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:

A panoply of errors: polymerase proofreading domain mutations in cancer

Key Points

  • The proofreading exonuclease domains of the replicative DNA polymerases Pol δ and Pol ε perform an essential function in ensuring accurate DNA replication by proofreading and removing mispaired bases from the newly synthesized DNA strand.

  • Recent studies have shown that mutations in the proofreading domains of POLD1 and POLE (which encode the catalytic subunits of Pol δ and Pol ε, respectively, in humans) predispose to colonic polyposis and cancer, and that somatic POLE proofreading domain mutations occur in several tumour types, most commonly those of the endometrium and colorectum. Interestingly, somatic POLD1 proofreading domain mutations seem to be uncommon.

  • In several cases, the pathogenicity of these germline and somatic DNA polymerase proofreading domain mutations has been supported by studies using cell-free assays and Saccharomyces cerevisiae mutants, which confirm that they reduce or abolish exonuclease activity and increase the mutation rate.

  • Consistent with these studies, the most striking feature of tumours with somatic POLE proofreading domain mutations is their exceptional burden of base substitution mutations — 'ultramutation'. Other notable features are their characteristic mutation spectrum, with overrepresentation of C→A transversions and, in general, a strong tendency to microsatellite stability.

  • Endometrial cancers with somatic POLE proofreading domain mutations have an excellent prognosis, which may be because their ultramutation causes an abundance of antigenic neoepitopes, which in turn stimulate a potent antitumour immune response. The prognostic and immunological consequences of somatic POLE proofreading domain mutations in other tumour types await definition.

  • Future studies of DNA polymerase proofreading domain mutations in cancer may provide further insights into the mechanisms and consequences of a mutator phenotype in cancer, and help to improve care for patients with endometrial, colorectal and other cancers.

Abstract

Although it has long been recognized that the exonucleolytic proofreading activity intrinsic to the replicative DNA polymerases Pol δ and Pol ε is essential for faithful replication of DNA, evidence that defective DNA polymerase proofreading contributes to human malignancy has been limited. However, recent studies have shown that germline mutations in the proofreading domains of Pol δ and Pol ε predispose to cancer, and that somatic Pol ε proofreading domain mutations occur in multiple sporadic tumours, where they underlie a phenotype of 'ultramutation' and favourable prognosis. In this Review, we summarize the current understanding of the mechanisms and consequences of polymerase proofreading domain mutations in human malignancies, and highlight the potential utility of these variants as novel cancer biomarkers and 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: Frequency and location of germline and somatic Pol δ and Pol ε proofreading exonuclease domain mutations in cancers.

Similar content being viewed by others

References

  1. Loeb, L. A. Mutator phenotype may be required for multistage carcinogenesis. Cancer Res. 51, 3075–3079 (1991).

    CAS  PubMed  Google Scholar 

  2. Kunkel, T. A. DNA replication fidelity. J. Biol. Chem. 279, 16895–16898 (2004).

    Article  CAS  PubMed  Google Scholar 

  3. Loeb, L. A. & Monnat, R. J. DNA polymerases and human disease. Nat. Rev. Genet. 9, 594–604 (2008).

    Article  CAS  PubMed  Google Scholar 

  4. Morrison, A., Johnson, A. L., Johnston, L. H. & Sugino, A. Pathway correcting DNA replication errors in Saccharomyces cerevisiae. EMBO J. 12, 1467–1473 (1993).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Edelmann, W. et al. Mutation in the mismatch repair gene Msh6 causes cancer susceptibility. Cell 91, 467–477 (1997).

    Article  CAS  PubMed  Google Scholar 

  6. Albertson, T. M. et al. DNA polymerase ε and δ proofreading suppress discrete mutator and cancer phenotypes in mice. Proc. Natl Acad. Sci. USA 106, 17101–17104 (2009). Demonstration that although both Pole and Pold1 exonuclease-null mice develop tumours, the tumour spectrum differs between the two mutant alleles.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Goldsby, R. E. et al. High incidence of epithelial cancers in mice deficient for DNA polymerase δ proofreading. Proc. Natl Acad. Sci. USA 99, 15560–15565 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Goldsby, R. E. et al. Defective DNA polymerase-δ proofreading causes cancer susceptibility in mice. Nat. Med. 7, 638–639 (2001).

    Article  CAS  PubMed  Google Scholar 

  9. Fishel, R. et al. The human mutator gene homolog MSH2 and its association with hereditary nonpolyposis colon cancer. Cell 75, 1027–1038 (1993).

    Article  CAS  PubMed  Google Scholar 

  10. Aaltonen, L. A. et al. Clues to the pathogenesis of familial colorectal cancer. Science 260, 812–816 (1993).

    Article  CAS  PubMed  Google Scholar 

  11. Yoshida, R. et al. Concurrent genetic alterations in DNA polymerase proofreading and mismatch repair in human colorectal cancer. Eur. J. Hum. Genet. 19, 320–325 (2011).

    Article  PubMed  Google Scholar 

  12. Palles, C. et al. Germline mutations affecting the proofreading domains of POLE and POLD1 predispose to colorectal adenomas and carcinomas. Nat. Genet. 45, 136–144 (2012). Identification of germline POLE and POLD1 proofreading domain mutations in patients with intestinal polyposis and CRC.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. The Cancer Genome Atlas Network. Comprehensive molecular characterization of human colon and rectal cancer. Nature 487, 330–337 (2012).

  14. Erson-Omay, E. Z. et al. Somatic POLE mutations cause an ultramutated giant cell high-grade glioma subtype with better prognosis. Neuro Oncol. 17, 1356–1364 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Church, D. N. et al. DNA polymerase ε and δ exonuclease domain mutations in endometrial cancer. Hum. Mol. Genet. 22, 2820–2828 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Seshagiri, S. et al. Recurrent R-spondin fusions in colon cancer. Nature 488, 660–664 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. The Cancer Genome Atlas Network. Integrated genomic characterization of endometrial carcinoma. Nature 497, 67–73 (2013). Demonstration that ECs with somatic POLE proofreading domain mutations are ultramutated and may have a favourable prognosis.

  18. Shinbrot, E. et al. Exonuclease mutations in DNA polymerase ε reveal replication strand specific mutation patterns and human origins of replication. Genome Res. 24, 1740–1750 (2014). Comprehensive analysis of TCGA sequencing data to identify pathogenic somatic polymerase proofreading domain mutations across multiple tumour types.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Alexandrov, L. B. et al. Signatures of mutational processes in human cancer. Nature 500, 415–421 (2013). Pan-cancer analysis demonstrating that the POLE proofreading domain-mutant mutational signature is present in 0.5% of tumours.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Church, D. N. et al. Prognostic significance of POLE proofreading mutations in endometrial cancer. J. Natl Cancer Inst. 107, 1–8 (2015).

    Article  CAS  Google Scholar 

  21. Billingsley, C. C. et al. Polymerase ε (POLE) mutations in endometrial cancer: clinical outcomes and implications for Lynch syndrome testing. Cancer 121, 386–394 (2014).

    Article  CAS  PubMed  Google Scholar 

  22. Stelloo, E. et al. Refining prognosis and identifying targetable pathways for high-risk endometrial cancer; a TransPORTEC initiative. Mod. Pathol. 28, 836–844 (2015).

    Article  CAS  PubMed  Google Scholar 

  23. Talhouk, A. et al. A clinically applicable molecular-based classification for endometrial cancers. Br. J. Cancer 113, 299–310 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Meng, B. et al. POLE exonuclease domain mutation predicts long progression-free survival in grade 3 endometrioid carcinoma of the endometrium. Gynecol. Oncol. 134, 15–19 (2014).

    Article  CAS  PubMed  Google Scholar 

  25. van Gool, I. C. et al. POLE proofreading mutations elicit an anti-tumor immune response in endometrial cancer. Clin. Cancer Res. 21, 3347–3355 (2015). First demonstration that somatic POLE proofreading domain mutations are associated with a cytotoxic T cell response.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Howitt, B. E. et al. Association of polymerase e-mutated and microsatellite-instable endometrial cancers with neoantigen load, number of tumor-infiltrating lymphocytes, and expression of PD-1 and PD-L1. JAMA Oncol. 1, 1319–1323 (2015).

    Article  PubMed  Google Scholar 

  27. Henninger, E. E. & Pursell, Z. F. DNA polymerase ε and its roles in genome stability. IUBMB Life 66, 339–351 (2014).

    Article  CAS  PubMed  Google Scholar 

  28. Prindle, M. J. & Loeb, L. A. DNA polymerase delta in DNA replication and genome maintenance. Env. Mol. Mutag. 53, 666–682 (2012).

    Article  CAS  Google Scholar 

  29. Pavlov, Y. I. & Shcherbakova, P. V. DNA polymerases at the eukaryotic fork—20 years later. Mutat. Res. 685, 45–53 (2010).

    Article  CAS  PubMed  Google Scholar 

  30. Johansson, E. & Dixon, N. Replicative DNA polymerases. Cold Spring Harb. Perspect. Biol. 5, a012799 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Morrison, A., Araki, H., Clark, A. B., Hamatake, R. K. & Sugino, A. A third essential DNA polymerase in S. cerevisiae. Cell 62, 1143–1151 (1990).

    Article  CAS  PubMed  Google Scholar 

  32. Budd, M. & Campbell, J. L. Temperature-sensitive mutations in the yeast DNA polymerase I gene. Proc. Natl Acad. Sci. USA 84, 2838–2842 (1987).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Budd, M. E. & Campbell, J. L. DNA polymerases δ and ε are required for chromosomal replication in Saccharomyces cerevisiae. Mol. Cell. Biol. 13, 496–505 (1993).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Francesconi, S., Park, H. & Wang, T. S. Fission yeast with DNA polymerase δ temperature-sensitive alleles exhibits cell division cycle phenotype. Nucleic Acids Res. 21, 3821–3828 (1993).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Bernad, A., Blanco, L., Lázaro, J., Martín, G. & Salas, M. A conserved 3′→5′ exonuclease active site in prokaryotic and eukaryotic DNA polymerases. Cell 59, 219–228 (1989).

    Article  CAS  PubMed  Google Scholar 

  36. Shevelev, I. V. & Hübscher, U. The 3′ 5′ exonucleases. Nat. Rev. Mol. Cell Biol. 3, 364–376 (2002).

    Article  CAS  PubMed  Google Scholar 

  37. Baranovskiy, A. G. et al. X-ray structure of the complex of regulatory subunits of human DNA polymerase δ. Cell Cycle 7, 3026–3036 (2008).

    Article  CAS  PubMed  Google Scholar 

  38. Podust, V. N., Chang, L. S., Ott, R., Dianov, G. L. & Fanning, E. Reconstitution of human DNA polymerase δ using recombinant baculoviruses: the p12 subunit potentiates DNA polymerizing activity of the four-subunit enzyme. J. Biol. Chem. 277, 3894–3901 (2002).

    Article  CAS  PubMed  Google Scholar 

  39. Zhou, Y., Meng, X., Zhang, S., Lee, E. Y. & Lee, M. Y. Characterization of human DNA polymerase δ and its subassemblies reconstituted by expression in the MultiBac system. PLoS ONE 7, e319156 (2012).

    Google Scholar 

  40. Garbacz, M. et al. Fidelity consequences of the impaired interaction between DNA polymerase ε and the GINS complex. DNA Repair 29, 23–35 (2015).

    Article  CAS  PubMed  Google Scholar 

  41. Langston, L. D. et al. CMG helicase and DNA polymerase ε form a functional 15-subunit holoenzyme for eukaryotic leading-strand DNA replication. Proc. Natl Acad. Sci. USA 111, 15390–15395 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Sengupta, S., van Deursen, F., de Piccoli, G. & Labib, K. Dpb2 integrates the leading-strand DNA polymerase into the eukaryotic replisome. Curr. Biol. 23, 543–552 (2013).

    Article  CAS  PubMed  Google Scholar 

  43. Aksenova, A. et al. Mismatch repair-independent increase in spontaneous mutagenesis in yeast lacking non-essential subunits of DNA polymerase ε. PLoS Genet. 6, e1001209 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Pursell, Z. F., Isoz, I., Lundström, E.-B., Johansson, E. & Kunkel, T. A. Yeast DNA polymerase ε participates in leading-strand DNA replication. Science 317, 127–130 (2007). Demonstration that Pol ε replicates the leading strand in S. cerevisiae.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. McElhinny, S. A., Gordenin, D. A., Stith, C. M., Burgers, P. M. J. & Kunkel, T. A. Division of labor at the eukaryotic replication fork. Mol. Cell 30, 137–144 (2008).

    Article  CAS  Google Scholar 

  46. Miyabe, I., Kunkel, T. A. & Carr, A. M. The major roles of DNA polymerases ε and δ at the eukaryotic replication fork are evolutionarily conserved. PLoS Genet. 7, e1002407 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Vazquez, E. & Antequera, F. Replication dynamics in fission and budding yeasts through DNA polymerase tracking. Bioessays 37, 1067–1073 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Georgescu, R. E. et al. Reconstitution of a eukaryotic replisome reveals suppression mechanisms that define leading/lagging strand operation. eLife 4, e04988 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Georgescu, R. E. et al. Mechanism of asymmetric polymerase assembly at the eukaryotic replication fork. Nat. Struct. Mol. Biol. 21, 664–670 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Hogg, M. et al. Structural basis for processive DNA synthesis by yeast DNA polymerase ε. Nat. Struct. Mol. Biol. 21, 49–55 (2014). First report of the crystal structure of the S. cerevisiae Pol ε catalytic subunit.

    Article  CAS  PubMed  Google Scholar 

  51. Johnson, Robert, E., Klassen, R., Prakash, L. & Prakash, S. A major role of DNA polymerase δ in replication of both the leading and lagging DNA strands. Mol. Cell 59, 163–175 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Blank, A., Kim, B. Fau-Loeb, L. A. & Loeb, L. A. DNA polymerase δ is required for base excision repair of DNA methylation damage in Saccharomyces cerevisiae. Proc. Natl Acad. Sci. USA 91, 9047–9051 (1994).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Stucki, M. et al. Mammalian base excision repair by DNA polymerases δ and ε. Oncogene 17, 835–843 (1998).

    Article  CAS  PubMed  Google Scholar 

  54. Nishida, C., Reinhard, P. & Linn, S. DNA repair synthesis in human fibroblasts requires DNA polymerase δ. J. Biol. Chem. 263, 501–510 (1988).

    CAS  PubMed  Google Scholar 

  55. Lehmann, A. R. DNA polymerases and repair synthesis in NER in human cells. DNA Repair 10, 730–733 (2011).

    Article  CAS  PubMed  Google Scholar 

  56. Tran, H. T., Gordenin, D. A. & Resnick, M. A. The 3′→5′ exonucleases of DNA polymerases δ and ε and the 5′→3′ exonuclease Exo1 have major roles in postreplication mutation avoidance in Saccharomyces cerevisiae. Mol. Cell. Biol. 19, 2000–2007 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Bowen, N. et al. Reconstitution of long and short patch mismatch repair reactions using Saccharomyces cerevisiae proteins. Proc. Natl Acad. Sci. USA 110, 18472–18477 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Zhang, Y. et al. Reconstitution of 5′-directed human mismatch repair in a purified system. Cell 122, 693–705 (2005).

    Article  CAS  PubMed  Google Scholar 

  59. Costantino, L. et al. Break-induced replication repair of damaged forks induces genomic duplications in human cells. Science 343, 88–91 (2014).

    Article  CAS  PubMed  Google Scholar 

  60. Lydeard, J. R., Jain, S., Yamaguchi, M. & Haber, J. E. Break-induced replication and telomerase-independent telomere maintenance require Pol32. Nature 448, 820–823 (2007).

    Article  CAS  PubMed  Google Scholar 

  61. Pursell, Z. F. & Kunkel, T. A. DNA polymerase ε: a polymerase of unusual size (and complexity). Prog. Nucleic Acid Res. Mol. Biol. 82, 101–145 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Ganai, R. a., Bylund, G. O. & Johansson, E. Switching between polymerase and exonuclease sites in DNA polymerase ε. Nucleic Acids Res. 4, 1–11 (2014).

    Google Scholar 

  63. Simon, M., Giot, L. & Faye, G. The 3′ to 5′ exonuclease activity located in the DNA polymerase delta subunit of Saccharomyces cerevisiae is required for accurate replication. EMBO J. 10, 2165–2170 (1991).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Murphy, K., Darmawan, H., Schultz, A., da Silva, E. F. & Reha-Krantz, L. J. A method to select for mutator DNA polymerase δs in Saccharomyces cerevisiae. Genome 49, 403–410 (2006).

    Article  CAS  PubMed  Google Scholar 

  65. Datta, A. et al. Checkpoint-dependent activation of mutagenic repair in Saccharomyces cerevisiae pol3-01 mutants. Mol. Cell 6, 593–603 (2000).

    Article  CAS  PubMed  Google Scholar 

  66. Williams, L. N. et al. dNTP pool levels modulate mutator phenotypes of error-prone DNA polymerase ε variants. Proc. Natl Acad. Sci. USA 112, E2457–E2466 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Bellido, F. et al. POLE and POLD1 mutations in 529 kindred with familial colorectal cancer and/or polyposis: review of reported cases and recommendations for genetic testing and surveillance. Genet. Med. https://dx.doi.org/10.1038/gim.2015.75 (2015).

  68. Spier, I. et al. Frequency and phenotypic spectrum of germline mutations in POLE and seven other polymerase genes in 266 patients with colorectal adenomas and carcinomas. Int. J. Cancer 137, 320–331 (2015).

    Article  CAS  PubMed  Google Scholar 

  69. Chubb, D. et al. Genetic diagnosis of high-penetrance susceptibility for colorectal cancer (CRC) is achievable for a high proportion of familial CRC by exome sequencing. J. Clin. Oncol. 33, 426–432 (2015).

    Article  CAS  PubMed  Google Scholar 

  70. Valle, L. et al. New insights into POLE and POLD1 germline mutations in familial colorectal cancer and polyposis. Hum. Mol. Genet. 23, 3506–3512 (2014).

    Article  CAS  PubMed  Google Scholar 

  71. Elsayed, F. A. et al. Germline variants in POLE are associated with early onset mismatch repair deficient colorectal cancer. Eur. J. Hum. Genet. 23, 1080–1084 (2015).

    Article  CAS  PubMed  Google Scholar 

  72. Rohlin, A. et al. A mutation in POLE predisposing to a multi-tumour phenotype. Int. J. Oncol. 45, 77–81 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Hansen, M. F. et al. A novel POLE mutation associated with cancers of colon, pancreas, ovaries and small intestine. Fam. Cancer 14, 437–448 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Aoude, L. G. et al. POLE mutations in families predisposed to cutaneous melanoma. Fam. Cancer 14, 621–628 (2015).

    Article  CAS  PubMed  Google Scholar 

  75. Zou, Y. et al. Frequent POLE1 p. S297F mutation in Chinese patients with ovarian endometrioid carcinoma. Mutat. Res. 761, 49–52 (2014).

    Article  CAS  PubMed  Google Scholar 

  76. Jones, S. et al. Genomic analyses of gynaecologic carcinosarcomas reveal frequent mutations in chromatin remodelling genes. Nat. Commun. 5, 5006 (2014).

    Article  CAS  PubMed  Google Scholar 

  77. Shlien, A. et al. Combined hereditary and somatic mutations of replication error repair genes result in rapid onset of ultra-hypermutated cancers. Nat. Genet. 47, 257–262 (2015). Demonstration that acquisition of pathogenic POLD1 and POLE proofreading domain mutations is associated with rapid tumour growth in patients with CMMR-D.

    Article  CAS  PubMed  Google Scholar 

  78. The Cancer Genome Atlas Network. Widespread genetic heterogeneity in multiple myeloma: implications for targeted therapy. Cancer Cell 25, 91–101 (2014).

  79. The Cancer Genome Atlas Network. Comprehensive molecular characterization of gastric adenocarcinoma. Nature 513, 202–209 (2014).

  80. Imielinski, M. et al. Mapping the hallmarks of lung adenocarcinoma with massively parallel sequencing. Cell 150, 1107–1120 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. The Cancer Genome Atlas Network. Comprehensive molecular profiling of lung adenocarcinomas. Nature 511, 543–550 (2014).

  82. Kane, D. P. & Shcherbakova, P. V. A common cancer-associated DNA polymerase ε mutation causes an exceptionally strong mutator phenotype, indicating fidelity defects distinct from loss of proofreading. Cancer Res. 74, 1895–1901 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Hussein, Y. R. et al. Clinicopathological analysis of endometrial carcinomas harboring somatic POLE exonuclease domain mutations. Mod. Pathol. 28, 505–514 (2014).

    Article  CAS  PubMed  Google Scholar 

  84. Bertagnolli, M. M. et al. Microsatellite instability and loss of heterozygosity at chromosomal location 18q: prospective evaluation of biomarkers for stages II and III colon cancer—a study of CALGB 9581 and 89803. J. Clin. Oncol. 29, 3153–3162 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  85. Hutchins, G. et al. Value of mismatch repair, KRAS, and BRAF mutations in predicting recurrence and benefits from chemotherapy in colorectal cancer. J. Clin. Oncol. 29, 1261–1270 (2011).

    Article  PubMed  Google Scholar 

  86. Nelson, G. S. et al. MMR deficiency is common in high-grade endometrioid carcinomas and is associated with an unfavorable outcome. Gynecol. Oncol. 131, 309–314 (2013).

    Article  CAS  PubMed  Google Scholar 

  87. Diaz-Padilla, I. et al. Mismatch repair status and clinical outcome in endometrial cancer: a systematic review and meta-analysis. Crit. Rev. Oncol. Hematol. 88, 154–167 (2013).

    Article  PubMed  Google Scholar 

  88. Heitzer, E. & Tomlinson, I. Replicative DNA polymerase mutations in cancer. Curr. Opin. Genet. Dev. 24, 107–113 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Williams, L. N., Herr, A. J. & Preston, B. D. Emergence of DNA polymerase ε antimutators that escape error-induced extinction in yeast. Genetics 193, 751–770 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Herr, A. J., Kennedy, S. R., Knowels, G. M., Schultz, E. M. & Preston, B. D. DNA replication error-induced extinction of diploid yeast. Genetics 196, 677–691 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Heyer, J., Yang, K., Lipkin, M., Edelmann, W. & Kucherlapati, R. Mouse models for colorectal cancer. Oncogene 18, 5325–5333 (1999).

    Article  CAS  PubMed  Google Scholar 

  92. Sole, R. V. & Deisboeck, T. S. An error catastrophe in cancer? J. Theor. Biol. 228, 47–54 (2004).

    Article  PubMed  Google Scholar 

  93. Loeb, L. A. Human cancers express mutator phenotypes: origin, consequences and targeting. Nat. Rev. Cancer 11, 450–457 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Herr, A. J. et al. Mutator suppression and escape from replication error-induced extinction in yeast. PLoS Genet. 7, e1002282 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Ghodgaonkar, M. M. et al. Phenotypic characterization of missense polymerase-δ mutations using an inducible protein-replacement system. Nat. Commun. 5, 4990–4990 (2014).

    Article  CAS  PubMed  Google Scholar 

  96. Rizvi, N. A. et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 348, 124–128 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Korona, D. A., LeCompte, K. G. & Pursell, Z. F. The high fidelity and unique error signature of human DNA polymerase. Nucleic Acids Res. 39, 1763–1773 (2011).

    Article  CAS  PubMed  Google Scholar 

  98. Schwarz, J. M., Cooper, D. N. Schuelke, M. & Seelow, D. MutationTaster2: mutation prediction for the deep-sequencing age. Nat. Methods 11, 361–362 (2014).

    Article  CAS  PubMed  Google Scholar 

  99. Kumar, P., Henikoff, S. & Ng, P. C. Predicting the effects of coding non-synonymous variants on protein function using the SIFT algorithm. Nat. Protoc. 4, 1073–1081 (2009).

    Article  CAS  PubMed  Google Scholar 

  100. Adzhubei, I. A. et al. A method and server for predicting damaging missense mutations. Nat. Methods 7, 248–249 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Fox, E. J., Prindle, M. J. & Loeb, L. A. Do mutator mutations fuel tumorigenesis? Cancer Metastasis Rev. 32, 353–361 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Llosa, N. J. et al. The vigorous immune microenvironment of microsatellite instable colon cancer is balanced by multiple counter-inhibitory checkpoints. Cancer Discov. 5, 43–51 (2015).

    Article  CAS  PubMed  Google Scholar 

  103. McFarland, C. D., Mirny, L. A. & Korolev, K. S. Tug-of-war between driver and passenger mutationsin cancer and other adaptive processes. Proc. Natl Acad. Sci. USA 111, 15138–15143 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Vasen, H. F. et al. Revised guidelines for the clinical management of Lynch syndrome (HNPCC): recommendations by a group of European experts. Gut 62, 812–823 (2013).

    Article  CAS  PubMed  Google Scholar 

  105. Hamid, O. et al. Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. N. Engl. J. Med. 369, 134–144 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Brahmer, J. R. et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N. Engl. J. Med. 366, 2455–2465 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Topalian, S. L. et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N. Engl. J. Med. 366, 2443–2454 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Snyder, A. et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N. Engl. J. Med. 371, 2189–2199 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Tumeh, P. C. et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature 515, 568–571 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Twyman-Saint Victor, C. et al. Radiation and dual checkpoint blockade activate non-redundant immune mechanisms in cancer. Nature 520, 373–377 (2015).

    Article  CAS  PubMed  Google Scholar 

  112. Jin, Y. H. et al. Cadmium is a mutagen that acts by inhibiting mismatch repair. Nat. Genet. 34, 326–329 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Mertz, T. M., Sharma, S., Chabes, A. & Shcherbakova, P. V. Colon cancer-associated mutator DNA polymerase δ variant causes expansion of dNTP pools increasing its own infidelity. Proc. Natl Acad. Sci. USA 112, E2467–E2476 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Hyman, D. M. et al. Vemurafenib in multiple nonmelanoma cancers with BRAF V600 mutations. N. Engl. J. Med. 373, 726–736 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Loeb, L. A., Bielas, J. H. & Beckman, R. A. Cancers exhibit a mutator phenotype: clinical implications. Cancer Res. 68, 3551–3557 (2008).

    Article  CAS  PubMed  Google Scholar 

  116. Swan, M. K., Johnson, R. E., Prakash, L., Prakash, S. & Aggarwal, A. K. Structural basis of high-fidelity DNA synthesis by yeast DNA polymerase δ. Nat. Struct. Mol. Biol. 16, 979–986 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Wang, J., Yu, P., Lin, T. C., Konigsberg, W. H. & Steitz, T. A. Crystal structures of an NH2-terminal fragment of T4 DNA polymerase and its complexes with single-stranded DNA and with divalent metal ions. Biochemistry 35, 8110–8119 (1996).

    Article  CAS  PubMed  Google Scholar 

  118. Hoang, L. N. et al. Polymerase epsilon exonuclease domain mutations in ovarian endometrioid carcinoma. Int. J. Gynecol. Cancer 25, 1187–1193 (2015).

    Article  PubMed  Google Scholar 

  119. Cerami, E. et al. The cBio Cancer Genomics Portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2, 401–404 (2014).

    Article  Google Scholar 

  120. Abdus Sattar, A. K., Lin, T. C., Jones, C. & Konigsberg, W. H. Functional consequences and exonuclease kinetic parameters of point mutations in bacteriophage T4 DNA polymerase. Biochemistry 35, 16621–16629 (1996).

    Article  CAS  PubMed  Google Scholar 

  121. Stenzinger, A. et al. Mutations in POLE and survival of colorectal cancer patients — link to disease stage and treatment. Cancer Med. 3, 1527–1538 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors thank J. Grimes for help in generating the Pol δ and Pol ε structures and N. de Wind for helpful comments on the manuscript. Work in the host laboratories is supported by an Academy of Medical Sciences/Health Foundation Clinician Scientist Fellowship Award (to D.N.C.), Cancer Research UK (programme grant C6199/A10417 to I.T.), the European Research Council (EVOCAN grant agreement: 340560 to I.T.), the Dutch Cancer Society (grant ref: UL2012-5719 to T.B.) and the Medical Research Council (grant Ref. MR/L016591/1 to S.E.K.) and core funding to the Wellcome Trust Centre for Human Genetics from the Wellcome Trust (ref: 090532/Z/09/Z). D.N.C. has received funding from the Oxford Cancer Centre, UK.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to David N. Church.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Related links

PowerPoint slides

Glossary

Mismatch repair

(MMR). A mechanism of post-replicative DNA repair that removes mismatched bases and small insertions and deletions (indels) from the newly synthesized DNA strand.

Ultramutated

Describes tumours with an exceptional frequency of mutations, often exceeding 100 mutations per megabase and often caused by mutations in the gene encoding the catalytic subunit of DNA polymerase ε (POLE).

Neoepitopes

Novel epitopes, created by somatic mutations in tumours, that are capable of eliciting an antitumour immune response.

B family polymerases

Replicative and repair DNA polymerases that are grouped on the basis of sequence and structural similarities, and include the prokaryotic DNA polymerase Pol II, and the eukaryotic DNA polymerases Pol ζ, Pol α, Pol δ and Pol ε.

GINS

A multiprotein complex essential for eukaryotic DNA replication. The name is derived from the Japanese go, ichi, ni and san (meaning five, one, two and three), and refers to the constituent components in budding yeast: Sld5, Psf1, Psf2 and Psf3.

Mutator phenotype

Elevation of the mutation rate above that expected for normally dividing cells. It has been argued that the acquisition of a mutator phenotype is a characteristic of many cancers.

Microsatellite-stable

Absence of small insertions and deletions (indels) at repetitive DNA microsatellites as a result of proficient DNA mismatch repair.

Intestinal polyposis

A phenotype of multiple colonic polyps and increased risk of colorectal cancer that may be due to a germline mutation in a known predisposition gene (for example, adenomatous polyposis coli (APC) or MUTYH) or may lack an obvious genetic cause.

Mutational hot spot

A genomic region that is the site of mutations in cancers at a frequency greater than that expected by chance: for example, codon 600 in the serine/threonine kinase BRAF.

Multinucleated giant cells

Abnormally large cells with multiple nuclei and typically bizarre morphology that are observed in association with chronic inflammation and some types of malignancy.

Microsatellite instability

(MSI). The presence of small insertions and deletions (indels) at repetitive DNA microsatellites that result from defective DNA mismatch repair.

Synthetically lethal

A type of genetic interaction in which the combination of mutations in two genes results in cell death, whereas cells harbouring only one of the mutations are viable.

Antimutator mutations

Mutations that result in an increase in the fidelity of DNA replication; examples have been reported in both Escherichia coli and Saccharomyces cerevisiae, although their occurrence in human cancer is currently not proved.

Polymerase proofreading-associated polyposis

(PPAP). A high-penetrance, dominantly inherited condition characterized by multiple colorectal adenomas and carcinomas, caused by germline mutations in the genes encoding the catalytic subunits of the DNA polymerases Pol δ and Pol ε (POLD1 and POLE, respectively).

Lynch syndrome

A tumour predisposition syndrome caused by germline mutation of mismatch repair genes, which manifests as increased risk of carcinomas of the colon and rectum, endometrium, ovary, stomach and several other organs.

MUTYH-associated polyposis

A syndrome characterized by multiple intestinal adenomas and an increased risk of colorectal carcinoma. It is caused by germline mutations in the base excision repair gene MUTYH.

Basket trial

Also known as a bucket trial. A study in which patients with a specific molecular marker are recruited for treatment with a corresponding targeted therapy, irrespective of tumour histology or tissue of origin.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Rayner, E., van Gool, I., Palles, C. et al. A panoply of errors: polymerase proofreading domain mutations in cancer. Nat Rev Cancer 16, 71–81 (2016). https://doi.org/10.1038/nrc.2015.12

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrc.2015.12

This article is cited by

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