Article  CAS  Google Scholar 

  74. Fan, Y. H. et al. USP7 inhibitor P22077 inhibits neuroblastoma growth via inducing p53-mediated apoptosis. Cell Death Dis. 4, e867 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Nishi, R. et al. Systematic characterization of deubiquitylating enzymes for roles in maintaining genome integrity. Nature Cell Biol. 16, 1016–1026 (2014). This paper describes a systematic screen of DUBs to identify their involvement in the DDR.

    Article  CAS  PubMed  Google Scholar 

  76. Tomkinson, A. E., Howes, T. R. & Wiest, N. E. DNA ligases as therapeutic targets. Transl. Cancer Res. 2, 1219 (2013).

    PubMed  Google Scholar 

  77. van Pel, D. M. et al. An evolutionarily conserved synthetic lethal interaction network identifies FEN1 as a broad-spectrum target for anticancer therapeutic development. PLoS Genet. 9, e1003254 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Panda, H. et al. Amino acid Asp181 of 5′-flap endonuclease 1 is a useful target for chemotherapeutic development. Biochemistry 48, 9952–9958 (2009).

    Article  CAS  PubMed  Google Scholar 

  79. Dorjsuren, D. et al. Complementary non- radioactive assays for investigation of human flap endonuclease 1 activity. Nucleic Acids Res. 39, e11 (2011).

    Article  CAS  PubMed  Google Scholar 

  80. McWhirter, C. et al. Development of a high-throughput fluorescence polarization DNA cleavage assay for the identification of FEN1 inhibitors. J. Biomol. Screen 18, 567–575 (2013).

    Article  CAS  PubMed  Google Scholar 

  81. Nguyen, G. H. et al. A small molecule inhibitor of the BLM helicase modulates chromosome stability in human cells. Chem. Biol. 20, 55–62 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Aggarwal, M., Banerjee, T., Sommers, J. A. & Brosh, R. M. Jr. Targeting an Achilles' heel of cancer with a WRN helicase inhibitor. Cell Cycle 12, 3329–3335 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. D'Antonio, M. et al. Recessive cancer genes engage in negative genetic interactions with their functional paralogs. Cell Rep. 5, 1519–1526 (2013).

    Article  CAS  PubMed  Google Scholar 

  84. Turner, N. C. et al. A synthetic lethal siRNA screen identifying genes mediating sensitivity to a PARP inhibitor. EMBO J. 27, 1368–1377 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Allinson, S. L. DNA end-processing enzyme polynucleotide kinase as a potential target in the treatment of cancer. Future Oncol. 6, 1031–1042 (2010).

    Article  CAS  PubMed  Google Scholar 

  86. Oliver, A. W., Swift, S., Lord, C. J., Ashworth, A. & Pearl, L. H. Structural basis for recruitment of BRCA2 by PALB2. EMBO Rep. 10, 990–996 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Bastian, M., Heymann, S. & Jacomy, M. Gephi: an open source software for exploring and manipulating networks International AAAI Conference on Weblogs and Social Media (2009).

    Google Scholar 

  88. TCGA. Comprehensive molecular characterization of human colon and rectal cancer. Nature 487, 330–337 (2012).

  89. TCGA. Comprehensive genomic characterization of squamous cell lung cancers. Nature 489, 519–525 (2012).

  90. TCGA. Comprehensive molecular portraits of human breast tumours. Nature 490, 61–70 (2012).

  91. TCGA. Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N. Engl. J. Med. 368, 2059–2074 (2013).

  92. TCGA. Comprehensive molecular characterization of clear cell renal cell carcinoma. Nature 499, 43–49 (2013).

  93. TCGA. Comprehensive molecular characterization of urothelial bladder carcinoma. Nature 507, 315–322 (2014).

  94. Smoot, M. E., Ono, K., Ruscheinski, J., Wang, P. L. & Ideker, T. Cytoscape 2.8: new features for data integration and network visualization. Bioinformatics 27, 431–432 (2011).

    Article  CAS  PubMed  Google Scholar 

  95. Kupfer, G. M. Fanconi anemia: a signal transduction and DNA repair pathway. Yale J. Biol. Med. 86, 491–497 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  96. Daley, J. M., Kwon, Y., Niu, H. & Sung, P. Investigations of homologous recombination pathways and their regulation. Yale J. Biol. Med. 86, 453–461 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  97. Murray, J. M. & Carr, A. M. Smc5/6: a link between DNA repair and unidirectional replication? Nature Rev. Mol. Cell Biol. 9, 177–182 (2008).

    Article  CAS  Google Scholar 

  98. Pastwa, E. & Blasiak, J. Non-homologous DNA end joining. Acta Biochim. Pol. 50, 891–908 (2003).

    CAS  PubMed  Google Scholar 

  99. Wang, Z., Wu, X. & Friedberg, E. C. Molecular mechanism of base excision repair of uracil-containing DNA in yeast cell-free extracts. J. Biol. Chem. 272, 24064–24071 (1997).

    Article  CAS  PubMed  Google Scholar 

  100. Eker, A. P., Quayle, C., Chaves, I. & van der Horst, G. T. DNA repair in mammalian cells: Direct DNA damage reversal: elegant solutions for nasty problems. Cell. Mol. Life Sci. 66, 968–980 (2009).

    Article  CAS  PubMed  Google Scholar 

  101. Larrea, A. A., Lujan, S. A. & Kunkel, T. A. SnapShot: DNA mismatch repair. Cell 141, 730.e1 (2010).

    Article  PubMed  Google Scholar 

  102. Jaspers, N. G. & Hoeijmakers, J. H. DNA repair. Nucleotide excision-repair in the test tube. Curr. Biol. 5, 700–702 (1995).

    Article  CAS  PubMed  Google Scholar 

  103. Prakash, S., Johnson, R. E. & Prakash, L. Eukaryotic translesion synthesis DNA polymerases: specificity of structure and function. Annu. Rev. Biochem. 74, 317–353 (2005).

    Article  CAS  PubMed  Google Scholar 

  104. Chambers, A. L. & Downs, J. A. The RSC and INO80 chromatin-remodeling complexes in DNA double-strand break repair. Prog. Mol. Biol. Transl. Sci. 110, 229–261 (2012).

    Article  CAS  PubMed  Google Scholar 

  105. Verdun, R. E. & Karlseder, J. Replication and protection of telomeres. Nature 447, 924–931 (2007).

    Article  CAS  PubMed  Google Scholar 

  106. Bardin, A. J. & Amon, A. Men and sin: what's the difference? Nature Rev. Mol. Cell Biol. 2, 815–826 (2001).

    Article  CAS  Google Scholar 

  107. Jackson, S. P. & Durocher, D. Regulation of DNA damage responses by ubiquitin and SUMO. Mol. Cell 49, 795–807 (2013).

    Article  CAS  PubMed  Google Scholar 

  108. Vogelstein, B., Lane, D. & Levine, A. J. Surfing the p53 network. Nature 408, 307–310 (2000).

    Article  CAS  PubMed  Google Scholar 

  109. Marston, A. L. Chromosome segregation in budding yeast: sister chromatid cohesion and related mechanisms. Genetics 196, 31–63 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors would like to thank the following principal investigators from the Sussex Genome Damage and Stability Centre for help with curating the DDR gene list and for providing deep insights into the pathways and networks that make up the DDR: A. Carr, J. Murray, J. Downs, E. Hoffmann, A. Lehmann, P. Jeggo, A. Bianchi, J. Baxter, K. Caldecott and V. Savic. The authors would also like to thank C. Richardson and J. Tym for technical assistance, A. Eyre-Walker and C. Parry-Morris for useful discussions, and A. Carr, P. Beswick and P. Workman for critical reading of the manuscript. This work was supported by a Daphne Jackson Fellowship funded by the Medical Research Council (F.M.G.P.); Cancer Research UK core funding to the Cancer Therapeutics Unit at the Institute of Cancer Research in London C309/A11566 (B.A.-L.); and a Cancer Research UK Programme Grant C302/A14532 (L.H.P.). Author contributions: L.H.P., S.E.W., B.A.-L. and F.M.G.P. conceived the project; F.M.G.P. and B.A.-L. designed the analysis; A.C.S., B.A.-L. and F.M.G.P. performed the data analysis and informatics; and L.H.P., B.A.-L. and F.M.G.P. wrote the paper.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Bissan Al-Lazikani or Frances M. G. Pearl.

Ethics declarations

Competing interests

B.A.-L. is an employee of The Institute of Cancer Research (ICR), which has a commercial interest in inhibitors in oncology targets, and operates a 'Rewards to Inventors' scheme. L.H.P. is a director and shareholder in Domainex Limited a biotechnology company, developing novel drugs for a range of human diseases. S.E.W. is a consultant for BioCrea GmbH.

Related links

PowerPoint slides

Supplementary information

Supplementary information S1 (table)

Current DDR targets with drugs or compounds in clinical trials (PDF 182 kb)

Supplementary information S2 (box)

Methods (PDF 348 kb)

Supplementary information S3 (table)

Hierarchical classification of DDR genes (PDF 364 kb)

Supplementary information S4 (figure)

(ZIP 194 kb)

Supplementary information S5 (table)

Genes within the DDR implicated in genetic diseases (PDF 250 kb)

Supplementary information S6 (table)

Genes predicted to be oncogenes or tumour suppressors according to the 20:20 rule (PDF 105 kb)

Supplementary information S7 (table)

Mutational frequency per disease (PDF 143 kb)

Supplementary information S8 (table)

Pathway disruption of the DDR in 15 TCGA cancers (PDF 271 kb)

Supplementary information S9 (figure)

(ZIP 27 kb)

Supplementary information S10 (table)

SSL predictions (PDF 1367 kb)

Supplementary information S11 (table)

Targets with small molecule modulators (PDF 121 kb)

Supplementary information S12 (table)

Ligand druggability predictions (PDF 154 kb)

Supplementary information S13 (table)

Structure druggability predictions (PDF 322 kb)

Supplementary information S14 (table)

Network druggability predictions (PDF 315 kb)

Supplementary information S15 (figure)

(ZIP 182 kb)

Supplementary information S16 (table)

Kinases that regulate DDR proteins (PDF 402 kb)

Supplementary information S17 (table)

DDR proteins with reported ubiquitination sites (PDF 1569 kb)

Supplementary information S18 (table)

E3 ligases with reported interactions with DDR proteins (PDF 287 kb)

Supplementary information S19 (table)

DUBs reported to interact with DDR proteins (PDF 232 kb)

Supplementary information S20 (table)

Duggability predictions for best pathway-based candidates (PDF 156 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Pearl, L., Schierz, A., Ward, S. et al. Therapeutic opportunities within the DNA damage response. Nat Rev Cancer 15, 166–180 (2015). https://doi.org/10.1038/nrc3891

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrc3891

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