J Breast Cancer.  2018 Sep;21(3):233-243. 10.4048/jbc.2018.21.e41.

The Epigenetics of Triple-Negative and Basal-Like Breast Cancer: Current Knowledge

Affiliations
  • 1Research Center for Functional Genomics, Biomedicine and Translational Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania.
  • 2Faculty of Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania.
  • 3Division of Dental Propaedeutics, Aesthetic, Department of Prosthetic Dentistry and Dental Materials, “Iuliu Hatieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania. irimie.alexandra@umfcluj.ro
  • 4MedFUTURE Research Center for Advanced Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania.
  • 5Department of Functional Genomics and Experimental Pathology, The Oncology Institute “Prof. Dr. I Chiricuta”, Cluj-Napoca, Romania.

Abstract

Breast cancer has the highest incidence among all malignancies diagnosed in women. Therapies have significantly improved over the years due to extensive molecular and clinical research; in a large number of cases, targeted therapies have provided better prognosis. However, one specific subtype remains elusive to targeted therapies-the triple-negative breast cancer. This immunohistochemically defined subtype is resistant to both endocrine and targeted therapies, leading to its poor prognosis. A field that is of great promise in current cancer research is epigenetics. By studying the epigenetic mechanisms underlying tumorigenesis-DNA methylation, histone modifications, and noncoding RNAs-advances in cancer treatment, diagnosis, and prevention are possible. This review aims to synthesize the epigenetic discoveries that have been made related to the triple-negative breast cancer.

Keyword

Breast neoplasms; DNA methylation; Histone code; Triple negative breast neoplasms; Untranslated RNA

MeSH Terms

Breast Neoplasms*
Breast*
Diagnosis
DNA Methylation
Epigenomics*
Female
Histone Code
Humans
Incidence
Methylation
Prognosis
RNA, Untranslated
Triple Negative Breast Neoplasms
RNA, Untranslated

Figure

  • Figure 1 Classification systems developed for triple-negative breast cancer (TNBC). (A) Intrinsic subtypes of breast cancer, first established in 2001 by Sørlie et al. [5]. This classification is most widely used by both clinicians and researchers due to numerous confirmed prognostic, predictive and therapeutic correlations. The pie chart represents the distribution of these intrinsic subtypes among the imunohistochemically defined TNBC, the majority being represented by basal-like tumors. (B) TNBCtype-4 comprised of six subtypes was established in 2011 by Lehmann et al. [12] and subsequently redefined to TNBCtype-4 in 2016 [13]. The pie chart represents the distribution of these subtypes among TNBC, again the majority being represented by basal-like tumors. ER=estrogen receptor; PR=progesterone receptor; HER2=human epidermal growth factor receptor 2; BL=basal-like; M=mesenchymal; LAR=luminal androgen receptor; EGF=epidermal growth factor; NGF=nerve growth factor; MET=tyrosine-protein kinase Met encoded by the MET gene; IGF1R=insulin-like growth factor 1 receptor.

  • Figure 2 Patterns of methylation described in triple-negative breast cancer. Hypermethylation of CpG islands and shores with hypomethylation of intragenic regions leads to epigenetic silencing. At nucleosomal level the patterns in DNA methylation translate to tri-methylation of lysine 27 on histone H3 (H3K27me3), another marker of epigenetic silencing.

  • Figure 3 HOTAIR in basal-like breast cancer. HOTAIR forms a complex with the enhancer of zeste homolog 2 (EZH2) subunit of the polycomb repressor complex 2 (PRC2) complex, which leads to H3K27 trimethylation and maintenance of the basal-like phenotype. H3K27me3=tri-methylation of lysine 27 on histone H3.


Reference

1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA Cancer J Clin. 2018; 68:7–30.
Article
2. Gierach GL, Burke A, Anderson WF. Epidemiology of triple negative breast cancers. Breast Dis. 2010; 32:5–24.
Article
3. Chiorean R, Braicu C, Berindan-Neagoe I. Another review on triple negative breast cancer: are we on the right way towards the exit from the labyrinth? Breast. 2013; 22:1026–1033.
Article
4. Perou CM, Sørlie T, Eisen MB, van de Rijn M, Jeffrey SS, Rees CA, et al. Molecular portraits of human breast tumours. Nature. 2000; 406:747–752.
Article
5. Sørlie T, Perou CM, Tibshirani R, Aas T, Geisler S, Johnsen H, et al. Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad Sci U S A. 2001; 98:10869–10874.
Article
6. Senkus E, Kyriakides S, Penault-Llorca F, Poortmans P, Thompson A, Zackrisson S, et al. Primary breast cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2013; 24:Suppl 6. vi7–vi23.
Article
7. Lachapelle J, Foulkes WD. Triple-negative and basal-like breast cancer: implications for oncologists. Curr Oncol. 2011; 18:161–164.
Article
8. Foulkes WD, Smith IE, Reis-Filho JS. Triple-negative breast cancer. N Engl J Med. 2010; 363:1938–1948.
Article
9. Lehmann BD, Pietenpol JA. Identification and use of biomarkers in treatment strategies for t riple-negative breast cancer subtypes. J Pathol. 2014; 232:142–150.
Article
10. Perou CM. Molecular stratification of triple-negative breast cancers. Oncologist. 2011; 16:Suppl 1. 61–70.
Article
11. Weigelt B, Baehner FL, Reis-Filho JS. The contribution of gene expression profiling to breast cancer classification, prognostication and prediction: a retrospective of the last decade. J Pathol. 2010; 220:263–280.
Article
12. Lehmann BD, Bauer JA, Chen X, Sanders ME, Chakravarthy AB, Shyr Y, et al. Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies. J Clin Invest. 2011; 121:2750–2767.
Article
13. Lehmann BD, Jovanović B, Chen X, Estrada MV, Johnson KN, Shyr Y, et al. Refinement of triple-negative breast cancer molecular subtypes: implications for neoadjuvant chemotherapy selection. PLoS One. 2016; 11:e0157368.
Article
14. Burstein MD, Tsimelzon A, Poage GM, Covington KR, Contreras A, Fuqua SA, et al. Comprehensive genomic analysis identifies novel subtypes and targets of triple-negative breast cancer. Clin Cancer Res. 2015; 21:1688–1698.
Article
15. Jézéquel P, Loussouarn D, Guérin-Charbonnel C, Campion L, Vanier A, Gouraud W, et al. Gene-expression molecular subtyping of triple-negative breast cancer tumours: importance of immune response. Breast Cancer Res. 2015; 17:43.
Article
16. Ahn SG, Kim SJ, Kim C, Jeong J. Molecular classification of triple-negative breast cancer. J Breast Cancer. 2016; 19:223–230.
Article
17. Kanwal R, Gupta K, Gupta S. Cancer epigenetics: an introduction. Methods Mol Biol. 2015; 1238:3–25.
Article
18. Basse C, Arock M. The increasing roles of epigenetics in breast cancer: implications for pathogenicity, biomarkers, prevention and treatment. Int J Cancer. 2015; 137:2785–2794.
Article
19. Tammen SA, Friso S, Choi SW. Epigenetics: the link between nature and nurture. Mol Aspects Med. 2013; 34:753–764.
Article
20. Jones PA, Baylin SB. The epigenomics of cancer. Cell. 2007; 128:683–692.
Article
21. Elsheikh SE, Green AR, Rakha EA, Powe DG, Ahmed RA, Collins HM, et al. Global histone modifications in breast cancer correlate with tumor phenotypes, prognostic factors, and patient outcome. Cancer Res. 2009; 69:3802–3809.
Article
22. Palazzo AF, Lee ES. Non-coding RNA: what is functional and what is junk? Front Genet. 2015; 6:2.
Article
23. Stirzaker C, Zotenko E, Song JZ, Qu W, Nair SS, Locke WJ, et al. Methylome sequencing in triple-negative breast cancer reveals distinct methylation clusters with prognostic value. Nat Commun. 2015; 6:5899.
Article
24. Harburg GC, Hinck L. Navigating breast cancer: axon guidance molecules as breast cancer tumor suppressors and oncogenes. J Mammary Gland Biol Neoplasia. 2011; 16:257–270.
Article
25. Robichaux MA, Cowan CW. Signaling mechanisms of axon guidance and early synaptogenesis. Curr Top Behav Neurosci. 2014; 16:19–48.
Article
26. Braicu C, Chiorean R, Irimie A, Chira S, Tomuleasa C, Neagoe E, et al. Novel insight into triple-negative breast cancers, the emerging role of angiogenesis, and antiangiogenic therapy. Expert Rev Mol Med. 2016; 18:e18.
Article
27. Branham MT, Marzese DM, Laurito SR, Gago FE, Orozco JI, Tello OM, et al. Methylation profile of triple-negative breast carcinomas. Oncogenesis. 2012; 1:e17.
Article
28. Greenup R, Buchanan A, Lorizio W, Rhoads K, Chan S, Leedom T, et al. Prevalence of BRCA mutations among women with triple-negative breast cancer (TNBC) in a genetic counseling cohort. Ann Surg Oncol. 2013; 20:3254–3258.
Article
29. Pop LA, Cojocneanu-Petric RM, Pileczki V, Morar-Bolba G, Irimie A, Lazar V, et al. Genetic alterations in sporadic triple negative breast cancer. Breast. 2018; 38:30–38.
Article
30. Lord CJ, Ashworth A. BRCAness revisited. Nat Rev Cancer. 2016; 16:110–120.
Article
31. Lips EH, Mulder L, Oonk A, van der Kolk LE, Hogervorst FB, Imholz AL, et al. Triple-negative breast cancer: BRCAness and concordance of clinical features with BRCA1-mutation carriers. Br J Cancer. 2013; 108:2172–2177.
Article
32. Murria R, Palanca S, de Juan I, Alenda C, Egoavil C, Seguí FJ, et al. Immunohistochemical, genetic and epigenetic profiles of hereditary and triple negative breast cancers: relevance in personalized medicine. Am J Cancer Res. 2015; 5:2330–2343.
33. Sharma P, Stecklein SR, Kimler BF, Sethi G, Petroff BK, Phillips TA, et al. The prognostic value of BRCA1 promoter methylation in early stage triple negative breast cancer. J Cancer Ther Res. 2014; 3:1–11.
Article
34. Stefansson OA, Villanueva A, Vidal A, Martí L, Esteller M. BRCA1 epigenetic inactivation predicts sensitivity to platinum-based chemotherapy in breast and ovarian cancer. Epigenetics. 2012; 7:1225–1229.
Article
35. Veeck J, Ropero S, Setien F, Gonzalez-Suarez E, Osorio A, Benitez J, et al. BRCA1 CpG island hypermethylation predicts sensitivity to poly(adenosine diphosphate)-ribose polymerase inhibitors. J Clin Oncol. 2010; 28:e563–e564.
36. Mori H, Kubo M, Nishimura R, Osako T, Arima N, Okumura Y, et al. BRCAness as a biomarker for predicting prognosis and response to anthracycline-based adjuvant chemotherapy for patients with triple-negative breast cancer. PLoS One. 2016; 11:e0167016.
Article
37. Mathe A, Wong-Brown M, Locke WJ, Stirzaker C, Braye SG, Forbes JF, et al. DNA methylation profile of triple negative breast cancer-specific genes comparing lymph node positive patients to lymph node negative patients. Sci Rep. 2016; 6:33435.
Article
38. Kong B, Lv ZD, Wang Y, Jin LY, Ding L, Yang ZC. Down-regulation of BRMS1 by DNA hypermethylation and its association with metastatic progression in triple-negative breast cancer. Int J Clin Exp Pathol. 2015; 8:11076–11083.
39. Kagara N, Huynh KT, Kuo C, Okano H, Sim MS, Elashoff D, et al. Epigenetic regulation of cancer stem cell genes in triple-negative breast cancer. Am J Pathol. 2012; 181:257–267.
Article
40. Cancer Genome Atlas Network. Comprehensive molecular portraits of human breast tumours. Nature. 2012; 490:61–70.
41. Holm K, Hegardt C, Staaf J, Vallon-Christersson J, Jönsson G, Olsson H, et al. Molecular subtypes of breast cancer are associated with characteristic DNA methylation patterns. Breast Cancer Res. 2010; 12:R36.
Article
42. Sunami E, Shinozaki M, Sim MS, Nguyen SL, Vu AT, Giuliano AE, et al. Estrogen receptor and HER2/neu status affect epigenetic differences of tumor-related genes in primary breast tumors. Breast Cancer Res. 2008; 10:R46.
Article
43. Hughes LA, Melotte V, de Schrijver J, de Maat M, Smit VT, Bovée JV, et al. The CpG island methylator phenotype: what's in a name? Cancer Res. 2013; 73:5858–5868.
Article
44. Roll JD, Rivenbark AG, Jones WD, Coleman WB. DNMT3b overexpression contributes to a hypermethylator phenotype in human breast cancer cell lines. Mol Cancer. 2008; 7:15.
Article
45. Sandhu R, Rivenbark AG, Coleman WB. Enhancement of chemotherapeutic efficacy in hypermethylator breast cancer cells through targeted and pharmacologic inhibition of DNMT3b. Breast Cancer Res Treat. 2012; 131:385–399.
Article
46. Liu YR, Jiang YZ, Xu XE, Yu KD, Jin X, Hu X, et al. Comprehensive transcriptome analysis identifies novel molecular subtypes and subtype-specific RNAs of triple-negative breast cancer. Breast Cancer Res. 2016; 18:33.
Article
47. Shen X, Xie B, Ma Z, Yu W, Wang W, Xu D, et al. Identification of novel long non-coding RNAs in triple-negative breast cancer. Oncotarget. 2015; 6:21730–21739.
Article
48. Chen C, Li Z, Yang Y, Xiang T, Song W, Liu S. Microarray expression profiling of dysregulated long non-coding RNAs in triple-negative breast cancer. Cancer Biol Ther. 2015; 16:856–865.
Article
49. Jadaliha M, Zong X, Malakar P, Ray T, Singh DK, Freier SM, et al. Functional and prognostic significance of long non-coding RNA MALAT1 as a metastasis driver in ER negative lymph node negative breast cancer. Oncotarget. 2016; 7:40418–40436.
Article
50. Su X, Malouf GG, Chen Y, Zhang J, Yao H, Valero V, et al. Comprehensive analysis of long non-coding RNAs in human breast cancer clinical subtypes. Oncotarget. 2014; 5:9864–9876.
Article
51. Liu J, Shen L, Yao J, Li Y, Wang Y, Chen H, et al. Forkhead box C1 promoter upstream transcript, a novel long non-coding RNA, regulates proliferation and migration in basal-like breast cancer. Mol Med Rep. 2014; 11:3155–3159.
Article
52. Gupta RA, Shah N, Wang KC, Kim J, Horlings HM, Wong DJ, et al. Long non-coding RNA HOTAIR reprograms chromatin state to promote cancer metastasis. Nature. 2010; 464:1071–1076.
Article
53. Zhuang Y, Nguyen HT, Burow ME, Zhuo Y, El-Dahr SS, Yao X, et al. Elevated expression of long intergenic non-coding RNA HOTAIR in a basal-like variant of MCF-7 breast cancer cells. Mol Carcinog. 2015; 54:1656–1667.
Article
54. Wang YL, Overstreet AM, Chen MS, Wang J, Zhao HJ, Ho PC, et al. Combined inhibition of EGFR and c-ABL suppresses the growth of triple-negative breast cancer growth through inhibition of HOTAIR. Oncotarget. 2015; 6:11150–11161.
Article
55. Gasparini P, Cascione L, Fassan M, Lovat F, Guler G, Balci S, et al. MicroRNA expression profiling identifies a four microRNA signature as a novel diagnostic and prognostic biomarker in triple negative breast cancers. Oncotarget. 2014; 5:1174–1184.
Article
56. Fkih M'hamed I, Privat M, Ponelle F, Penault-Llorca F, Kenani A, Bignon YJ. Identification of miR-10b, miR-26a, miR-146a and miR-153 as potential triple-negative breast cancer biomarkers. Cell Oncol (Dordr). 2015; 38:433–442.
57. Kumaraswamy E, Wendt KL, Augustine LA, Stecklein SR, Sibala EC, Li D, et al. BRCA1 regulation of epidermal growth factor receptor (EGFR) expression in human breast cancer cells involves microRNA-146a and is critical for its tumor suppressor function. Oncogene. 2015; 34:4333–4346.
Article
58. Garcia AI, Buisson M, Bertrand P, Rimokh R, Rouleau E, Lopez BS, et al. Down-regulation of BRCA1 expression by miR-146a and miR-146b-5p in triple negative sporadic breast cancers. EMBO Mol Med. 2011; 3:279–290.
Article
59. Damiano V, Brisotto G, Borgna S, di Gennaro A, Armellin M, Perin T, et al. Epigenetic silencing of miR-200c in breast cancer is associated with aggressiveness and is modulated by ZEB1. Genes Chromosomes Cancer. 2017; 56:147–158.
Article
60. Humphries B, Wang Z, Oom AL, Fisher T, Tan D, Cui Y, et al. MicroRNA-200b targets protein kinase Cα and suppresses triple-negative breast cancer metastasis. Carcinogenesis. 2014; 35:2254–2263.
Article
61. Tsouko E, Wang J, Frigo DE, Aydoğdu E, Williams C. miR-200a inhibits migration of triple-negative breast cancer cells through direct repression of the EPHA2 oncogene. Carcinogenesis. 2015; 36:1051–1060.
Article
62. Li D, Wang H, Song H, Xu H, Zhao B, Wu C, et al. The microRNAs miR-200b-3p and miR-429-5p target the LIMK1/CFL1 pathway to inhibit growth and motility of breast cancer cells. Oncotarget. 2017; 8:85276–85289.
Article
63. Mathe A, Scott RJ, Avery-Kiejda KA. miRNAs and other epigenetic changes as biomarkers in triple negative breast cancer. Int J Mol Sci. 2015; 16:28347–28376.
Article
64. Xi Y, Shi J, Li W, Tanaka K, Allton KL, Richardson D, et al. Histone modification profiling in breast cancer cell lines highlights commonalities and differences among subtypes. BMC Genomics. 2018; 19:150.
Article
65. Khaled WT, Choon Lee S, Stingl J, Chen X, Raza Ali H, Rueda OM, et al. BCL11A is a triple-negative breast cancer gene with critical functions in stem and progenitor cells. Nat Commun. 2015; 6:5987.
Article
66. Moody RR, Lo MC, Meagher JL, Lin CC, Stevers NO, Tinsley SL, et al. Probing the interaction between the histone methyltransferase/deacetylase subunit RBBP4/7 and the transcription factor BCL11A in epigenetic complexes. J Biol Chem. 2018; 293:2125–2136.
Article
67. Ocaña A, Nieto-Jiménez C, Pandiella A. BET inhibitors as novel therapeutic agents in breast cancer. Oncotarget. 2017; 8:71285–71291.
Article
68. Filippakopoulos P, Knapp S. Targeting bromodomains: epigenetic readers of lysine acetylation. Nat Rev Drug Discov. 2014; 13:337–356.
Article
69. Sahai V, Redig AJ, Collier KA, Eckerdt FD, Munshi HG. Targeting BET bromodomain proteins in solid tumors. Oncotarget. 2016; 7:53997–54009.
Article
70. Shu S, Lin CY, He HH, Witwicki RM, Tabassum DP, Roberts JM, et al. Response and resistance to BET bromodomain inhibitors in triple-negative breast cancer. Nature. 2016; 529:413–417.
Article
71. Sahni JM, Gayle SS, Bonk KL, Vite LC, Yori JL, Webb B, et al. Bromodomain and extraterminal protein inhibition blocks growth of triple-negative breast cancers through the suppression of aurora kinases. J Biol Chem. 2016; 291:23756–23768.
Article
72. Nieto-Jiménez C, Alcaraz-Sanabria A, Pérez-Peña J, Corrales-Sánchez V, Serrano-Heras G, Galán-Moya EM, et al. Targeting basal-like breast tumors with bromodomain and extraterminal domain (BET) and pololike kinase inhibitors. Oncotarget. 2017; 8:19478–19490.
Article
73. Vázquez R, Riveiro ME, Astorgues-Xerri L, Odore E, Rezai K, Erba E, et al. The bromodomain inhibitor OTX015 (MK-8628) exerts anti-tumor activity in triple-negative breast cancer models as single agent and in combination with everolimus. Oncotarget. 2017; 8:7598–7613.
Article
74. Gregoire JM, Fleury L, Salazar-Cardozo C, Alby F, Masson V, Arimondo PB, et al. Identification of epigenetic factors regulating the mesenchyme to epithelium transition by RNA interference screening in breast cancer cells. BMC Cancer. 2016; 16:700.
Article
75. Lavigne AC, Castells M, Mermet J, Kocanova S, Dalvai M, Bystricky K. Increased macroH2A1.1 expression correlates with poor survival of triple-negative breast cancer patients. PLoS One. 2014; 9:e98930.
Article
76. Healey MA, Hu R, Beck AH, Collins LC, Schnitt SJ, Tamimi RM, et al. Association of H3K9me3 and H3K27me3 repressive histone marks with breast cancer subtypes in the Nurses' Health Study. Breast Cancer Res Treat. 2014; 147:639–651.
Article
77. Li G, Wang D, Ma W, An K, Liu Z, Wang X, et al. Transcriptomic and epigenetic analysis of breast cancer stem cells. Epigenomics. 2018; 10:765–783.
Article
78. Mottamal M, Zheng S, Huang TL, Wang G. Histone deacetylase inhibitors in clinical studies as templates for new anticancer agents. Molecules. 2015; 20:3898–3941.
Article
79. Wang ZT, Chen ZJ, Jiang GM, Wu YM, Liu T, Yi YM, et al. Histone deacetylase inhibitors suppress mutant p53 transcription via HDAC8/YY1 signals in triple negative breast cancer cells. Cell Signal. 2016; 28:506–515.
Article
80. Pileczki V, Pop L, Braicu C, Budisan L, Bolba Morar G, Del C Monroig-Bosque P, et al. Double gene siRNA knockdown of mutant p53 and TNF induces apoptosis in triple-negative breast cancer cells. Onco Targets Ther. 2016; 9:6921–6933.
Article
81. Braicu C, Pileczki V, Pop L, Petric RC, Chira S, Pointiere E, et al. Dual targeted therapy with p53 siRNA and epigallocatechingallate in a triple negative breast cancer cell model. PLoS One. 2015; 10:e0120936.
Article
82. Braicu C, Pileczki V, Irimie A, Berindan-Neagoe I. p53siRNA therapy reduces cell proliferation, migration and induces apoptosis in triple negative breast cancer cells. Mol Cell Biochem. 2013; 381:61–68.
Article
83. Tate CR, Rhodes LV, Segar HC, Driver JL, Pounder FN, Burow ME, et al. Targeting triple-negative breast cancer cells with the histone deacetylase inhibitor panobinostat. Breast Cancer Res. 2012; 14:R79.
Article
84. Min A, Im SA, Kim DK, Song SH, Kim HJ, Lee KH, et al. Histone deacetylase inhibitor, suberoylanilide hydroxamic acid (SAHA), enhances anti-tumor effects of the poly (ADP-ribose) polymerase (PARP) inhibitor olaparib in triple-negative breast cancer cells. Breast Cancer Res. 2015; 17:33.
Article
85. Topalian SL, Drake CG, Pardoll DM. Targeting the PD-1/B7-H1(PD-L1) pathway to activate anti-tumor immunity. Curr Opin Immunol. 2012; 24:207–212.
Article
86. Terranova-Barberio M, Thomas S, Ali N, Pawlowska N, Park J, Krings G, et al. HDAC inhibition potentiates immunotherapy in triple negative breast cancer. Oncotarget. 2017; 8:114156–114172.
Article
87. Schech A, Kazi A, Yu S, Shah P, Sabnis G. Histone deacetylase inhibitor entinostat inhibits tumor-initiating cells in triple-negative breast cancer cells. Mol Cancer Ther. 2015; 14:1848–1857.
Article
88. Palmieri D, Lockman PR, Thomas FC, Hua E, Herring J, Hargrave E, et al. Vorinostat inhibits brain metastatic colonization in a model of triple-negative breast cancer and induces DNA double-strand breaks. Clin Cancer Res. 2009; 15:6148–6157.
Article
89. Mekala JR, Naushad SM, Ponnusamy L, Arivazhagan G, Sakthiprasad V, Pal-Bhadra M. Epigenetic regulation of miR-200 as the potential strategy for the therapy against triple-negative breast cancer. Gene. 2018; 641:248–258.
Article
90. Zhu Y, Bai K, Yu J, Guo M. Association between histone methyltransferase hSETD1A and prognosis in patients with triple-negative breast cancer after surgery: a retrospective study in the Chinese female population. Medicine (Baltimore). 2016; 95:e3783.
91. Wu Q, Madany P, Akech J, Dobson JR, Douthwright S, Browne G, et al. The SWI/SNF ATPases are required for triple negative breast cancer cell proliferation. J Cell Physiol. 2015; 230:2683–2694.
Article
92. Wu Q, Madany P, Dobson JR, Schnabl JM, Sharma S, Smith TC, et al. The BRG1 chromatin remodeling enzyme links cancer cell metabolism and proliferation. Oncotarget. 2016; 7:38270–38281.
Article
93. Wu Q, Sharma S, Cui H, LeBlanc SE, Zhang H, Muthuswami R, et al. Targeting the chromatin remodeling enzyme BRG1 increases the efficacy of chemotherapy drugs in breast cancer cells. Oncotarget. 2016; 7:27158–27175.
Article
94. Festuccia C, Mancini A, Colapietro A, Gravina GL, Vitale F, Marampon F, et al. The first-in-class alkylating deacetylase inhibitor molecule tinostamustine shows antitumor effects and is synergistic with radiotherapy in preclinical models of glioblastoma. J Hematol Oncol. 2018; 11:32.
Article
Full Text Links
  • JBC
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr