Generic placeholder image

Current Molecular Medicine

Editor-in-Chief

ISSN (Print): 1566-5240
ISSN (Online): 1875-5666

Review Article

Epigenetic Reprogramming and Landscape of Transcriptomic Interactions: Impending Therapeutic Interference of Triple-Negative Breast Cancer in Molecular Medicine

Author(s): Suman Kumar Ray and Sukhes Mukherjee*

Volume 22, Issue 10, 2022

Published on: 14 January, 2022

Page: [835 - 850] Pages: 16

DOI: 10.2174/1566524021666211206092437

Price: $65

Abstract

The mechanisms governing the development and progression of cancers are believed to be the consequence of hereditary deformities and epigenetic modifications. Accordingly, epigenetics has become an incredible and progressively explored field of research to discover better prevention and therapy for neoplasia, especially triple-negative breast cancer (TNBC). It represents 15-20% of all invasive breast cancers and will, in general, have bellicose histological highlights and poor clinical outcomes. In the early phases of triple-negative breast carcinogenesis, epigenetic deregulation modifies chromatin structure and influences the plasticity of cells. It up-keeps the oncogenic reprogramming of malignant progenitor cells with the acquisition of unrestrained selfrenewal capacities. Genomic impulsiveness in TNBC prompts mutations, copy number variations, as well as genetic rearrangements, while epigenetic remodeling includes an amendment by DNA methylation, histone modification, and noncoding RNAs of gene expression profiles. It is currently evident that epigenetic mechanisms assume a significant part in the pathogenesis, maintenance, and therapeutic resistance of TNBC. Although TNBC is a heterogeneous malaise that is perplexing to describe and treat, the ongoing explosion of genetic and epigenetic research will help to expand these endeavors. Latest developments in transcriptome analysis have reformed our understanding of human diseases, including TNBC at the molecular medicine level. It is appealing to envision transcriptomic biomarkers to comprehend tumor behavior more readily regarding its cellular microenvironment. Understanding these essential biomarkers and molecular changes will propel our capability to treat TNBC adequately. This review will depict the different aspects of epigenetics and the landscape of transcriptomics in triple-negative breast carcinogenesis and their impending application for diagnosis, prognosis, and treatment decision with the view of molecular medicine.

Keywords: Epigenetic reprogramming, triple negative breast cancer, genomic impulsiveness, transcriptome, biomarkers, treatment decision.

Next »
[1]
Ferlay J, Colombet M, Soerjomataram I, et al. Estimating the global cancer incidence and mortality in 2018: GLOBOCAN sources and methods. Int J Cancer 2019; 144(8): 1941-53.
[http://dx.doi.org/10.1002/ijc.31937] [PMID: 30350310]
[2]
Ghoncheh M, Pournamdar Z, Salehiniya H. Incidence and mortality and epidemiology of breast cancer in the world. Asian Pac J Cancer Prev 2016; 17(S3): 43-6.
[http://dx.doi.org/10.7314/APJCP.2016.17.S3.43] [PMID: 27165206]
[3]
Torre LA, Islami F, Siegel RL, Ward EM, Jemal A. Global cancer in women: burden and trends. Cancer Epidemiol Biomarkers Prev 2017; 26(4): 444-57.
[http://dx.doi.org/10.1158/1055-9965.EPI-16-0858] [PMID: 28223433]
[4]
Gierach GL, Burke A, Anderson WF. Epidemiology of triple negative breast cancers. Breast Dis 2010; 32(1-2): 5-24.
[http://dx.doi.org/10.3233/BD-2010-0319] [PMID: 21965309]
[5]
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(6): 1026-33.
[http://dx.doi.org/10.1016/j.breast.2013.08.007] [PMID: 24063766]
[6]
Senkus E, Kyriakides S, Penault-Llorca F, et al. Primary breast cancer: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol 2013; 24(Suppl. 6): vi7-vi23.
[http://dx.doi.org/10.1093/annonc/mdt284] [PMID: 23970019]
[7]
Lachapelle J, Foulkes WD. Triple-negative and basal-like breast cancer: Implications for oncologists. Curr Oncol 2011; 18(4): 161-4.
[http://dx.doi.org/10.3747/co.v18i4.824] [PMID: 21874112]
[8]
Ray SK, Mukherjee S. Current headway in cancer immunotherapy emphasising the practice of genetically engineered T-cells to target selected tumor antigen. Crit Rev Immunol 2021; 41(1): 23-40.
[http://dx.doi.org/10.1615/CritRevImmunol.2020037044] [PMID: 33822523]
[9]
Ray SK, Meshram Y, Mukherjee S. Cancer immunology and CAR-T cells: a turning point therapeutic approach in cancer treatment focusing on colorectal carcinoma with clinical landscape. Curr Mol Med 2021; 21(3): 221-36.
[http://dx.doi.org/10.2174/1566524020666200824103749] [PMID: 32838717]
[10]
Foulkes WD, Smith IE, Reis-Filho JS. Triple-negative breast cancer. N Engl J Med 2010; 363(20): 1938-48.
[http://dx.doi.org/10.1056/NEJMra1001389] [PMID: 21067385]
[11]
Lehmann BD, Pietenpol JA. Identification and use of biomarkers in treatment strategies for triple-negative breast cancer subtypes. J Pathol 2014; 232(2): 142-50.
[http://dx.doi.org/10.1002/path.4280] [PMID: 24114677]
[12]
Perou CM. Molecular stratification of triple-negative breast cancers. Oncologist 2011; 16(Suppl. 1): 61-70.
[http://dx.doi.org/10.1634/theoncologist.2011-S1-61] [PMID: 21278442]
[13]
Fedele M, Cerchia L, Chiappetta G. The epithelial-to-mesenchymal transition in breast cancer: focus on basal-like carcinomas. Cancers (Basel) 2017; 9(10): 134.
[http://dx.doi.org/10.3390/cancers9100134] [PMID: 28974015]
[14]
Moreno-Bueno G, Portillo F, Cano A. Transcriptional regulation of cell polarity in EMT and cancer. Oncogene 2008; 27(55): 6958-69.
[http://dx.doi.org/10.1038/onc.2008.346] [PMID: 19029937]
[15]
Sundararajan V, Tan M, Tan TZ, Ye J, Thiery JP, Huang RY. SNAI1 recruits HDAC1 to suppress SNAI2 transcription during epithelial to mesenchymal transition. Sci Rep 2019; 9(1): 8295.
[http://dx.doi.org/10.1038/s41598-019-44826-8] [PMID: 31165775]
[16]
Xu R, Won JY, Kim CH, Kim DE, Yim H. Roles of the phosphorylation of transcriptional factors in epithelial-mesenchymal transition. J Oncol 2019; 2019: 5810465.
[http://dx.doi.org/10.1155/2019/5810465] [PMID: 31275381]
[17]
Muhammad N, Bhattacharya S, Steele R, Phillips N, Ray RB. Involvement of c-Fos in the promotion of cancer stem-like cell properties in head and neck squamous cell carcinoma. Clin Cancer Res 2017; 23(12): 3120-8.
[http://dx.doi.org/10.1158/1078-0432.CCR-16-2811] [PMID: 27965308]
[18]
Hu X, Harvey SE, Zheng R, et al. The RNA-binding protein AKAP8 suppresses tumor metastasis by antagonizing EMT-associated alternative splicing. Nat Commun 2020; 11(1): 486.
[http://dx.doi.org/10.1038/s41467-020-14304-1] [PMID: 31980632]
[19]
Ebright RY, Lee S, Wittner BS, Niederhoffer KL, Nicholson BT, Bardia A. Deregulation of ribosomal protein expression and translation promotes breast cancer metastasis. Science 2020; 367(6485): 1468-73.
[http://dx.doi.org/10.1126/science.aay0939]
[20]
Wu X, Zhang X, Yu L, et al. Zinc finger protein 367 promotes metastasis by inhibiting the Hippo pathway in breast cancer. Oncogene 2020; 39(12): 2568-82.
[http://dx.doi.org/10.1038/s41388-020-1166-y] [PMID: 31988454]
[21]
Kelly AD, Issa JJ. The promise of epigenetic therapy: Reprogramming the cancer epigenome. Curr Opin Genet Dev 2017; 42: 68-77.
[http://dx.doi.org/10.1016/j.gde.2017.03.015] [PMID: 28412585]
[22]
Kamps R, Brandão RD, Bosch BJ, et al. Next-generation sequencing in oncology: genetic diagnosis, risk prediction and cancer classification. Int J Mol Sci 2017; 18(2): 308.
[http://dx.doi.org/10.3390/ijms18020308] [PMID: 28146134]
[23]
Lehmann BD, Bauer JA, Chen X, et al. Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies. J Clin Invest 2011; 121(7): 2750-67.
[http://dx.doi.org/10.1172/JCI45014] [PMID: 21633166]
[24]
Lehmann BD. Jovanović B, Chen X, et al. Refinement of triple-negative breast cancer molecular subtypes: Implications for neoadjuvant chemotherapy selection. PLoS One 2016; 11(6): e0157368.
[http://dx.doi.org/10.1371/journal.pone.0157368] [PMID: 27310713]
[25]
Burstein MD, Tsimelzon A, Poage GM, et al. Comprehensive genomic analysis identifies novel subtypes and targets of triple-negative breast cancer. Clin Cancer Res 2015; 21(7): 1688-98.
[http://dx.doi.org/10.1158/1078-0432.CCR-14-0432] [PMID: 25208879]
[26]
Jézéquel P, Loussouarn D, Guérin-Charbonnel C, et al. Gene-expression molecular subtyping of triple-negative breast cancer tumours: Importance of immune response. Breast Cancer Res 2015; 17: 43.
[http://dx.doi.org/10.1186/s13058-015-0550-y] [PMID: 25887482]
[27]
Fragomeni SM, Sciallis A, Jeruss JS. Molecular subtypes and local-regional control of breast cancer. Surg Oncol Clin N Am 2018; 27(1): 95-120.
[http://dx.doi.org/10.1016/j.soc.2017.08.005] [PMID: 29132568]
[28]
Yin L, Duan JJ, Bian XW, Yu SC. Triple-negative breast cancer molecular subtyping and treatment progress. Breast Cancer Res 2020; 22(1): 61.
[http://dx.doi.org/10.1186/s13058-020-01296-5] [PMID: 32517735]
[29]
Kumar N, Zhao D, Bhaumik D, Sethi A, Gann PH. Quantification of intrinsic subtype ambiguity in luminal a breast cancer and its relationship to clinical outcomes. BMC Cancer 2019; 19(1): 215.
[http://dx.doi.org/10.1186/s12885-019-5392-z] [PMID: 30849944]
[30]
Russnes HG, Lingjærde OC, Børresen-Dale AL, Caldas C. Breast cancer molecular stratification: from intrinsic subtypes to integrative clusters. Am J Pathol 2017; 187(10): 2152-62.
[http://dx.doi.org/10.1016/j.ajpath.2017.04.022] [PMID: 28733194]
[31]
Wang DY, Jiang Z, Ben-David Y, Woodgett JR, Zacksenhaus E. Molecular stratification within triple-negative breast cancer subtypes. Sci Rep 2019; 9(1): 19107.
[http://dx.doi.org/10.1038/s41598-019-55710-w] [PMID: 31836816]
[32]
Herschkowitz JI, Zhao W, Zhang M, et al. Comparative oncogenomics identifies breast tumors enriched in functional tumor-initiating cells. Proc Natl Acad Sci USA 2012; 109(8): 2778-83.
[http://dx.doi.org/10.1073/pnas.1018862108] [PMID: 21633010]
[33]
Netanely D, Avraham A, Ben-Baruch A, Evron E, Shamir R. Expression and methylation patterns partition luminal-A breast tumors into distinct prognostic subgroups. Breast Cancer Res 2016; 18(1): 74.
[http://dx.doi.org/10.1186/s13058-016-0724-2] [PMID: 27386846]
[34]
Noberini R, Restellini C, Savoia EO, et al. Profiling of epigenetic features in clinical samples reveals novel widespread changes in cancer. Cancers (Basel) 2019; 11(5): 723.
[http://dx.doi.org/10.3390/cancers11050723] [PMID: 31137727]
[35]
Parrella P. The value of epigenetic biomarkers in breast cancer. Biomarkers Med 2018; 12(9): 937-40.
[http://dx.doi.org/10.2217/bmm-2018-0187] [PMID: 30041537]
[36]
Zolota V, Tzelepi V, Piperigkou Z, et al. Epigenetic alterations in triple-negative breast cancer-the critical role of extracellular matrix. Cancers (Basel) 2021; 13(4): 713.
[http://dx.doi.org/10.3390/cancers13040713] [PMID: 33572395]
[37]
Pearson GW. Control of Invasion by epithelial-to-mesenchymal transition programs during metastasis. J Clin Med 2019; 8(5): 646.
[http://dx.doi.org/10.3390/jcm8050646] [PMID: 31083398]
[38]
Banyard J, Bielenberg DR. The role of EMT and MET in cancer dissemination. Connect Tissue Res 2015; 56(5): 403-13.
[http://dx.doi.org/10.3109/03008207.2015.1060970] [PMID: 26291767]
[39]
Yang L, Shi P, Zhao G, et al. Targeting cancer stem cell pathways for cancer therapy. Signal Transduct Target Ther 2020; 5(1): 8.
[http://dx.doi.org/10.1038/s41392-020-0110-5] [PMID: 32296030]
[40]
Feng Y, Spezia M, Huang S, et al. Breast cancer development and progression: risk factors, cancer stem cells, signaling pathways, genomics, and molecular pathogenesis. Genes Dis 2018; 5(2): 77-106.
[http://dx.doi.org/10.1016/j.gendis.2018.05.001] [PMID: 30258937]
[41]
Simeone P, Trerotola M, Franck J, et al. The multiverse nature of epithelial to mesenchymal transition. Semin Cancer Biol 2019; 58: 1-10.
[http://dx.doi.org/10.1016/j.semcancer.2018.11.004] [PMID: 30453041]
[42]
Moyret-Lalle C, Ruiz E, Puisieux A. Epithelial-mesenchymal transition transcription factors and miRNAs: “Plastic surgeons” of breast cancer. World J Clin Oncol 2014; 5(3): 311-22.
[http://dx.doi.org/10.5306/wjco.v5.i3.311] [PMID: 25114847]
[43]
Liu F, Gu LN, Shan BE, Geng CZ, Sang MX. Biomarkers for EMT and MET in breast cancer: an update. Oncol Lett 2016; 12(6): 4869-76.
[http://dx.doi.org/10.3892/ol.2016.5369] [PMID: 28105194]
[44]
Gómez Tejeda Zañudo J, Guinn MT, Farquhar K, et al. Towards control of cellular decision-making networks in the epithelial-to-mesenchymal transition. Phys Biol 2019; 16(3): 031002.
[http://dx.doi.org/10.1088/1478-3975/aaffa1] [PMID: 30654341]
[45]
Kaszak I. Witkowska-Piłaszewicz O, Niewiadomska Z, Dworecka-Kaszak B, Ngosa Toka F, Jurka P. Role of cadherins in cancer-a review. Int J Mol Sci 2020; 21(20): 7624.
[http://dx.doi.org/10.3390/ijms21207624] [PMID: 33076339]
[46]
Medici D, Hay ED, Olsen BR. Snail and slug promote epithelial-mesenchymal transition through beta-catenin-T-cell factor-4-dependent expression of transforming growth factor-beta3. Mol Biol Cell 2008; 19(11): 4875-87.
[http://dx.doi.org/10.1091/mbc.e08-05-0506] [PMID: 18799618]
[47]
Kourtidis A, Lu R, Pence LJ, Anastasiadis PZ. A central role for cadherin signaling in cancer. Exp Cell Res 2017; 358(1): 78-85.
[http://dx.doi.org/10.1016/j.yexcr.2017.04.006] [PMID: 28412244]
[48]
Fintha A, Gasparics Á, Rosivall L, Sebe A. Therapeutic targeting of fibrotic epithelial-mesenchymal transition-an outstanding challenge. Front Pharmacol 2019; 10: 388.
[http://dx.doi.org/10.3389/fphar.2019.00388] [PMID: 31057405]
[49]
Xu Y, Zhou X, Mei M, Ren Y. Reprograming carcinoma associated fibroblasts by microRNAs. Curr Mol Med 2017; 17(5): 341-9.
[http://dx.doi.org/10.2174/1566524018666171205113959] [PMID: 29210650]
[50]
Tagawa H, Ikeda S, Sawada K. Role of microRNA in the pathogenesis of malignant lymphoma. Cancer Sci 2013; 104(7): 801-9.
[http://dx.doi.org/10.1111/cas.12160] [PMID: 23551855]
[51]
Wang J, Zhang Y, Wei H, et al. The mir-675-5p regulates the progression and development of pancreatic cancer via the UBQLN1-ZEB1-mir200 axis. Oncotarget 2017; 8(15): 24978-87.
[http://dx.doi.org/10.18632/oncotarget.15330] [PMID: 28212565]
[52]
Korpal M, Kang Y. The emerging role of miR-200 family of microRNAs in epithelial-mesenchymal transition and cancer metastasis. RNA Biol 2008; 5(3): 115-9.
[http://dx.doi.org/10.4161/rna.5.3.6558] [PMID: 19182522]
[53]
Feng YH, Tsao CJ. Emerging role of microRNA-21 in cancer. Biomed Rep 2016; 5(4): 395-402.
[http://dx.doi.org/10.3892/br.2016.747] [PMID: 27699004]
[54]
Poulos RC, Olivier J, Wong JWH. The interaction between cytosine methylation and processes of DNA replication and repair shape the mutational landscape of cancer genomes. Nucleic Acids Res 2017; 45(13): 7786-95.
[http://dx.doi.org/10.1093/nar/gkx463] [PMID: 28531315]
[55]
Jovanovic J, Rønneberg JA, Tost J, Kristensen V. The epigenetics of breast cancer Molecular Oncology 2010; 4: 242e254.
[http://dx.doi.org/10.1016/j.molonc.2010.04.002]
[56]
Cheng Y, He C, Wang M, et al. Targeting epigenetic regulators for cancer therapy: mechanisms and advances in clinical trials. Signal Transduct Target Ther 2019; 4: 62.
[http://dx.doi.org/10.1038/s41392-019-0095-0] [PMID: 31871779]
[57]
Casalino L, Verde P. Multifaceted roles of DNA methylation in neoplastic transformation, from tumor suppressors to EMT and metastasis. Genes (Basel) 2020; 11(8): 922.
[http://dx.doi.org/10.3390/genes11080922] [PMID: 32806509]
[58]
Zhang J, Yang C, Wu C, Cui W, Wang L. DNA Methyltransferases in cancer: Biology, paradox, aberrations, and targeted therapy. Cancers (Basel) 2020; 12(8): 2123.
[http://dx.doi.org/10.3390/cancers12082123] [PMID: 32751889]
[59]
Gujar H, Weisenberger DJ, Liang G. The roles of human DNA methyltransferases and their isoforms in shaping the epigenome. Genes (Basel) 2019; 10(2): 172.
[http://dx.doi.org/10.3390/genes10020172] [PMID: 30813436]
[60]
Hannen R, Bartsch JW. Essential roles of telomerase reverse transcriptase hTERT in cancer stemness and metastasis. FEBS Lett 2018; 592(12): 2023-31.
[http://dx.doi.org/10.1002/1873-3468.13084] [PMID: 29749098]
[61]
Hill VK, Ricketts C, Bieche I, et al. Genome-wide DNA methylation profiling of CpG islands in breast cancer identifies novel genes associated with tumorigenicity. Cancer Res 2011; 71(8): 2988-99.
[http://dx.doi.org/10.1158/0008-5472.CAN-10-4026] [PMID: 21363912]
[62]
Xi Y, Shi J, Li W, et al. Histone modification profiling in breast cancer cell lines highlights commonalities and differences among subtypes. BMC Genomics 2018; 19(1): 150.
[http://dx.doi.org/10.1186/s12864-018-4533-0] [PMID: 29458327]
[63]
Rajan PK, Udoh UA, Sanabria JD, et al. The role of histone acetylation-/methylation-mediated apoptotic gene regulation in hepatocellular carcinoma. Int J Mol Sci 2020; 21(23): 8894.
[http://dx.doi.org/10.3390/ijms21238894] [PMID: 33255318]
[64]
Basse C, Arock M. The increasing roles of epigenetics in breast cancer: implications for pathogenicity, biomarkers, prevention and treatment. Int J Cancer 2015; 137(12): 2785-94.
[http://dx.doi.org/10.1002/ijc.29347] [PMID: 25410431]
[65]
Jia YM, Xie YT, Wang YJ, Han JY, Tian XX, Fang WG. Association of genetic polymorphisms in CDH1 and CTNNB1 with breast cancer susceptibility and patients’ prognosis among Chinese Han women. PLoS One 2015; 10(8): e0135865.
[http://dx.doi.org/10.1371/journal.pone.0135865] [PMID: 26285011]
[66]
Nandy D, Rajam SM, Dutta D. A three layered histone epigenetics in breast cancer metastasis. Cell Biosci 2020; 10: 52.
[http://dx.doi.org/10.1186/s13578-020-00415-1] [PMID: 32257110]
[67]
Nakagawa H, Fujita M. Whole genome sequencing analysis for cancer genomics and precision medicine. Cancer Sci 2018; 109(3): 513-22.
[http://dx.doi.org/10.1111/cas.13505] [PMID: 29345757]
[68]
Rheinbay E, Nielsen MM, Abascal F, et al. Analyses of non-coding somatic drivers in 2,658 cancer whole genomes. Nature 2020; 578(7793): 102-11.
[http://dx.doi.org/10.1038/s41586-020-1965-x] [PMID: 32025015]
[69]
Rajendran BK, Deng CX. Characterization of potential driver mutations involved in human breast cancer by computational approaches. Oncotarget 2017; 8(30): 50252-72.
[http://dx.doi.org/10.18632/oncotarget.17225] [PMID: 28477017]
[70]
Shi Y, Jin J, Ji W, Guan X. Therapeutic landscape in mutational triple negative breast cancer. Mol Cancer 2018; 17(1): 99.
[http://dx.doi.org/10.1186/s12943-018-0850-9] [PMID: 30007403]
[71]
Byler S, Goldgar S, Heerboth S, et al. Genetic and epigenetic aspects of breast cancer progression and therapy. Anticancer Res 2014; 34(3): 1071-7.
[PMID: 24596345]
[72]
Curtis C, Shah SP, Chin SF, et al. The genomic and transcriptomic architecture of 2,000 breast tumours reveals novel subgroups. Nature 2012; 486(7403): 346-52.
[http://dx.doi.org/10.1038/nature10983] [PMID: 22522925]
[73]
The Cancer Genome Atlas Network Comprehensive molecular portraits of human breast tumours. Nature 2012; 490: 61-70.
[http://dx.doi.org/10.1038/nature11412]
[74]
Deshpande V, Luebeck J, Nguyen ND, et al. Exploring the landscape of focal amplifications in cancer using ampliconarchitect. Nat Commun 2019; 10(1): 392.
[http://dx.doi.org/10.1038/s41467-018-08200-y] [PMID: 30674876]
[75]
Medina-Jaime AD, Reyes-Vargas F, Martinez-Gaytan V, et al. ESR1 and PGR gene promoter methylation and correlations with estrogen and progesterone receptors in ductal and lobular breast cancer. Asian Pac J Cancer Prev 2014; 15(7): 3041-4.
[http://dx.doi.org/10.7314/APJCP.2014.15.7.3041] [PMID: 24815444]
[76]
Setiawan VW, Monroe KR, Wilkens LR, Kolonel LN, Pike MC, Henderson BE. Breast cancer risk factors defined by estrogen and progesterone receptor status: the multiethnic cohort study. Am J Epidemiol 2009; 169(10): 1251-9.
[http://dx.doi.org/10.1093/aje/kwp036] [PMID: 19318616]
[77]
Sun Z, Asmann YW, Kalari KR, et al. Integrated analysis of gene expression, CpG island methylation, and gene copy number in breast cancer cells by deep sequencing. PLoS One 2011; 6(2): e17490.
[http://dx.doi.org/10.1371/journal.pone.0017490] [PMID: 21364760]
[78]
Di Leva G, Gasparini P, Piovan C, et al. MicroRNA cluster 221-222 and estrogen receptor α interactions in breast cancer. J Natl Cancer Inst 2010; 102(10): 706-21.
[http://dx.doi.org/10.1093/jnci/djq102] [PMID: 20388878]
[79]
Howard EW, Yang X. microRNA regulation in estrogen receptor-positive breast cancer and endocrine therapy. Biol Proced Online 2018; 20: 17.
[http://dx.doi.org/10.1186/s12575-018-0082-9] [PMID: 30214383]
[80]
Song Q, An Q, Niu B, Lu X, Zhang N, Cao X. Role of miR-221/222 in tumor development and the underlying mechanism. J Oncol 2019; 2019: 7252013.
[http://dx.doi.org/10.1155/2019/7252013] [PMID: 31929798]
[81]
Kawazu M, Saso K, Tong KI, et al. Histone demethylase JMJD2B functions as a co-factor of estrogen receptor in breast cancer proliferation and mammary gland development. PLoS One 2011; 6(3): e17830.
[http://dx.doi.org/10.1371/journal.pone.0017830] [PMID: 21445275]
[82]
Zhao Z, Shilatifard A. Epigenetic modifications of histones in cancer. Genome Biol 2019; 20(1): 245.
[http://dx.doi.org/10.1186/s13059-019-1870-5] [PMID: 31747960]
[83]
Idrissou M, Sanchez A, Penault-Llorca F, Bignon YJ, Bernard-Gallon D. Epi-drugs as triple-negative breast cancer treatment. Epigenomics 2020; 12(8): 725-42.
[http://dx.doi.org/10.2217/epi-2019-0312] [PMID: 32396394]
[84]
Kong WY, Yee ZY, Mai CW, Fang CM, Abdullah S, Ngai SC. Zebularine and trichostatin A sensitized human breast adenocarcinoma cells towards tumor necrosis factor-related apoptosis inducing ligand (TRAIL)-induced apoptosis. Heliyon 2019; 5(9): e02468.
[http://dx.doi.org/10.1016/j.heliyon.2019.e02468] [PMID: 31687564]
[85]
Billam M, Sobolewski MD, Davidson NE. Effects of a novel DNA methyltransferase inhibitor zebularine on human breast cancer cells. Breast Cancer Res Treat 2010; 120(3): 581-92.
[http://dx.doi.org/10.1007/s10549-009-0420-3] [PMID: 19459041]
[86]
Sheng J, Shi W, Guo H, et al. The inhibitory effect of (-)-epigallocatechin-3-gallate on breast cancer progression via reducing SCUBE2 methylation and DNMT activity. Molecules 2019; 24(16): 2899.
[http://dx.doi.org/10.3390/molecules24162899] [PMID: 31404982]
[87]
Cai FF, Kohler C, Zhang B, Wang MH, Chen WJ, Zhong XY. Epigenetic therapy for breast cancer. Int J Mol Sci 2011; 12(7): 4465-87.
[http://dx.doi.org/10.3390/ijms12074465] [PMID: 21845090]
[88]
Jenke R, Reßing N, Hansen FK, Aigner A, Büch T. Anticancer therapy with HDAC inhibitors: Mechanism-based combination strategies and future perspectives. Cancers (Basel) 2021; 13(4): 634.
[http://dx.doi.org/10.3390/cancers13040634] [PMID: 33562653]
[89]
Thurn KT, Thomas S, Moore A, Munster PN. Rational therapeutic combinations with histone deacetylase inhibitors for the treatment of cancer. Future Oncol 2011; 7(2): 263-83.
[http://dx.doi.org/10.2217/fon.11.2] [PMID: 21345145]
[90]
Suraweera A, O’Byrne KJ, Richard DJ. Combination Therapy with histone deacetylase inhibitors (HDACi) for the treatment of cancer: achieving the full therapeutic potential of HDACi. Front Oncol 2018; 8: 92.
[http://dx.doi.org/10.3389/fonc.2018.00092] [PMID: 29651407]
[91]
Bezu L, Chuang AW, Liu P, Kroemer G, Kepp O. Immunological effects of epigenetic modifiers. Cancers (Basel) 2019; 11(12): 1911.
[http://dx.doi.org/10.3390/cancers11121911] [PMID: 31805711]
[92]
Jezkova E, Zubor P, Kajo K, et al. Impact of RASSF1A gene methylation on the metastatic axillary nodal status in breast cancer patients. Oncol Lett 2017; 14(1): 758-66.
[http://dx.doi.org/10.3892/ol.2017.6204] [PMID: 28693231]
[93]
Wang X, Liu Y, Sun H, et al. DNA Methylation in RARβ gene as a mediator of the association between healthy lifestyle and breast cancer: a case-control study. Cancer Manag Res 2020; 12: 4677-84.
[http://dx.doi.org/10.2147/CMAR.S244606] [PMID: 32606959]
[94]
Aubele M, Schmitt M, Napieralski R, et al. The predictive value of PITX2 DNA methylation for high-risk breast cancer therapy: Current guidelines, medical needs, and challenges. Dis Markers 2017; 2017: 4934608.
[http://dx.doi.org/10.1155/2017/4934608] [PMID: 29138528]
[95]
Subhash S, Kanduri M. Comprehensive DNA methylation analysis using a methyl-CpG-binding domain capture-based method in chronic lymphocytic leukemia patients. J Vis Exp 2017; (124): 55773.
[http://dx.doi.org/10.3791/55773] [PMID: 28654059]
[96]
Fang F, Turcan S, Rimner A, et al. Breast cancer methylomes establish an epigenomic foundation for metastasis. Sci Transl Med 2011; 3(75): 75ra25.
[http://dx.doi.org/10.1126/scitranslmed.3001875] [PMID: 21430268]
[97]
Yu JR, Lee CH, Oksuz O, Stafford JM, Reinberg D. PRC2 is high maintenance. Genes Dev 2019; 33(15-16): 903-35.
[http://dx.doi.org/10.1101/gad.325050.119] [PMID: 31123062]
[98]
Trimboli RM, Giorgi Rossi P, Battisti NML, et al. Do we still need breast cancer screening in the era of targeted therapies and precision medicine? Insights Imaging 2020; 11(1): 105.
[http://dx.doi.org/10.1186/s13244-020-00905-3] [PMID: 32975658]
[99]
Pinker K, Chin J, Melsaether AN, Morris EA, Moy L. Precision medicine and radiogenomics in breast cancer: new approaches toward diagnosis and treatment. Radiology 2018; 287(3): 732-47.
[http://dx.doi.org/10.1148/radiol.2018172171] [PMID: 29782246]
[100]
Krzyszczyk P, Acevedo A, Davidoff EJ, et al. The growing role of precision and personalized medicine for cancer treatment. Technology (Singap World Sci) 2018; 6(3-4): 79-100.
[http://dx.doi.org/10.1142/S2339547818300020] [PMID: 30713991]
[101]
Hosseini A, Khoury AL, Esserman LJ. Precision surgery and avoiding over-treatment. Eur J Surg Oncol 2017; 43(5): 938-43.
[http://dx.doi.org/10.1016/j.ejso.2017.02.003] [PMID: 28238520]
[102]
Gnant M, Sestak I, Filipits M, et al. Identifying clinically relevant prognostic subgroups of postmenopausal women with node-positive hormone receptor-positive early-stage breast cancer treated with endocrine therapy: a combined analysis of ABCSG-8 and ATAC using the PAM50 risk of recurrence score and intrinsic subtype. Ann Oncol 2015; 26(8): 1685-91.
[http://dx.doi.org/10.1093/annonc/mdv215] [PMID: 25935792]
[103]
Cardoso F, van’t Veer LJ, Bogaerts J, et al. 70-gene signature as an aid to treatment decisions in early-stage breast cancer. N Engl J Med 2016; 375(8): 717-29.
[http://dx.doi.org/10.1056/NEJMoa1602253] [PMID: 27557300]
[104]
Sparano JA, Gray RJ, Makower DF, et al. Adjuvant chemotherapy guided by a 21-gene expression assay in breast cancer. N Engl J Med 2018; 379(2): 111-21.
[http://dx.doi.org/10.1056/NEJMoa1804710] [PMID: 29860917]
[105]
Curigliano G, Burstein HJ, Winer EP, et al. Deescalating and escalating treatments for early-stage breast cancer: the St. Gallen international expert consensus conference on the primary therapy of early breast cancer. Ann Oncol 28(8): 1700-12.
[http://dx.doi.org/10.1093/annonc/mdx308] [PMID: 28838210]
[106]
Ohnstad HO, Borgen E, Falk RS, et al. Prognostic value of PAM50 and risk of recurrence score in patients with early-stage breast cancer with long-term follow-up. Breast Cancer Res 2017; 19(1): 120.
[http://dx.doi.org/10.1186/s13058-017-0911-9] [PMID: 29137653]
[107]
Esserman LJ, Yau C, Thompson CK, et al. Use of molecular tools to identify patients with indolent breast cancers with ultralow risk over 2 decades. JAMA Oncol 2017; 3(11): 1503-10.
[http://dx.doi.org/10.1001/jamaoncol.2017.1261] [PMID: 28662222]
[108]
Wang ZT, Chen ZJ, Jiang GM, et al. Histone deacetylase inhibitors suppress mutant p53 transcription via HDAC8/YY1 signals in triple negative breast cancer cells. Cell Signal 2016; 28(5): 506-15.
[http://dx.doi.org/10.1016/j.cellsig.2016.02.006] [PMID: 26876786]
[109]
Peixoto P, Grandvallet C, Feugeas JP, Guittaut M, Hervouet E. Epigenetic control of autophagy in cancer cells: a key process for cancer-related phenotypes. Cells 2019; 8(12): 1656.
[http://dx.doi.org/10.3390/cells8121656] [PMID: 31861179]
[110]
Temian DC, Pop LA, Irimie AI, Berindan-Neagoe I. The epigenetics of triple-negative and basal-like breast cancer: current knowledge. J Breast Cancer 2018; 21(3): 233-43.
[http://dx.doi.org/10.4048/jbc.2018.21.e41] [PMID: 30275851]
[111]
Ediriweera MK, Tennekoon KH, Samarakoon SR. Emerging role of histone deacetylase inhibitors as anti-breast-cancer agents. Drug Discov Today 2019; 24(3): 685-702.
[http://dx.doi.org/10.1016/j.drudis.2019.02.003] [PMID: 30776482]
[112]
Garmpis N, Damaskos C, Garmpi A, et al. Histone deacetylases as new therapeutic targets in triple-negative breast cancer: progress and promises. Cancer Genomics Proteomics 2017; 14(5): 299-313.
[http://dx.doi.org/10.21873/cgp.20041] [PMID: 28870998]
[113]
Pasculli B, Barbano R, Parrella P. Epigenetics of breast cancer: Biology and clinical implication in the era of precision medicine. Semin Cancer Biol 2018; 51: 22-35.
[http://dx.doi.org/10.1016/j.semcancer.2018.01.007] [PMID: 29339244]
[114]
Li Y, Seto E. HDACs and HDAC Inhibitors in cancer development and therapy. Cold Spring Harb Perspect Med 2016; 6(10): a026831.
[http://dx.doi.org/10.1101/cshperspect.a026831] [PMID: 27599530]
[115]
Yardley DA, Ismail-Khan RR, Melichar B, et al. Randomized phase II, double-blind, placebo-controlled study of exemestane with or without entinostat in postmenopausal women with locally recurrent or metastatic estrogen receptor-positive breast cancer progressing on treatment with a nonsteroidal aromatase inhibitor. J Clin Oncol 2013; 31(17): 2128-35.
[http://dx.doi.org/10.1200/JCO.2012.43.7251] [PMID: 23650416]
[116]
Connolly RM, Li H, Jankowitz RC, et al. Combination epigenetic therapy in advanced breast cancer with 5-azacitidine and entinostat: a phase ii national cancer institute/stand up to cancer study. Clin Cancer Res 2017; 23(11): 2691-701.
[http://dx.doi.org/10.1158/1078-0432.CCR-16-1729] [PMID: 27979916]
[117]
Chalakur-Ramireddy NKR, Pakala SB. Combined drug therapeutic strategies for the effective treatment of triple negative breast cancer. Biosci Rep 2018; 38(1): BSR20171357.
[http://dx.doi.org/10.1042/BSR20171357] [PMID: 29298879]
[118]
Terranova-Barberio M, Thomas S, Ali N, et al. HDAC inhibition potentiates immunotherapy in triple negative breast cancer. Oncotarget 2017; 8(69): 114156-72.
[http://dx.doi.org/10.18632/oncotarget.23169] [PMID: 29371976]
[119]
Ray SK, Mukherjee S. LncRNAs as new architects in cancer biomarkers, and potential therapeutic targets in addition to interface with epitranscriptomics: is incipient targets in cancer? Curr Cancer Drug Targets 2021; 21(5): 416-27.
[http://dx.doi.org/10.2174/1568009620666210106122421] [PMID: 33413062]
[120]
Ray SK, Mukherjee S. Cell free DNA as an evolving liquid biopsy biomarker for initial diagnosis and therapeutic nursing in cancer- an evolving aspect in medical biotechnology. Curr Pharm Biotechnol 2020.
[http://dx.doi.org/10.2174/1389201021666201211102710] [PMID: 33308128]
[121]
Ray SK, Mukherjee S. Cancer stem cells: Current status and therapeutic implications in cancer therapy- a new paradigm. Curr Stem Cell Res Ther 2021; 16(8): 970-9.
[http://dx.doi.org/10.2174/1574888X16666210203105800] [PMID: 33563175]
[122]
Shaheed SU, Tait C, Kyriacou K, Linforth R, Salhab M, Sutton C. Evaluation of nipple aspirate fluid as a diagnostic tool for early detection of breast cancer. Clin Proteomics 2018; 15: 3.
[http://dx.doi.org/10.1186/s12014-017-9179-4] [PMID: 29344009]
[123]
Salta S, Nunes PS, Fontes-Sousa M, et al. A DNA methylation-based test for breast cancer detection in circulating cell-free DNA. J Clin Med 2018; 7(11): 420.
[http://dx.doi.org/10.3390/jcm7110420] [PMID: 30405052]
[124]
Cao X, Tang Q, Holland-Letz T, et al. Evaluation of promoter methylation of RASSF1A and ATM in peripheral blood of breast cancer patients and healthy control individuals. Int J Mol Sci 2018; 19(3): 900.
[http://dx.doi.org/10.3390/ijms19030900] [PMID: 29562656]
[125]
Shah R, Smith P, Purdie C, et al. The prolyl 3-hydroxylases P3H2 and P3H3 are novel targets for epigenetic silencing in breast cancer. Br J Cancer 2009; 100(10): 1687-96.
[http://dx.doi.org/10.1038/sj.bjc.6605042] [PMID: 19436308]
[126]
Lee HJ, An HJ, Kim TH, et al. Fascin expression is inversely correlated with breast cancer metastasis suppressor 1 and predicts a worse survival outcome in node-negative breast cancer patients. J Cancer 2017; 8(16): 3122-9.
[http://dx.doi.org/10.7150/jca.22046] [PMID: 29158783]
[127]
Puhalla S, Bhattacharya S, Davidson NE. Hormonal therapy in breast cancer: a model disease for the personalization of cancer care. Mol Oncol 2012; 6(2): 222-36.
[http://dx.doi.org/10.1016/j.molonc.2012.02.003] [PMID: 22406404]
[128]
de Ruijter TC, van der Heide F, Smits KM, Aarts MJ, van Engeland M, Heijnen VCG. Prognostic DNA methylation markers for hormone receptor breast cancer: a systematic review. Breast Cancer Res 2020; 22(1): 13.
[http://dx.doi.org/10.1186/s13058-020-1250-9] [PMID: 32005275]
[129]
Gourley C, Balmaña J, Ledermann JA, et al. Moving from poly (ADP-Ribose) polymerase inhibition to targeting DNA repair and DNA damage response in cancer therapy. J Clin Oncol 2019; 37(25): 2257-69.
[http://dx.doi.org/10.1200/JCO.18.02050] [PMID: 31050911]
[130]
Tung NM, Garber JE. BRCA1/2 testing: therapeutic implications for breast cancer management. Br J Cancer 2018; 119(2): 141-52.
[http://dx.doi.org/10.1038/s41416-018-0127-5] [PMID: 29867226]
[131]
Nicolas E, Bertucci F, Sabatier R, Gonçalves A. Targeting BRCA deficiency in breast cancer: what are the clinical evidences and the next perspectives? Cancers (Basel) 2018; 10(12): 506.
[http://dx.doi.org/10.3390/cancers10120506] [PMID: 30544963]
[132]
Xie Y, Gou Q, Wang Q, Zhong X, Zheng H. The role of BRCA status on prognosis in patients with triple-negative breast cancer. Oncotarget 2017; 8(50): 87151-62.
[http://dx.doi.org/10.18632/oncotarget.19895] [PMID: 29152070]
[133]
Trenner A, Sartori AA. Harnessing DNA double-strand break repair for cancer treatment. Front Oncol 2019; 9: 1388.
[http://dx.doi.org/10.3389/fonc.2019.01388] [PMID: 31921645]
[134]
Noordermeer SM, van Attikum H. PARP inhibitor resistance: a tug-of-war in BRCA-mutated cells. Trends Cell Biol 2019; 29(10): 820-34.
[http://dx.doi.org/10.1016/j.tcb.2019.07.008] [PMID: 31421928]
[135]
Zhou P, Wang J, Mishail D, Wang CY. Recent advancements in PARP inhibitors-based targeted cancer therapy. Precis Clin Med 2020; 3(3): 187-201.
[http://dx.doi.org/10.1093/pcmedi/pbaa030] [PMID: 32983586]
[136]
Peyraud F, Italiano A. Combined PARP inhibition and immune checkpoint therapy in solid tumors. Cancers (Basel) 2020; 12(6): 1502.
[http://dx.doi.org/10.3390/cancers12061502] [PMID: 32526888]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy