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.

  • Opinion
  • Published:

Poised epigenetic states and acquired drug resistance in cancer

Abstract

Epigenetic events, which are somatically inherited through cell division, are potential drivers of acquired drug resistance in cancer. The high rate of epigenetic change in tumours generates diversity in gene expression patterns that can rapidly evolve through drug selection during treatment, leading to the development of acquired resistance. This will potentially confound stratified chemotherapy decisions that are solely based on mutation biomarkers. Poised epigenetic states in tumour cells may drive multistep epigenetic fixation of gene expression during the acquisition of drug resistance, which has implications for clinical strategies to prevent the emergence of drug resistance.

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: Epigenetics of acquired drug resistance.

Similar content being viewed by others

References

  1. Agarwal, R. & Kaye, S. B. Ovarian cancer: strategies for overcoming resistance to chemotherapy. Nature Rev. Cancer 3, 502–516 (2003).

    Article  CAS  Google Scholar 

  2. Camidge, D. R., Pao, W. & Sequist, L. V. Acquired resistance to TKIs in solid tumours: learning from lung cancer. Nature Rev. Clin. Oncol. 11, 473–481 (2014).

    Article  CAS  Google Scholar 

  3. Pujade-Lauraine, E. et al. Pegylated liposomal doxorubicin and carboplatin compared with paclitaxel and carboplatin for patients with platinum-sensitive ovarian cancer in late relapse. J. Clin. Oncol. 28, 3323–3329 (2010).

    Article  CAS  PubMed  Google Scholar 

  4. Kaplan, B., Qazi, Y. & Wellen, J. R. Strategies for the management of adverse events associated with mTOR inhibitors. Transplant. Rev. 28, 126–133 (2014).

    Article  Google Scholar 

  5. Shen, D.-W., Pastan, I. & Gottesman, M. M. In situ hybridization analysis of acquisition and loss of the human multidrug-resistance gene. Cancer Res. 48, 4334–4339 (1988).

    CAS  PubMed  Google Scholar 

  6. Zeller, C. et al. Candidate DNA methylation drivers of acquired cisplatin resistance in ovarian cancer identified by methylome and expression profiling. Oncogene 31, 4567–4576 (2012).

    Article  CAS  PubMed  Google Scholar 

  7. Papouli, E., Cejka, P. & Jiricny, J. Dependence of the cytotoxicity of DNA-damaging agents on the mismatch repair status of human cells. Cancer Res. 64, 3391–3394 (2004).

    Article  CAS  PubMed  Google Scholar 

  8. Yatabe, Y., Tavaré, S. & Shibata, D. Investigating stem cells in human colon by using methylation patterns. Proc. Natl Acad. Sci. 98, 10839–10844 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Vogelstein, B. et al. Cancer genome landscapes. Science 339, 1546–1558 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Easwaran, H., Tsai, H.-C. & Baylin, S. B. Cancer epigenetics: tumor heterogeneity, plasticity of stem-like states, and drug resistance. Mol. Cell 54, 716–727 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Baylin, S. B. & Jones, P. A. A decade of exploring the cancer epigenome — biological and translational implications. Nature. Rev. Cancer 11, 726–734 (2011).

    Article  CAS  Google Scholar 

  12. Glasspool, R. M., Teodoridis, J. M. & Brown, R. Epigenetics as a mechanism driving polygenic clinical drug resistance. Br. J. Cancer 94, 1087–1092 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Burrell, R. A. & Swanton, C. The evolution of the unstable cancer genome. Curr. Opin. Genet. Dev. 24, 61–67 (2014).

    Article  CAS  PubMed  Google Scholar 

  14. Cherblanc, F., Chapman-Rothe, N., Brown, R. & Fuchter, M. J. Current limitations and future opportunities for epigenetic therapies. Future Med. Chem. 4, 425–446 (2012).

    Article  CAS  PubMed  Google Scholar 

  15. Kaina, B. & Christmann, M. DNA repair in resistance to alkylating anticancer drugs. Int. J. Clin. Pharmacol. Ther. 40, 354–367 (2002).

    Article  CAS  PubMed  Google Scholar 

  16. Hegi, M. E. et al. MGMT gene silencing and benefit from temozolomide in glioblastoma. New Engl. J. Med. 352, 997–1003 (2005).

    Article  CAS  PubMed  Google Scholar 

  17. Wick, W. et al. Temozolomide chemotherapy alone versus radiotherapy alone for malignant astrocytoma in the elderly: the NOA-08 randomised, phase 3 trial. Lancet Oncol. 13, 707–715 (2012).

    Article  CAS  PubMed  Google Scholar 

  18. Malmström, A. et al. Temozolomide versus standard 6-week radiotherapy versus hypofractionated radiotherapy in patients older than 60 years with glioblastoma: the Nordic randomised, phase 3 trial. Lancet Oncol. 13, 916–926 (2012).

    Article  CAS  PubMed  Google Scholar 

  19. Brandes, A. A. et al. O6-methylguanine DNA-methyltransferase methylation status can change between first surgery for newly diagnosed glioblastoma and second surgery for recurrence: clinical implications. Neuro Oncol. 12, 283–288 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Felsberg, J. et al. Promoter methylation and expression of MGMT and the DNA mismatch repair genes MLH1, MSH2, MSH6 and PMS2 in paired primary and recurrent glioblastomas. Int. J. Cancer 129, 659–670 (2011).

    Article  CAS  PubMed  Google Scholar 

  21. Parkinson, J. F. et al. Variation of O6-methylguanine-DNA methyltransferase (MGMT) promoter methylation in serial samples in glioblastoma. J. Neurooncol. 87, 71–78 (2008).

    Article  CAS  PubMed  Google Scholar 

  22. Jung, T.-Y. et al. Changes of the O6-methylguanine-DNA methyltransferase promoter methylation and MGMT protein expression after adjuvant treatment in glioblastoma. Oncol. Rep. 23, 1269–1276 (2010).

    Article  CAS  PubMed  Google Scholar 

  23. Christmann, M. et al. MGMT activity, promoter methylation and immunohistochemistry of pretreatment and recurrent malignant gliomas: a comparative study on astrocytoma and glioblastoma. Int. J. Cancer 127, 2106–2118 (2010).

    Article  CAS  PubMed  Google Scholar 

  24. Park, C.-K. et al. The changes in MGMT promoter methylation status in initial and recurrent glioblastomas. Transl. Oncol. 5, 393–397 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Zhang, Y.-W. et al. Integrated analysis of DNA methylation and mRNA expression profiling reveals candidate genes associated with cisplatin resistance in non-small cell lung cancer. Epigenetics http://dx.doi.org/10.4161/epi.28601 (2014).

  26. Plumb, J. A., Strathdee, G., Sludden, J., Kaye, S. B. & Brown, R. Reversal of drug resistance in human tumor xenografts by 2′-deoxy-5-azacytidine-induced demethylation of the hMLH1 gene promoter. Cancer Res. 60, 6039–6044 (2000).

    CAS  PubMed  Google Scholar 

  27. Chang, X. et al. Identification of hypermethylated genes associated with cisplatin resistance in human cancers. Cancer Res. 70, 2870–2879 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Strathdee, G., MacKean, M., Illand, M. & Brown, R. A role for methylation of the hMLH1 promoter in loss of hMLH1 expression and drug resistance in ovarian cancer. Oncogene 18, 2335–2341 (1999).

    Article  CAS  PubMed  Google Scholar 

  29. Gifford, G., Paul, J., Vasey, P. A., Kaye, S. B. & Brown, R. The acquisition of hMLH1 methylation in plasma DNA after chemotherapy predicts poor survival for ovarian cancer patients. Clin. Cancer Res. 10, 4420–4426 (2004).

    Article  CAS  PubMed  Google Scholar 

  30. McShane, L. M. et al. Reporting recommendations for tumor marker prognostic studies. J. Clin. Oncol. 23, 9067–9072 (2005).

    Article  PubMed  Google Scholar 

  31. Balko, J. M. et al. Profiling of residual breast cancers after neoadjuvant chemotherapy identifies DUSP4 deficiency as a mechanism of drug resistance. Nature Med. 18, 1052–1059 (2012).

    Article  CAS  PubMed  Google Scholar 

  32. Watanabe, Y. et al. A change in promoter methylation of hMLH1 is a cause of acquired resistance to platinum-based chemotherapy in epithelial ovarian cancer. Anticancer Res. 27, 1449–1452 (2007).

    CAS  PubMed  Google Scholar 

  33. Holohan, C., Van Schaeybroeck, S., Longley, D. B. & Johnston, P. G. Cancer drug resistance: an evolving paradigm. Nature Rev. Cancer 13, 714–726 (2013).

    Article  CAS  Google Scholar 

  34. Monsma, D. J. et al. Using a rhabdomyosarcoma patient-derived xenograft to examine precision medicine approaches and model acquired resistance. Pediatr. Blood Cancer 61, 1570–1577 (2014).

    Article  CAS  PubMed  Google Scholar 

  35. Sternberg, S. H., Redding, S., Jinek, M., Greene, E. C. & Doudna, J. A. DNA interrogation by the CRISPR RNA-guided endonuclease Cas9. Nature 507, 62–67 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Mendenhall, E. M. et al. Locus-specific editing of histone modifications at endogenous enhancers. Nature Biotech. 31, 1133–1136 (2013).

    Article  CAS  Google Scholar 

  37. Murtaza, M. et al. Non-invasive analysis of acquired resistance to cancer therapy by sequencing of plasma DNA. Nature 497, 108–112 (2013).

    Article  CAS  PubMed  Google Scholar 

  38. Beck, B. & Blanpain, C. Unravelling cancer stem cell potential. Nature Rev. Cancer 13, 727–738 (2013).

    Article  CAS  Google Scholar 

  39. Shlush, L. I. et al. Identification of pre-leukaemic haematopoietic stem cells in acute leukaemia. Nature 506, 328–333 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Rizzo, S. et al. Ovarian cancer stem cell-like side populations are enriched following chemotherapy and overexpress EZH2. Mol. Cancer Ther. 10, 325–335 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Meacham, C. E. & Morrison, S. J. Tumour heterogeneity and cancer cell plasticity. Nature 501, 328–337 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Anderson, K. et al. Genetic variegation of clonal architecture and propagating cells in leukaemia. Nature 469, 356–361 (2011).

    Article  CAS  PubMed  Google Scholar 

  43. Ding, L. et al. Clonal evolution in relapsed acute myeloid leukaemia revealed by whole-genome sequencing. Nature 481, 506–510 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Mullighan, C. G. et al. Genomic analysis of the clonal origins of relapsed acute lymphoblastic leukemia. Science 322, 1377–1380 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Sharma, S. V. et al. A chromatin-mediated reversible drug-tolerant state in cancer cell subpopulations. Cell 141, 69–80 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Knoechel, B. et al. An epigenetic mechanism of resistance to targeted therapy in T cell acute lymphoblastic leukemia. Nature Genet. 46, 364–370 (2014).

    Article  CAS  PubMed  Google Scholar 

  47. Gupta, P. B. et al. Stochastic state transitions give rise to phenotypic equilibrium in populations of cancer cells. Cell 146, 633–644 (2011).

    Article  CAS  PubMed  Google Scholar 

  48. Chaffer, C. L. et al. Normal and neoplastic nonstem cells can spontaneously convert to a stem-like state. Proc. Natl Acad. Sci. 108, 7950–7955 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  49. Schlesinger, Y. et al. Polycomb-mediated methylation on Lys27 of histone H3 pre-marks genes for de novo methylation in cancer. Nature Genet. 39, 232–236 (2006).

    Article  CAS  PubMed  Google Scholar 

  50. Ohm, J. E. et al. A stem cell–like chromatin pattern may predispose tumor suppressor genes to DNA hypermethylation and heritable silencing. Nature Genet. 39, 237–242 (2007).

    Article  CAS  PubMed  Google Scholar 

  51. Widschwendter, M. et al. Epigenetic stem cell signature in cancer. Nature Genet. 39, 157–158 (2007).

    Article  CAS  PubMed  Google Scholar 

  52. Easwaran, H. et al. A DNA hypermethylation module for the stem/progenitor cell signature of cancer. Genome Res. 22, 837–849 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Zhuang, J. et al. The dynamics and prognostic potential of DNA methylation changes at stem cell gene loci in women's cancer. PLoS Genet. 8, e1002517 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Bernstein, B. E. et al. A bivalent chromatin structure marks key developmental genes in embryonic stem cells. Cell 125, 315–326 (2006).

    Article  CAS  PubMed  Google Scholar 

  55. Azuara, V. et al. Chromatin signatures of pluripotent cell lines. Nature Cell Biol. 8, 532–538 (2006).

    Article  CAS  PubMed  Google Scholar 

  56. Chapman-Rothe, N. et al. Chromatin H3K27me3/H3K4me3 histone marks define gene sets in high grade serous ovarian cancer that distinguish malignant, tumour sustaining and chemo-resistant ovarian tumour cells. Oncogene 19, 4586–4592 (2012).

    Google Scholar 

  57. Chaffer, C. L. et al. Poised chromatin at the ZEB1 promoter enables breast cancer cell plasticity and enhances tumorigenicity. Cell 154, 61–74 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Jansen, M. P. et al. Hallmarks of aromatase inhibitor drug resistance revealed by epigenetic profiling in breast cancer. Cancer Res. 73, 6632–6641 (2013).

    Article  CAS  PubMed  Google Scholar 

  59. Magnani, L. et al. Genome-wide reprogramming of the chromatin landscape underlies endocrine therapy resistance in breast cancer. Proc. Natl Acad. Sci. 110, 1490–1499 (2013).

    Article  Google Scholar 

  60. Verma, S. K. et al. Identification of potent, selective, cell-active inhibitors of the histone lysine methyltransferase EZH2. ACS Med. Chem. Lett. 3, 1091–1096 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. McCabe, M. T. et al. EZH2 inhibition as a therapeutic strategy for lymphoma with EZH2-activating mutations. Nature 492, 108–112 (2012).

    Article  CAS  PubMed  Google Scholar 

  62. Knutson, S. K. et al. A selective inhibitor of EZH2 blocks H3K27 methylation and kills mutant lymphoma cells. Nature Chem. Biol. 8, 890–896 (2012).

    Article  CAS  Google Scholar 

  63. Qi, W. et al. Selective inhibition of Ezh2 by a small molecule inhibitor blocks tumor cells proliferation. Proc. Natl Acad. Sci. 109, 21360–21365 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  64. Konze, K. D. et al. An orally bioavailable chemical probe of the lysine methyltransferases EZH2 and EZH1. ACS Chem. Biol. 8, 1324–1334 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Garapaty-Rao, S. et al. Identification of EZH2 and EZH1 small molecule inhibitors with selective impact on diffuse large B cell lymphoma cell growth. Chem. Biol. 20, 1329–1339 (2013).

    Article  CAS  PubMed  Google Scholar 

  66. Varambally, S. et al. The polycomb group protein EZH2 is involved in progression of prostate cancer. Nature 419, 624–629 (2002).

    Article  CAS  PubMed  Google Scholar 

  67. Bracken, A. P. et al. EZH2 is downstream of the pRB-E2F pathway, essential for proliferation and amplified in cancer. EMBO J. 22, 5323–5335 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Li, H., Cai, Q., Godwin, A. K. & Zhang, R. Enhancer of zeste homolog 2 promotes the proliferation and invasion of epithelial ovarian cancer cells. Mol. Cancer Res. 8, 1610–1618 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Knutson, S. K. et al. Durable tumor regression in genetically altered malignant rhabdoid tumors by inhibition of methyltransferase EZH2. Proc. Natl Acad. Sci. 110, 7922–7927 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  70. Thomas, A. et al. Drug-induced apoptosis in B cell chronic lymphocytic leukemia: relationship between p53 gene mutation and bcl-2/bax proteins in drug resistance. Oncogene 12, 1055–1062 (1996).

    CAS  PubMed  Google Scholar 

  71. Perego, P. et al. Association between cisplatin resistance and mutation of p53 gene and reduced BAX expression in ovarian carcinoma cell systems. Cancer Res. 56, 556–562 (1996).

    CAS  PubMed  Google Scholar 

  72. Senisterra, G. et al. Small-molecule inhibition of MLL activity by disruption of its interaction with WDR5. Biochem. J. 449, 151–159 (2013).

    Article  CAS  PubMed  Google Scholar 

  73. Chen, Q., Van der Sluis, P. C., Boulware, D., Hazlehurst, L. A. & Dalton, W. S. The FANC/BRCA pathway is involved in melphalan induced DNA interstrand crosslink repair and accounts for melphalan resistance in multiple myeloma cells. Blood 106, 698–705 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Edwards, S. L. et al. Resistance to therapy caused by intragenic deletion in BRCA2. Nature 451, 1111–1115 (2008).

    Article  CAS  PubMed  Google Scholar 

  75. Kitange, G. J. et al. Induction of MGMT expression is associated with temozolomide resistance in glioblastoma xenografts. Neuro Oncol. 11, 281–291 (2009).

    CAS  Google Scholar 

  76. Zeller, C. & Brown, R. Therapeutic modulation of epigenetic drivers of drug resistance in ovarian cancer. Ther. Adv. Med. Oncol. 2, 319–329 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Matei, D. et al. Epigenetic resensitization to platinum in ovarian cancer. Cancer Res. 72, 2197–2205 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Glasspool, R. et al. A randomised, Phase II trial of the DNA-hypomethylating agent 5-aza-2′-deoxycytidine (decitabine) in combination with carboplatin versus carboplatin alone in patients with recurrent, partially platinum-sensitive ovarian cancer. Br. J. Cancer 110, 1923–1929 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Fan, H. et al. Low-dose decitabine-based chemoimmunotherapy for patients with refractory advanced solid tumors: a Phase I/II report. J. Immunol. Res. 2014, 371087 (2014).

    PubMed  PubMed Central  Google Scholar 

  80. Appleton, K. et al. Phase I and pharmacodynamic trial of the DNA methyltransferase inhibitor decitabine and carboplatin in solid tumors. J. Clin. Oncol. 25, 4603–4609 (2007).

    Article  CAS  PubMed  Google Scholar 

  81. Chapman-Rothe, N. & Brown, R. Approaches to target the genome and its epigenome in cancer. Future Med. Chem. 1, 1481–1495 (2009).

    Article  CAS  PubMed  Google Scholar 

  82. Kitange, G. J. et al. Inhibition of histone deacetylation potentiates the evolution of acquired temozolomide resistance linked to MGMT upregulation in glioblastoma xenografts. Clin. Cancer Res. 18, 4070–4079 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Li, S. et al. Endocrine-therapy-resistant ESR1 variants revealed by genomic characterization of breast-cancer-derived xenografts. Cell Rep. 4, 1116–1130 (2013).

    Article  CAS  PubMed  Google Scholar 

  84. Topp, M. D. et al. Molecular correlates of platinum response in human high-grade serous ovarian cancer patient-derived xenografts. Mol. Oncol. 8, 656–668 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  85. Jaspers, J. E. et al. Loss of 53BP1 causes PARP inhibitor resistance in BRCA1-mutated mouse mammary tumors. Cancer Discov. 3, 68–81 (2013).

    Article  CAS  PubMed  Google Scholar 

  86. O'Hagan, H. M. et al. Oxidative damage targets complexes containing DNA methyltransferases, SIRT1, and polycomb members to promoter CpG Islands. Cancer Cell 20, 606–619 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Bauer, S. et al. Phase I study of panobinostat and imatinib in patients with treatment-refractory metastatic gastrointestinal stromal tumors. Br. J. Cancer 110, 1155–1162 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Garrido-Laguna, I. et al. A Phase I/II study of decitabine in combination with panitumumab in patients with wild-type (wt) KRAS metastatic colorectal cancer. Invest. New Drugs 31, 1257–1264 (2013).

    Article  CAS  PubMed  Google Scholar 

  89. Falchook, G. S. et al. Methylation and histone deacetylase inhibition in combination with platinum treatment in patients with advanced malignancies. Invest. New Drugs 31, 1192–1200 (2013).

    Article  CAS  PubMed  Google Scholar 

  90. Fu, S. et al. Phase 1b-2a study to reverse platinum resistance through use of a hypomethylating agent, azacitidine, in patients with platinum-resistant or platinum-refractory epithelial ovarian cancer. Cancer 117, 1661–1669 (2011).

    Article  CAS  PubMed  Google Scholar 

  91. Sonpavde, G. et al. Azacitidine favorably modulates PSA kinetics correlating with plasma DNA LINE-1 hypomethylation in men with chemonaïve castration-resistant prostate cancer. Urol. Oncol. 29, 682–689 (2011).

    Article  CAS  PubMed  Google Scholar 

  92. Munster, P. et al. A Phase II study of the histone deacetylase inhibitor vorinostat combined with tamoxifen for the treatment of patients with hormone therapy-resistant breast cancer. Br. J. Cancer 104, 1828–1835 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Dizon, D. S. et al. A Phase II evaluation of belinostat and carboplatin in the treatment of recurrent or persistent platinum-resistant ovarian, fallopian tube, or primary peritoneal carcinoma: a Gynecologic Oncology Group study. Gynecol. Oncol. 125, 367–371 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Dizon, D. S. et al. Phase II activity of belinostat (PXD-101), carboplatin, and paclitaxel in women with previously treated ovarian cancer. Int. J. Gynecol. Cancer 22, 979–986 (2012).

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

The authors thank S. Kaye and E. Loomis for comments on the manuscript. The authors' research on epigenetics of drug resistance is supported by grants from Cancer Research UK (A13086) and Ovarian Cancer Action, London, UK.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Robert Brown.

Ethics declarations

Competing interests

Grants from Cancer Research UK and Ovarian Cancer Action, which is acknowledged (R.B.). Patent on histone methyltransferase inhibitors (R.B.). E.C., L.M., C.S.W.-B and J.B. declare no competing interests.

Related links

FURTHER INFORMATION

BriTROC website

PowerPoint slides

Supplementary information

41568_2014_BFnrc3819_MOESM11_ESM.pdf

Supplementary information S1 | Systematic review of the published literature on epigenetic mechanisms involved in drug resistance in clinical studies (PDF 369 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Brown, R., Curry, E., Magnani, L. et al. Poised epigenetic states and acquired drug resistance in cancer. Nat Rev Cancer 14, 747–753 (2014). https://doi.org/10.1038/nrc3819

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrc3819

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