Skip to main content

Epigenetic Regulation in Breast Cancer

  • Chapter
  • First Online:
Translational Research in Breast Cancer

Part of the book series: Advances in Experimental Medicine and Biology ((AEMB,volume 1187))

Abstract

Aberrant epigenetic alteration has been associated with development of various cancers, including breast cancer. Since epigenetic modifications such as DNA methylation and histone modification are reversible, epigenetic enzymes, including histone modifying enzymes and DNA methyltransferases, emerge as attractive targets for cancer therapy. Although epi-drugs targeting histone deacetylation or DNA methylation have received FDA approval for cancer therapy, a very modest anti-tumor activity has been observed with monotherapy in clinical studies of breast cancer. To improve efficacy of epi-drugs in breast cancer, combination of epi-drugs with other therapies currently has been investigated. Additionally, basic researches to elucidate molecular causes of cancer should be extensively and intensively conducted in order to find novel epigenetic druggable targets. In this chapter, we summarize how epigenetic regulation affects the development of breast cancer and how to control cancer phenotype by modulating abnormal epigenetic modifications, and then suggest future research directions in epigenetics for breast cancer treatment.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 109.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 139.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 199.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

Similar content being viewed by others

References

  1. Esteller M. CpG island hypermethylation and tumor suppressor genes: a booming present, a brighter future. Oncogene. 2002;21(35):5427–40.

    Article  CAS  PubMed  Google Scholar 

  2. Jones PA, Baylin SB. The fundamental role of epigenetic events in cancer. Nat Rev Genet. 2002;3(6):415–28.

    Article  CAS  PubMed  Google Scholar 

  3. Radpour R, Barekati Z, Kohler C, Schumacher MM, Grussenmeyer T, Jenoe P, et al. Integrated epigenetics of human breast cancer: synoptic investigation of targeted genes, microRNAs and proteins upon demethylation treatment. PLoS One. 2011;6(11):e27355.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Dawson MA, Kouzarides T. Cancer epigenetics: from mechanism to therapy. Cell. 2012;150(1):12–27.

    Article  CAS  PubMed  Google Scholar 

  5. Jenuwein T, Allis CD. Translating the histone code. Science. 2001;293(5532):1074–80.

    Article  CAS  PubMed  Google Scholar 

  6. Venteicher AS, Meng Z, Mason PJ, Veenstra TD, Artandi SE. Identification of ATPases pontin and reptin as telomerase components essential for holoenzyme assembly. Cell. 2008;132(6):945–57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Bauer A, Chauvet S, Huber O, Usseglio F, Rothbacher U, Aragnol D, et al. Pontin52 and reptin52 function as antagonistic regulators of beta-catenin signalling activity. EMBO J. 2000;19(22):6121–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Kim JH, Choi HJ, Kim B, Kim MH, Lee JM, Kim IS, et al. Roles of sumoylation of a reptin chromatin-remodelling complex in cancer metastasis. Nat Cell Biol. 2006;8(6):631–9.

    Article  CAS  PubMed  Google Scholar 

  9. Kim JH, Lee JM, Nam HJ, Choi HJ, Yang JW, Lee JS, et al. SUMOylation of pontin chromatin-remodeling complex reveals a signal integration code in prostate cancer cells. Proc Natl Acad Sci U S A. 2007;104(52):20793–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Shen X, Mizuguchi G, Hamiche A, Wu C. A chromatin remodelling complex involved in transcription and DNA processing. Nature. 2000;406(6795):541–4.

    Article  CAS  PubMed  Google Scholar 

  11. Lee JS, Kim Y, Kim IS, Kim B, Choi HJ, Lee JM, et al. Negative regulation of hypoxic responses via induced Reptin methylation. Mol Cell. 2010;39(1):71–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Graeber TG, Osmanian C, Jacks T, Housman DE, Koch CJ, Lowe SW, et al. Hypoxia-mediated selection of cells with diminished apoptotic potential in solid tumours. Nature. 1996;379(6560):88–91.

    Article  CAS  PubMed  Google Scholar 

  13. Semenza GL. HIF-1 and tumor progression: pathophysiology and therapeutics. Trends Mol Med. 2002;8(4 Suppl):S62–7.

    Article  CAS  PubMed  Google Scholar 

  14. Bruick RK. Oxygen sensing in the hypoxic response pathway: regulation of the hypoxia-inducible transcription factor. Genes Dev. 2003;17(21):2614–23.

    Article  CAS  PubMed  Google Scholar 

  15. Eales KL, Hollinshead KE, Tennant DA. Hypoxia and metabolic adaptation of cancer cells. Oncogenesis. 2016;5:e190.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Rankin EB, Giaccia AJ. The role of hypoxia-inducible factors in tumorigenesis. Cell Death Differ. 2008;15(4):678–85.

    Article  CAS  PubMed  Google Scholar 

  17. Lee JS, Kim Y, Bhin J, Shin HJ, Nam HJ, Lee SH, et al. Hypoxia-induced methylation of a pontin chromatin remodeling factor. Proc Natl Acad Sci U S A. 2011;108(33):13510–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Kim JH, Kim B, Cai L, Choi HJ, Ohgi KA, Tran C, et al. Transcriptional regulation of a metastasis suppressor gene by Tip60 and beta-catenin complexes. Nature. 2005;434(7035):921–6.

    Article  CAS  PubMed  Google Scholar 

  19. Giguere V. Orphan nuclear receptors: from gene to function. Endocr Rev. 1999;20(5):689–725.

    CAS  PubMed  Google Scholar 

  20. Kim H, Lee JM, Lee G, Bhin J, Oh SK, Kim K, et al. DNA damage-induced RORalpha is crucial for p53 stabilization and increased apoptosis. Mol Cell. 2011;44(5):797–810.

    Article  CAS  PubMed  Google Scholar 

  21. Lee JM, Kim IS, Kim H, Lee JS, Kim K, Yim HY, et al. RORalpha attenuates Wnt/beta-catenin signaling by PKCalpha-dependent phosphorylation in colon cancer. Mol Cell. 2010;37(2):183–95.

    Article  CAS  PubMed  Google Scholar 

  22. Moretti RM, Montagnani Marelli M, Motta M, Limonta P. Role of the orphan nuclear receptor ROR alpha in the control of the metastatic behavior of androgen-independent prostate cancer cells. Oncol Rep. 2002;9(5):1139–43.

    CAS  PubMed  Google Scholar 

  23. Cao R, Wang L, Wang H, Xia L, Erdjument-Bromage H, Tempst P, et al. Role of histone H3 lysine 27 methylation in Polycomb-group silencing. Science. 2002;298(5595):1039–43.

    Article  CAS  PubMed  Google Scholar 

  24. Jones CA, Ng J, Peterson AJ, Morgan K, Simon J, Jones RS. The Drosophila esc and E(z) proteins are direct partners in polycomb group-mediated repression. Mol Cell Biol. 1998;18(5):2825–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Kuzmichev A, Jenuwein T, Tempst P, Reinberg D. Different EZH2-containing complexes target methylation of histone H1 or nucleosomal histone H3. Mol Cell. 2004;14(2):183–93.

    Article  CAS  PubMed  Google Scholar 

  26. Kleer CG, Cao Q, Varambally S, Shen R, Ota I, Tomlins SA, et al. EZH2 is a marker of aggressive breast cancer and promotes neoplastic transformation of breast epithelial cells. Proc Natl Acad Sci U S A. 2003;100(20):11606–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Varambally S, Dhanasekaran SM, Zhou M, Barrette TR, Kumar-Sinha C, Sanda MG, et al. The polycomb group protein EZH2 is involved in progression of prostate cancer. Nature. 2002;419(6907):624–9.

    Article  CAS  PubMed  Google Scholar 

  28. Lee JM, Lee JS, Kim H, Kim K, Park H, Kim JY, et al. EZH2 generates a methyl degron that is recognized by the DCAF1/DDB1/CUL4 E3 ubiquitin ligase complex. Mol Cell. 2012;48(4):572–86.

    Article  CAS  PubMed  Google Scholar 

  29. Bruick RK, McKnight SL. A conserved family of prolyl-4-hydroxylases that modify HIF. Science. 2001;294(5545):1337–40.

    Article  CAS  PubMed  Google Scholar 

  30. Epstein AC, Gleadle JM, McNeill LA, Hewitson KS, O'Rourke J, Mole DR, et al. C. elegans EGL-9 and mammalian homologs define a family of dioxygenases that regulate HIF by prolyl hydroxylation. Cell. 2001;107(1):43–54.

    Article  CAS  PubMed  Google Scholar 

  31. Huang LE, Gu J, Schau M, Bunn HF. Regulation of hypoxia-inducible factor 1alpha is mediated by an O2-dependent degradation domain via the ubiquitin-proteasome pathway. Proc Natl Acad Sci U S A. 1998;95(14):7987–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Maxwell PH, Wiesener MS, Chang GW, Clifford SC, Vaux EC, Cockman ME, et al. The tumour suppressor protein VHL targets hypoxia-inducible factors for oxygen-dependent proteolysis. Nature. 1999;399(6733):271–5.

    Article  CAS  PubMed  Google Scholar 

  33. Ohh M, Park CW, Ivan M, Hoffman MA, Kim TY, Huang LE, et al. Ubiquitination of hypoxia-inducible factor requires direct binding to the beta-domain of the von Hippel-Lindau protein. Nat Cell Biol. 2000;2(7):423–7.

    Article  CAS  PubMed  Google Scholar 

  34. Cheng J, Kang X, Zhang S, Yeh ET. SUMO-specific protease 1 is essential for stabilization of HIF1alpha during hypoxia. Cell. 2007;131(3):584–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Duyndam MC, Hulscher ST, van der Wall E, Pinedo HM, Boven E. Evidence for a role of p38 kinase in hypoxia-inducible factor 1-independent induction of vascular endothelial growth factor expression by sodium arsenite. J Biol Chem. 2003;278(9):6885–95.

    Article  CAS  PubMed  Google Scholar 

  36. Jeong JW, Bae MK, Ahn MY, Kim SH, Sohn TK, Bae MH, et al. Regulation and destabilization of HIF-1alpha by ARD1-mediated acetylation. Cell. 2002;111(5):709–20.

    Article  CAS  PubMed  Google Scholar 

  37. Kim Y, Nam HJ, Lee J, Park DY, Kim C, Yu YS, et al. Methylation-dependent regulation of HIF-1alpha stability restricts retinal and tumour angiogenesis. Nat Commun. 2016;7:10347.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Phadke PA, Vaidya KS, Nash KT, Hurst DR, Welch DR. BRMS1 suppresses breast cancer experimental metastasis to multiple organs by inhibiting several steps of the metastatic process. Am J Pathol. 2008;172(3):809–17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Seraj MJ, Samant RS, Verderame MF, Welch DR. Functional evidence for a novel human breast carcinoma metastasis suppressor, BRMS1, encoded at chromosome 11q13. Cancer Res. 2000;60(11):2764–9.

    CAS  PubMed  Google Scholar 

  40. Kim B, Nam HJ, Pyo KE, Jang MJ, Kim IS, Kim D, et al. Breast cancer metastasis suppressor 1 (BRMS1) is destabilized by the Cul3-SPOP E3 ubiquitin ligase complex. Biochem Biophys Res Commun. 2011;415(4):720–6.

    Article  CAS  PubMed  Google Scholar 

  41. Hernandez-Munoz I, Lund AH, van der Stoop P, Boutsma E, Muijrers I, Verhoeven E, et al. Stable X chromosome inactivation involves the PRC1 Polycomb complex and requires histone MACROH2A1 and the CULLIN3/SPOP ubiquitin E3 ligase. Proc Natl Acad Sci U S A. 2005;102(21):7635–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Kwon JE, La M, Oh KH, Oh YM, Kim GR, Seol JH, et al. BTB domain-containing speckle-type POZ protein (SPOP) serves as an adaptor of Daxx for ubiquitination by Cul3-based ubiquitin ligase. J Biol Chem. 2006;281(18):12664–72.

    Article  CAS  PubMed  Google Scholar 

  43. Cogswell PC, Guttridge DC, Funkhouser WK, Baldwin AS Jr. Selective activation of NF-kappa B subunits in human breast cancer: potential roles for NF-kappa B2/p52 and for Bcl-3. Oncogene. 2000;19(9):1123–31.

    Article  CAS  PubMed  Google Scholar 

  44. Ohno H, Takimoto G, McKeithan TW. The candidate proto-oncogene bcl-3 is related to genes implicated in cell lineage determination and cell cycle control. Cell. 1990;60(6):991–7.

    Article  CAS  PubMed  Google Scholar 

  45. Ahmed SU, Milner J. Basal cancer cell survival involves JNK2 suppression of a novel JNK1/c-Jun/Bcl-3 apoptotic network. PLoS One. 2009;4(10):e7305.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  46. Choi HJ, Lee JM, Kim H, Nam HJ, Shin HJ, Kim D, et al. Bcl3-dependent stabilization of CtBP1 is crucial for the inhibition of apoptosis and tumor progression in breast cancer. Biochem Biophys Res Commun. 2010;400(3):396–402.

    Article  CAS  PubMed  Google Scholar 

  47. O'Neil BH, Buzkova P, Farrah H, Kashatus D, Sanoff H, Goldberg RM, et al. Expression of nuclear factor-kappaB family proteins in hepatocellular carcinomas. Oncology. 2007;72(1-2):97–104.

    Article  CAS  PubMed  Google Scholar 

  48. Thornburg NJ, Pathmanathan R, Raab-Traub N. Activation of nuclear factor-kappaB p50 homodimer/Bcl-3 complexes in nasopharyngeal carcinoma. Cancer Res. 2003;63(23):8293–301.

    CAS  PubMed  Google Scholar 

  49. Giguere V, Tini M, Flock G, Ong E, Evans RM, Otulakowski G. Isoform-specific amino-terminal domains dictate DNA-binding properties of ROR alpha, a novel family of orphan hormone nuclear receptors. Genes Dev. 1994;8(5):538–53.

    Article  CAS  PubMed  Google Scholar 

  50. Kim K, Lee JM, Yu YS, Kim H, Nam HJ, Moon HG, et al. RORalpha2 requires LSD1 to enhance tumor progression in breast cancer. Sci Rep. 2017;7(1):11994.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  51. Ley TJ, Ding L, Walter MJ, McLellan MD, Lamprecht T, Larson DE, et al. DNMT3A mutations in acute myeloid leukemia. N Engl J Med. 2010;363(25):2424–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Derissen EJ, Beijnen JH, Schellens JH. Concise drug review: azacitidine and decitabine. Oncologist. 2013;18(5):619–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Tsai HC, Li H, Van Neste L, Cai Y, Robert C, Rassool FV, et al. Transient low doses of DNA-demethylating agents exert durable antitumor effects on hematological and epithelial tumor cells. Cancer Cell. 2012;21(3):430–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Borges S, Doppler H, Perez EA, Andorfer CA, Sun Z, Anastasiadis PZ, et al. Pharmacologic reversion of epigenetic silencing of the PRKD1 promoter blocks breast tumor cell invasion and metastasis. Breast Cancer Res. 2013;15(2):R66.

    Article  PubMed  PubMed Central  Google Scholar 

  55. Olsen EA, Kim YH, Kuzel TM, Pacheco TR, Foss FM, Parker S, et al. Phase IIb multicenter trial of vorinostat in patients with persistent, progressive, or treatment refractory cutaneous T-cell lymphoma. J Clin Oncol. 2007;25(21):3109–15.

    Article  CAS  PubMed  Google Scholar 

  56. Piekarz RL, Frye R, Turner M, Wright JJ, Allen SL, Kirschbaum MH, et al. Phase II multi-institutional trial of the histone deacetylase inhibitor romidepsin as monotherapy for patients with cutaneous T-cell lymphoma. J Clin Oncol. 2009;27(32):5410–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Beckers T, Burkhardt C, Wieland H, Gimmnich P, Ciossek T, Maier T, et al. Distinct pharmacological properties of second generation HDAC inhibitors with the benzamide or hydroxamate head group. Int J Cancer. 2007;121(5):1138–48.

    Article  CAS  PubMed  Google Scholar 

  58. Kim YJ, Greer CB, Cecchini KR, Harris LN, Tuck DP, Kim TH. HDAC inhibitors induce transcriptional repression of high copy number genes in breast cancer through elongation blockade. Oncogene. 2013;32(23):2828–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Falahi F, van Kruchten M, Martinet N, Hospers GA, Rots MG. Current and upcoming approaches to exploit the reversibility of epigenetic mutations in breast cancer. Breast Cancer Res. 2014;16(4):412.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  60. Park PJ. ChIP-seq: advantages and challenges of a maturing technology. Nat Rev Genet. 2009;10(10):669–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Stunnenberg HG, Vermeulen M. Towards cracking the epigenetic code using a combination of high-throughput epigenomics and quantitative mass spectrometry-based proteomics. BioEssays. 2011;33(7):547–51.

    Article  CAS  PubMed  Google Scholar 

  62. Filippakopoulos P, Qi J, Picaud S, Shen Y, Smith WB, Fedorov O, et al. Selective inhibition of BET bromodomains. Nature. 2010;468(7327):1067–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Dawson MA, Prinjha RK, Dittmann A, Giotopoulos G, Bantscheff M, Chan WI, et al. Inhibition of BET recruitment to chromatin as an effective treatment for MLL-fusion leukaemia. Nature. 2011;478(7370):529–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Delmore JE, Issa GC, Lemieux ME, Rahl PB, Shi J, Jacobs HM, et al. BET bromodomain inhibition as a therapeutic strategy to target c-Myc. Cell. 2011;146(6):904–17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Mertz JA, Conery AR, Bryant BM, Sandy P, Balasubramanian S, Mele DA, et al. Targeting MYC dependence in cancer by inhibiting BET bromodomains. Proc Natl Acad Sci U S A. 2011;108(40):16669–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Zuber J, Rappaport AR, Luo W, Wang E, Chen C, Vaseva AV, et al. An integrated approach to dissecting oncogene addiction implicates a Myb-coordinated self-renewal program as essential for leukemia maintenance. Genes Dev. 2011;25(15):1628–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Asangani IA, Dommeti VL, Wang X, Malik R, Cieslik M, Yang R, et al. Therapeutic targeting of BET bromodomain proteins in castration-resistant prostate cancer. Nature. 2014;510(7504):278–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Garcia PL, Miller AL, Kreitzburg KM, Council LN, Gamblin TL, Christein JD, et al. The BET bromodomain inhibitor JQ1 suppresses growth of pancreatic ductal adenocarcinoma in patient-derived xenograft models. Oncogene. 2016;35(7):833–45.

    Article  CAS  PubMed  Google Scholar 

  69. Ocana A, Nieto-Jimenez C, Pandiella A. BET inhibitors as novel therapeutic agents in breast cancer. Oncotarget. 2017;8(41):71285–91.

    Article  PubMed  PubMed Central  Google Scholar 

  70. Shimamura T, Chen Z, Soucheray M, Carretero J, Kikuchi E, Tchaicha JH, et al. Efficacy of BET bromodomain inhibition in Kras-mutant non-small cell lung cancer. Clin Cancer Res. 2013;19(22):6183–92.

    Article  CAS  PubMed  Google Scholar 

  71. Nieto-Jimenez C, Alcaraz-Sanabria A, Perez-Pena J, Corrales-Sanchez V, Serrano-Heras G, Galan-Moya EM, et al. Targeting basal-like breast tumors with bromodomain and extraterminal domain (BET) and polo-like kinase inhibitors. Oncotarget. 2017;8(12):19478–90.

    Article  PubMed  PubMed Central  Google Scholar 

  72. Johnson DB, Estrada MV, Salgado R, Sanchez V, Doxie DB, Opalenik SR, et al. Melanoma-specific MHC-II expression represents a tumour-autonomous phenotype and predicts response to anti-PD-1/PD-L1 therapy. Nat Commun. 2016;7:10582.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Pauken KE, Sammons MA, Odorizzi PM, Manne S, Godec J, Khan O, et al. Epigenetic stability of exhausted T cells limits durability of reinvigoration by PD-1 blockade. Science. 2016;354(6316):1160–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Sen DR, Kaminski J, Barnitz RA, Kurachi M, Gerdemann U, Yates KB, et al. The epigenetic landscape of T cell exhaustion. Science. 2016;354(6316):1165–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Wherry EJ, Kurachi M. Molecular and cellular insights into T cell exhaustion. Nat Rev Immunol. 2015;15(8):486–99.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Sung Hee Baek .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2021 Springer Nature Singapore Pte Ltd.

About this chapter

Check for updates. Verify currency and authenticity via CrossMark

Cite this chapter

Nam, H.J., Baek, S.H. (2021). Epigenetic Regulation in Breast Cancer. In: Noh, DY., Han, W., Toi, M. (eds) Translational Research in Breast Cancer. Advances in Experimental Medicine and Biology, vol 1187. Springer, Singapore. https://doi.org/10.1007/978-981-32-9620-6_5

Download citation

Publish with us

Policies and ethics