Predicting resistance to endocrine therapy in breast cancer: It's time for epigenetic biomarkers (Review)

  • Authors:
    • Mário Fontes‑Sousa
    • Maria Amorim
    • Sofia Salta
    • Susana Palma De Sousa
    • Rui Henrique
    • Carmen Jerónimo
  • View Affiliations

  • Published online on: January 11, 2019     https://doi.org/10.3892/or.2019.6967
  • Pages: 1431-1438
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Notwithstanding the marked progress in breast cancer (BC) management, it still constitutes the most common malignancy in women and a major cause of morbidity and mortality, thus remaining a major health issue worldwide. Most BC cases are hormone receptor (HR) positive (luminal A or B molecular subtypes) and endocrine treatment (ET) is an important therapeutic modality at all disease stages. Nevertheless, despite substantial improvements in BC patient outcome, effectiveness of ET is limited, as up to 40% of patients eventually relapse or progress and endocrine resistant BC has a less favorable prognosis and constitutes a therapeutic challenge. The biological mechanisms underlying endocrine resistance are, however, still poorly understood. In this review, we focused on data regarding the main epigenetic mechanisms associated with the development of endocrine treated‑resistant BC described so far, including alterations in DNA methylation, non‑coding RNAs, chromatin remodeling, post‑translational histone modifications and histone variants. Notably, specific epigenetic alterations have been characterized in this subset of breast tumors and may be of clinical value for individualized patient management in the future.

Introduction

Most breast cancers (BC), over 2/3 of cases, express estrogen (ER) and progesterone (PR) receptors (1). This is extremely important since these are used as biomarkers for subtype classification, with implications in choice of treatment and prognosis in BC patients (2). Notably, endocrine therapies (ET) have been successfully used for treating ER positive BC patients with significant impact in patient outcome. Several endocrine drugs are approved for BC treatment, most notably tamoxifen, toremifene, anastrozole, letrozole, exemestane and fulvestrant, which may be used in different clinical contexts, such as chemoprophylaxis, neoadjuvant, adjuvant and palliative treatments. However, the effectiveness of ET is limited as up to 40% of patients may experience disease recurrence while on ET adjuvant treatment (1,3). Moreover, in the metastatic setting, acquired resistance to ET is virtually an universal feature, and is clinically defined in accordance to the 3rd ESO-ESMO International Consensus Guidelines (4) and many efforts have been made to understand the mechanisms involved in acquisition of acquired resistance to ET. These, however, remain mostly elusive and no biomarkers have been validated in this setting despite intense drug development and approval.

Epigenetics may be defined as mechanisms that regulate cell fate specifications, while the DNA remains unchanged (5). Some of these mechanisms include DNA methylation, non-coding RNAs, chromatin remodeling and histone post-translational modifications or variants. Collectively, these components constitute the epigenome machinery whose role is to define which information is available for transcription and for translation (5). DNA methylation is performed by specific enzymes, the DNA methyltransferases (DNMTs) that introduce a methyl group at the 5′ position of a cytosine ring inside CpG dinucleotides (6). Globally, promoter methylation of genes is associated with transcription inhibition (6). Furthermore, the N-terminal tails of histones may undergo post-translation modifications that subsequently impact the chromatin structure (7). The most well-studied histone post-translation modifications are histone acetylation and histone methylation. Histone acetylation is associated with gene expression and is carried out by histone acetyltransferases (HATs), while histone deacetylation is accomplished by histone deacetylases (HDACs) (7). Histone methylation, which depending on the residue and the number of methyl groups may lead either to transcription repression or activation (8), is catalyzed by histone methyltransferases (HMTs), while histone demethylation is performed by histone demethylases (HDMs) (7). In addition to post-translational histone modifications, histone variants that can replace canonical histones are an additional level of epigenetic complexity, and contribute to the shaping of the chromatin structure.

Non-coding RNAs (ncRNAs) comprise a hidden layer of internal signals that control various levels of gene expression (9). Among these, microRNAs (miRNAs) and long non-coding RNAs (lncRNAs) are the most frequently reported in BC. lncRNAs are ncRNA molecules usually longer than 200 nucleotides that do not fit into known classes of small or structural RNAs (9) and may act as protein-DNA or protein-protein scaffolds, miRNA sponges, protein decoys, or regulators of translation (10). miRNAs are endogenous, small non-coding single-stranded RNAs with ~22 nucleotides in length, that exert a finely tuned regulation of gene expression at the post-transcriptional level (11) by binding to mRNA targets, inducing its cleavage or repressing its translation (11).

Over the last few years, convincing data has suggested that altered epigenetic regulation may be involved in tumor initiation, progression and cancer resistance to therapy, including endocrine resistance, particularly in BC. For instance, ER expression is currently one of the foremost predictive biomarkers of response to ET, and altered expression of ER may be due to hypermethylation of CpG islands within its promoter, increased histone deacetylase activity in the ESR1 promoter or translational repression by miRNAs (12). Since ER was found to be deleted in only 15–20% of endocrine-resistant BC, several epigenetic mechanisms may be involved in the development of endocrine treatment-resistance (3), and some of these are depicted in Fig. 1.

Our objective was to review the published evidence regarding epigenetic mechanisms associated to ET resistance in BC, as it may be considered an emerging subject and worth special focus.

Evidence acquisition

For the selection of the most relevant bibliography, we conducted a PubMed® search using the terms ‘endocrine resistance’, ‘breast cancer’ and ‘epigenetic mechanisms’. Reference lists from key articles were also searched for additional relevant data. The criteria for article selection were: written in English, central theme based on ET resistance on BC and epigenetic mechanisms. Original studies were selected based on the detail of analysis, mechanistic support of data, novelty, and potential clinical usefulness of the findings. Chemotherapy/radiotherapy-resistance, HER2-enriched subtype or ‘triple negative’ BC citations were excluded for being outside the scope of this review.

DNA methylation

DNA methylation is one of the most common epigenetic changes and has been reported in multiple tumors, including BC (9,13). This epigenetic alteration is inherently stable and has been proposed as a promising cancer biomarker in multiple cancers since it can be sampled from less invasive sources such as liquid biopsies (plasma or urine) (1315). Thus, the role of DNA methylation as a predictor of ET resistance is a field of growing interest and has become the focus of several research teams (1618) since it may improve BC patients' risk stratification.

Notably, Stone et al reported that in endocrine treated-resistant cell lines, DNA hypermethylation occurs predominantly at estrogen-responsive enhancers, leading to reduced ER binding and subsequently to expression downregulation. Furthermore, luminal subtype BC patients with relapsed disease exhibited significantly higher methylation levels at all enhancer loci studied (19). By comparing anti-estrogen-resistant cell lines with the parental sensitive cell line, DNA methylation of the promoter region of genes was also suggested to play a role in the emergence of endocrine resistance (17,20) (Table I). Multicenter studies, including several cohorts of BC patients were able to confirm these findings. Specifically, PITX2 methylation levels were consistently identified as a valuable biomarker to predict outcome in low-risk BC patients (ER-positive, node-negative) treated with surgery followed by adjuvant tamoxifen (21,22). Nevertheless, multiple validations are still required before the implementation of these markers in the clinical setting (Table I). Thus, to date, no clinical trials have assessed the clinical relevance of these candidate biomarkers.

Table I.

DNA methylation of the promoter region of genes as predictive biomarkers to different modalities of endocrine therapies along with their role and the biological samples used in each study.

Table I.

DNA methylation of the promoter region of genes as predictive biomarkers to different modalities of endocrine therapies along with their role and the biological samples used in each study.

BiomarkerRoleAgentSamplesRef.
PTENHypermethylation is associated with resistanceTMXCell lines(47)
PTGER4Hypomethylation is associated with resistanceEDT (48)
CDK10Hypermethylation is associated with shorter PFS and OSTMXCell lines and(49)
HOXC10Hypermethylation is associated with resistanceEDT, AIs and TMXtumor tissues(50)
ESR1 CYP1B1High methylation levels are associated with a better outcomeTMXTumor tissues(16)
ID4Hypomethylation is associated with resistance (51)
NAT1Hypermethylation is associated with resistance (52)
PITX2Hypermethylation is associated with worse outcome and shorter MFS (21,22,53)
PRHypermethylation is associated with resistance (54)
PSAT1Hypermethylation is associated with good clinical benefit (55)

[i] PFS, progression-free survival; OS, overall survival; MFS, metastasis-free survival; TMX, tamoxifen; AIs, aromatase inhibitors; EDT, estrogen deprivation therapy; PTEN, phosphatase and tensin homolog; PTGER4, prostaglandin E receptor 4; CDK10, cyclin dependent kinase 10; HOXC10, homeobox C10; BRCA1, BRCA1 DNA repair associated; ESR1, estrogen receptor 1; CYP1B1, cytochrome P450 family 1 subfamily B member 1; ID4, inhibitor of DNA binding 4 HLH protein; NAT1, N-acetyltransferase 1; PITX2, paired like homeodomain 2; PR, progesterone receptor; PSAT1, phosphoserine aminotransferase 1.

Non-coding RNAs

As previously mentioned, decreased ER expression may be due to post-transcription regulation of miRNAs, including that of miR-221/222, whose overexpression has been associated with resistance to tamoxifen (23,24) and fulvestrant (25). Conversely, miR-342-3p levels were revealed to be positively correlated with ER mRNA expression in human BC and associated with tamoxifen sensitivity (26,27). miRNAs that regulate growth, survival, apoptosis, epithelial-mesenchymal transition (EMT) and metastasis of BC cells may be implicated in loss of responsiveness to ET. In particular, PTEN downregulation due to specific miRNAs, permitting abnormal Pi3K/Akt pathway activation, promote estrogen-independent growth and survival of BC cells leading to endocrine treatment resistance (28,29).

Several clinical trials are currently ongoing to evaluate the role of miRNAs as predictive biomarkers in BC. Specifically, trials such as NCT01231386 and NCT01722851, aim to identify circulating miRNAs aiding at the identification of biomarkers of early response to neoadjuvant therapy, including ET, which may be used as potential targets for personalized therapies. Conversely, the NCT01612871 trial was set to explore a panel of circulating miRNAs that could aid to monitor the disease status of the patient while on adjuvant ET (3032).

lncRNAs have also been associated with endocrine treatment resistance. Particularly, lncRNAs, breast cancer anti-estrogen resistance 4 (BRCAAR4) overexpression (33,34) and DSCAM antisense RNA 1 (DSCAM-AS1) (35), which contains an ER promoter binding motif, have been revealed to predict tamoxifen resistance in primary BC (Table II and Fig. 1).

Table II.

Non-coding RNAs involved in response (sensitivity/resistance) to different modalities of endocrine therapies along with their putative targets/mechanism and the biological samples used in each study.

Table II.

Non-coding RNAs involved in response (sensitivity/resistance) to different modalities of endocrine therapies along with their putative targets/mechanism and the biological samples used in each study.

ETRolemiRNAPutative targetAgentSamplesRefs.
AntiESensitivitymiR-375MTDHTMXCell lines(56)
miR-873CDK3 (57)
miR-320aARPP19, ESRRG (58)
Let-7b/iESR1 (ER-α36 variant) (59)
miR-451YWHAZ (60)
miR-17/20CCND1 (61)
miR-148aALCAM
miR-152 (62)
miR-200c/bZEB1/2TMX and FULV (63)
miR-15a/16BRCAL2TMXCell lines and xenografts(64)
miR-342-3pBMP7, GEMIN4 Cell lines and tumor tissues(26)
miR-26aEZH2 Tumor tissues(65)
miR-30cEGFR (66)
miR-10a
miR-126 (67)
ResistancemiR-10bHDAC4TMXCell lines(68)
lncRNA DSCAM-AS1Binding to the hnRNPL proteinTumor tissue and cell lines (35)
miR-519aCDKN1A, PTEN, RB1 (29)
lncRNA BRCAAR4 (34)
miR-221/222ESR1, CDKN1B, CTNNB1TMX and FULV (23,25,69)
miR-301FOXF2, PTEN, BBRCA3iso2, COL2A1TMXTumor tissue, cell lines and xenografts(28)
miR-155SOCS6 (70)
miR-210EFNA3, E2F3, RAD52, FGFRL1, MET Tumor tissue(71)
AIsSensitivityLet-7fCYP19A1LETCell lines(72)
miR-125b let-7cERBB2LET and ANATumor tissues and cell lines(73)
ResistancemiR-128aTGFBR1LETCell lines(74)
miR-181a BRCAL2L11 Cell lines, xenografts and tumor tissue(75)

[i] miR, microRNA; lncRNA, long non-coding RNA; ET, endocrine therapies; AntiE, anti estrogen; AIs, aromatase inhibitors; ANA, anastrozole; FULV, fulvestrant; DSCAM-AS1, DSCAM antisense RNA 1; BRCAAR4, breast cancer anti-estrogen resistance 4; MTDH, metadherin; CDK, cyclin-dependent kinase; ARPP19, cAMP-regulated phosphoprotein 19; ESRRG, estrogen related receptor gamma; YWHAZ, tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein ζ; CCND1, cyclin D1; ALCAM, activated leukocyte cell adhesion molecule; ZEB, zinc finger E-box-binding homeobox; BRCAL-2, B-cell lymphoma 2; BMP7, bone morphogenetic protein 7; GEMIN4, gem (nuclear organelle)-associated protein 4; EZH2, enhancer of zeste homolog 2; EGFR, epidermal growth factor receptor; HDAC4, histone deacetylase 4; HnRNPL, heterogeneous nuclear ribonucleoprotein L; CDKN, cyclin-dependent kinase inhibitor; PTEN, phosphatase and tensin homolog; RB1, retinoblastoma 1; ESR1, estrogen receptor 1; CTNNB1, catenin β 1; FOXF2, forkhead box F2; BBRCA3iso-2, BRCAL2 binding component 3 isoform 2; COL2A1, collagen type II alpha 1; SOCS, suppressor of cytokine signaling; EFNA3, ephrin A3; E2F3, E2F transcription factor 3; RAD52, RAD52 homolog DNA repair protein; FGFRL1, fibroblast growth factor receptor-like 1; MET, hepatocyte growth factor receptor; CYP19A1, cytochrome P450 family 19 subfamily A member 1; ERBB2, erb-b2 receptor tyrosine kinase 2; TGFBR1, transforming growth factor β-receptor 1; BRCAL2L11, BRCAL2 like 11; ZNF217, zinc finger protein 217.

Chromatin remodeling, post-translational histone modifications and histone variants

Histone post-translation modifications induce chromatin landscape changes that subsequently favor ER repression, thus promoting other signaling pathways that could lead to endocrine resistance, as exemplified by Magnani et al that revealed how the genome's accessibility is altered in drug-resistant vs. drug-responsive BC cells (36). Recently, expression of the H3K36 methyltransferase NSD2 was found to be higher in tamoxifen-resistant BC cell lines, associated with disease recurrence and worse survival (37). Furthermore, H3K37me3 profiles enabled the identification of patients with poor outcome after aromatase inhibitor (AI) treatment (38).

Furthermore, it was recently demonstrated that transcription repression performed by ER co-repressors confer tamoxifen sensitivity through recruitment of HDACs to DNA (39). This evidence suggests that loss of ER co-repressors may sensitize BC cells to the cytotoxic effects of HDACs inhibitors (HDACi). Notably, some clinical trials have demonstrated that HDACi appears to re-establish sensitivity to anti-estrogens in a subset of endocrine treated-resistant tumors (40,41). In addition, the ENCORE-301, a randomized phase II trial (41) tested entinostat, an oral HDACi, in the endocrine-resistance, more specifically AI in post-menopausal women. The results revealed modest improvement in PFS but much greater improvement in overall survival (OS)-median OS improved to 28.1 months in the experimental arm vs. 19.8 months (HR, 0.59; 95% CI, 0.36 to 0.97; P=0.036). Ongoing clinical trials are further testing entinostat in monotherapy or in combination. Moreover, in custom-generated tamoxifen resistant cell lines, treatment with HDACi re-established sensitivity to tamoxifen through significant Bcl-2 downregulation, growth arrest and apoptosis (42).

Histone variants, such as H2A.Z, an H2A variant, have been shown to be intimately linked to estrogen signaling (43). Notably, a study has already provided a link (yet uncharacterized) between H2A.Z and endocrine resistance by revealing that H2A.Z overexpression led to increased estrogen-independent proliferation (44). Furthermore, another study demonstrated that the histone HIST1H2BE, an H2B variant, was overexpressed not only in endocrine-resistant cell lines, but also in AI-treated tumors from patients which relapsed compared to those that did not (45).

Furthermore, an emerging class of transcription factors named ‘pioneer factors’, appear to be key players in shaping chromatin structure through binding to chromatin prior to transcription factors, making it accessible for transcription factors, together with histone post-translation modifications and histone variants [68–70]. PBX1 is an example of this class-its expression levels have been associated with reduced metastasis-free survival in ER-positive BC patients (46). Furthermore, a gene expression signature based on NOTCH-PBX1 activity was found to discriminate BC patients that are responsive to ET from those which are not. Notably, PBX1 knockdown was sufficient to arrest ER-resistant BC cell growth (36).

These and other chromatin remodeling complexes associated with endocrine resistance are summarized in Table III along with their putative role and the biological samples in which they were characterized.

Table III.

Chromatin remodeling, post-translational histone modifications and histone variants involved in response (sensitivity/resistance) to endocrine therapies along with their putative epigenetic mechanism and role in response.

Table III.

Chromatin remodeling, post-translational histone modifications and histone variants involved in response (sensitivity/resistance) to endocrine therapies along with their putative epigenetic mechanism and role in response.

BiomarkerEpigenetic mechanismRoleSamplesRefs.
H3K27me3 Post-translationalH3K27me3 profile predicts the treatmentTumor tissues
profileshistone modificationoutcomes for first-line AIs (38)
PBX1Chromatin remodelingResistance to ETCell lines and(46)
HDAC6 Post-translationalSensitivity to TMX bytumor tissues
histone modificationdeacetylation of alpha-tubulin (76,77)
HIST1H2BEHistone variantOverexpressed in AI-resistant tumors/cell lines
compared to AI-sensitive tumors/cell lines (45)
NSD2 Post-translationalHistone H3K36 methyltransferase
histone modificationthat confers resistance to TAM by upregulating
key glucose metabolic enzyme genes (37)
H3K27me3 Post-translationalResistance to ET due toCell lines
demethylationhistone modificationBrCal-2 expression (78)
H2A.Z Increased H2A.Z expression promotes
Histone variantcellular proliferation, namely when E2
levels are low and during TMX treatment (44)

[i] PBX1, PBX homeobox 1; HDAC6, histone deacetylase 6; HIST1H2BE, histone cluster 1 H2B family member E; NSD2, nuclear receptor binding SET domain protein 2; H2A.Z, H2A histone family member Z; ET, endrocrine therapies; TMX, tamoxifen; AIs, aromatase inhibitors.

Conclusion

Notwithstanding the prevalence of endocrine treatment resistance in BC, predictive and diagnostic biomarkers in this setting are markedly lacking in clinical practice. In this review, we summarized emerging evidence that epigenetic mechanisms may prove useful for this purpose. These would perform as non-invasive predictive biomarkers of treatment-resistance, providing affordable and sequential monitoring during the course of treatment. The concept of early detection (preclinical) of therapy resistance is compelling, as it could assist clinicians in choosing the most appropriate individualized therapeutic strategy.

Furthermore, some epigenetic modifications in addition to conveying information concerning prediction of response, are also appealingly targetable, in particular due to their reversible nature. The clinical usefulness of these findings, however, is still elusive, mostly due to lack of standardization in methodology, limiting reproducibility.

Promising results have been arising in clinically meaningful trials, such as ENCORE-301. A useful approach would be the integration of the candidate biomarkers into a panel, enabling its validation in a clinical trial setting. Hopefully, this will be accomplished in the near future.

Acknowledgments

Not applicable.

Funding

The present study was supported by a grant from the Research Center of Portuguese Oncology Institute of Porto (PI 74-CI-IPOP-19-2016) and the Portuguese Society of Oncology-YOuR Project. SS was supported by a PhD fellowship IPO/ESTIMA-1 NORTE-01-0145-FEDER-000027.

Availability of data and materials

The datasets used during the present study are available from the corresponding author upon reasonable request.

Authors' contributions

MFS, SPDS, RH and CJ conceived and designed the review. MFS, MA, SS performed the literature search and wrote the manuscript. SPDS, RH and CJ reviewed and edited the manuscript. All authors read and approved the manuscript and agree to be accountable for all aspects of the research in ensuring that the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Magnani L, Brunelle M, Gévry N and Lupien M: Chromatin landscape and endocrine response in breast cancer. Epigenomics. 4:675–683. 2012. View Article : Google Scholar : PubMed/NCBI

2 

Cheang MC, van de Rijn M and Nielsen TO: Gene expression profiling of breast cancer. Annu Rev Pathol. 3:67–97. 2008. View Article : Google Scholar : PubMed/NCBI

3 

Normanno N, Di Maio M, De Maio E, De Luca A, de Matteis A, Giordano A and Perrone F; NCI-Naple Breast Cancer Group, : Mechanisms of endocrine resistance and novel therapeutic strategies in breast cancer. Endocr Relat Cancer. 12:721–747. 2005. View Article : Google Scholar : PubMed/NCBI

4 

Cardoso F, Costa A, Senkus E, Aapro M, André F, Barrios CH, Bergh J, Bhattacharyya G, Biganzoli L, Cardoso MJ, et al: 3rd ESO-ESMO international consensus guidelines for Advanced Breast Cancer (ABC 3). Breast. 31:244–259. 2017. View Article : Google Scholar : PubMed/NCBI

5 

Rodríguez-Paredes M and Esteller M: Cancer epigenetics reaches mainstream oncology. Nat Med. 17:330–339. 2011. View Article : Google Scholar : PubMed/NCBI

6 

Jones PA: Functions of DNA methylation: Islands, start sites, gene bodies and beyond. Nat Rev Genet. 13:484–492. 2012. View Article : Google Scholar : PubMed/NCBI

7 

Bannister AJ and Kouzarides T: Regulation of chromatin by histone modifications. Cell Res. 21:381–395. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Zentner GE and Henikoff S: Regulation of nucleosome dynamics by histone modifications. Nat Struct Mol Biol. 20:259–266. 2013. View Article : Google Scholar : PubMed/NCBI

9 

Amorim M, Salta S, Henrique R and Jerónimo C: Decoding the usefulness of non-coding RNAs as breast cancer markers. J Transl Med. 14:2652016. View Article : Google Scholar : PubMed/NCBI

10 

Kung JT, Colognori D and Lee JT: Long noncoding RNAs: Past, present, and future. Genetics. 193:651–669. 2013. View Article : Google Scholar : PubMed/NCBI

11 

Huntzinger E and Izaurralde E: Gene silencing by microRNAs: Contributions of translational repression and mRNA decay. Nat Rev Genet. 12:99–110. 2011. View Article : Google Scholar : PubMed/NCBI

12 

Sharma D, Blum J, Yang X, Beaulieu N, Macleod AR and Davidson NE: Release of methyl CpG binding proteins and histone deacetylase 1 from the estrogen receptor α (ER) promoter upon reactivation in ER-negative human breast cancer cells. Mol Endocrinol. 19:1740–1751. 2005. View Article : Google Scholar : PubMed/NCBI

13 

Heyn H and Esteller M: DNA methylation profiling in the clinic: Applications and challenges. Nat Rev Genet. 13:679–692. 2012. View Article : Google Scholar : PubMed/NCBI

14 

Jerónimo C and Henrique R: Epigenetic biomarkers in urological tumors: A systematic review. Cancer Lett. 342:264–274. 2014. View Article : Google Scholar : PubMed/NCBI

15 

Costa-Pinheiro P, Montezuma D, Henrique R and Jerónimo C: Diagnostic and prognostic epigenetic biomarkers in cancer. Epigenomics. 7:1003–1015. 2015. View Article : Google Scholar : PubMed/NCBI

16 

Widschwendter M, Siegmund KD, Müller HM, Fiegl H, Marth C, Müller-Holzner E, Jones PA and Laird PW: Association of breast cancer DNA methylation profiles with hormone receptor status and response to tamoxifen. Cancer Res. 64:3807–3813. 2004. View Article : Google Scholar : PubMed/NCBI

17 

Fan M, Yan PS, Hartman-Frey C, Chen L, Paik H, Oyer SL, Salisbury JD, Cheng AS, Li L, Abbosh PH, et al: Diverse gene expression and DNA methylation profiles correlate with differential adaptation of breast cancer cells to the antiestrogens tamoxifen and fulvestrant. Cancer Res. 66:11954–11966. 2006. View Article : Google Scholar : PubMed/NCBI

18 

Musgrove EA and Sutherland RL: Biological determinants of endocrine resistance in breast cancer. Nat Rev Cancer. 9:631–643. 2009. View Article : Google Scholar : PubMed/NCBI

19 

Stone A, Zotenko E, Locke WJ, Korbie D, Millar EK, Pidsley R, Stirzaker C, Graham P, Trau M, Musgrove EA, et al: DNA methylation of oestrogen-regulated enhancers defines endocrine sensitivity in breast cancer. Nat Commun. 6:77582015. View Article : Google Scholar : PubMed/NCBI

20 

Williams KE, Anderton DL, Lee MP, Pentecost BT and Arcaro KF: High-density array analysis of DNA methylation in Tamoxifen-resistant breast cancer cell lines. Epigenetics. 9:297–307. 2014. View Article : Google Scholar : PubMed/NCBI

21 

Maier S, Nimmrich I, Koenig T, Eppenberger-Castori S, Bohlmann I, Paradiso A, Spyratos F, Thomssen C, Mueller V, Nährig J, et al: European Organisation for Research and Treatment of Cancer (EORTC) PathoBiology group: DNA-methylation of the homeodomain transcription factor PITX2 reliably predicts risk of distant disease recurrence in tamoxifen-treated, node-negative breast cancer patients: Technical and clinical validation in a multi-centre setting in collaboration with the European Organisation for Research and Treatment of Cancer (EORTC) PathoBiology group. Eur J Cancer. 43:1679–1686. 2007. View Article : Google Scholar : PubMed/NCBI

22 

Harbeck N, Nimmrich I, Hartmann A, Ross JS, Cufer T, Grützmann R, Kristiansen G, Paradiso A, Hartmann O, Margossian A, et al: Multicenter study using paraffin-embedded tumor tissue testing PITX2 DNA methylation as a marker for outcome prediction in tamoxifen-treated, node-negative breast cancer patients. J Clin Oncol. 26:5036–5042. 2008. View Article : Google Scholar : PubMed/NCBI

23 

Miller TE, Ghoshal K, Ramaswamy B, Roy S, Datta J, Shapiro CL, Jacob S and Majumder S: MicroRNA-221/222 confers tamoxifen resistance in breast cancer by targeting p27Kip1. J Biol Chem. 283:29897–29903. 2008. View Article : Google Scholar : PubMed/NCBI

24 

Zhao JJ, Lin J, Yang H, Kong W, He L, Ma X, Coppola D and Cheng JQ: MicroRNA-221/222 negatively regulates estrogen receptor α and is associated with tamoxifen resistance in breast cancer. J Biol Chem. 283:31079–31086. 2008. View Article : Google Scholar : PubMed/NCBI

25 

Rao X, Di Leva G, Li M, Fang F, Devlin C, Hartman-Frey C, Burow ME, Ivan M, Croce CM and Nephew KP: MicroRNA-221/222 confers breast cancer fulvestrant resistance by regulating multiple signaling pathways. Oncogene. 30:1082–1097. 2011. View Article : Google Scholar : PubMed/NCBI

26 

Cittelly DM, Das PM, Spoelstra NS, Edgerton SM, Richer JK, Thor AD and Jones FE: Downregulation of miR-342 is associated with tamoxifen resistant breast tumors. Mol Cancer. 9:3172010. View Article : Google Scholar : PubMed/NCBI

27 

He YJ, Wu JZ, Ji MH, Ma T, Qiao EQ, Ma R and Tang JH: miR-342 is associated with estrogen receptor-α expression and response to tamoxifen in breast cancer. Exp Ther Med. 5:813–818. 2013. View Article : Google Scholar : PubMed/NCBI

28 

Shi W, Gerster K, Alajez NM, Tsang J, Waldron L, Pintilie M, Hui AB, Sykes J, P'ng C, Miller N, et al: MicroRNA-301 mediates proliferation and invasion in human breast cancer. Cancer Res. 71:2926–2937. 2011. View Article : Google Scholar : PubMed/NCBI

29 

Ward A, Shukla K, Balwierz A, Soons Z, König R, Sahin O and Wiemann S: MicroRNA-519a is a novel oncomir conferring tamoxifen resistance by targeting a network of tumour-suppressor genes in ER+ breast cancer. J Pathol. 233:368–379. 2014. View Article : Google Scholar : PubMed/NCBI

30 

ClinicalTrials.gov: MIRNA Profiling of Breast Cancer in Patients Undergoing Neoadjuvant or Adjuvant Treatment for Locally Advanced & Inflammatory Breast Cancer. ClinicalTrials.gov2016. https://clinicaltrials.gov/ct2/show/NCT01231386

31 

ClinicalTrials.gov: Circulating miRNAs. ICORG 10–11, V2. ClinicalTrials.gov2017. https://clinicaltrials.gov/ct2/show/NCT01722851

32 

ClinicalTrials.gov: Circulating miRNAs as Biomarkers of Hormone Sensitivity in Breast Cancer (MIRHO). ClinicalTrials.gov. 2014, https://clinicaltrials.gov/ct2/show/NCT01612871

33 

Meijer D, van Agthoven T, Bosma PT, Nooter K and Dorssers LC: Functional screen for genes responsible for tamoxifen resistance in human breast cancer cells. Mol Cancer Res. 4:379–386. 2006. View Article : Google Scholar : PubMed/NCBI

34 

Godinho MF, Sieuwerts AM, Look MP, Meijer D, Foekens JA, Dorssers LC and Van Agthoven T: Relevance of BCAR4 in tamoxifen resistance and tumour aggressiveness of human breast cancer. Br J Cancer. 103:1284–1291. 2010. View Article : Google Scholar : PubMed/NCBI

35 

Niknafs YS, Han S, Ma T, Speers C, Zhang C, Wilder-Romans K, Iyer MK, Pitchiaya S, Malik R, Hosono Y, et al: The lncRNA landscape of breast cancer reveals a role for DSCAM-AS1 in breast cancer progression. Nat Commun. 7:127912016. View Article : Google Scholar : PubMed/NCBI

36 

Magnani L, Stoeck A, Zhang X, Lánczky A, Mirabella AC, Wang TL, Gyorffy B and Lupien M: Genome-wide reprogramming of the chromatin landscape underlies endocrine therapy resistance in breast cancer. Proc Natl Acad Sci USA. 110:E1490–E1499. 2013. View Article : Google Scholar : PubMed/NCBI

37 

Wang J, Duan Z, Nugent Z, Zou JX, Borowsky AD, Zhang Y, Tepper CG, Li JJ, Fiehn O, Xu J, et al: Reprogramming metabolism by histone methyltransferase NSD2 drives endocrine resistance via coordinated activation of pentose phosphate pathway enzymes. Cancer Lett. 378:69–79. 2016. View Article : Google Scholar : PubMed/NCBI

38 

Jansen MP, Knijnenburg T, Reijm EA, Simon I, Kerkhoven R, Droog M, Velds A, van Laere S, Dirix L, Alexi X, et al: Hallmarks of aromatase inhibitor drug resistance revealed by epigenetic profiling in breast cancer. Cancer Res. 73:6632–6641. 2013. View Article : Google Scholar : PubMed/NCBI

39 

Légaré S and Basik M: Minireview: The link between ERα corepressors and histone deacetylases in tamoxifen resistance in breast cancer. Mol Endocrinol. 30:965–976. 2016. View Article : Google Scholar : PubMed/NCBI

40 

Munster PN, Thurn KT, Thomas S, Raha P, Lacevic M, Miller A, Melisko M, Ismail-Khan R, Rugo H, Moasser M, 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. View Article : Google Scholar : PubMed/NCBI

41 

Yardley DA, Ismail-Khan RR, Melichar B, Lichinitser M, Munster PN, Klein PM, Cruickshank S, Miller KD, Lee MJ and Trepel JB: 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. 31:2128–2135. 2013. View Article : Google Scholar : PubMed/NCBI

42 

Raha P, Thomas S, Thurn KT, Park J and Munster PN: Combined histone deacetylase inhibition and tamoxifen induces apoptosis in tamoxifen-resistant breast cancer models, by reversing Bcl-2 overexpression. Breast Cancer Res. 17:262015. View Article : Google Scholar : PubMed/NCBI

43 

Gévry N, Hardy S, Jacques P-É, Laflamme L, Svotelis A, Robert F and Gaudreau L: Histone H2A.Z is essential for estrogen receptor signaling. Genes Dev. 23:1522–1533. 2009. View Article : Google Scholar : PubMed/NCBI

44 

Svotelis A, Gévry N, Grondin G and Gaudreau L: H2A.Z overexpression promotes cellular proliferation of breast cancer cells. Cell Cycle. 9:364–370. 2010. View Article : Google Scholar : PubMed/NCBI

45 

Nayak SR, Harrington E, Boone D, Hartmaier R, Chen J, Pathiraja TN, Cooper KL, Fine JL, Sanfilippo J, Davidson NE, et al: A role for histone H2B variants in endocrine-resistant breast cancer. Horm Cancer. 6:214–224. 2015. View Article : Google Scholar : PubMed/NCBI

46 

Magnani L, Ballantyne EB, Zhang X and Lupien M: PBX1 genomic pioneer function drives ERα signaling underlying progression in breast cancer. PLoS Genet. 7:e10023682011. View Article : Google Scholar : PubMed/NCBI

47 

Phuong NT, Kim SK, Lim SC, Kim HS, Kim TH, Lee KY, Ahn SG, Yoon JH and Kang KW: Role of PTEN promoter methylation in tamoxifen-resistant breast cancer cells. Breast Cancer Res Treat. 130:73–83. 2011. View Article : Google Scholar : PubMed/NCBI

48 

Hiken JF, McDonald JI, Decker KF, Sanchez C, Hoog J, VanderKraats ND, Jung KL, Akinhanmi M, Rois LE, Ellis MJ, et al: Epigenetic activation of the prostaglandin receptor EP4 promotes resistance to endocrine therapy for breast cancer. Oncogene. 36:2319–2327. 2017. View Article : Google Scholar : PubMed/NCBI

49 

Iorns E, Turner NC, Elliott R, Syed N, Garrone O, Gasco M, Tutt AN, Crook T, Lord CJ and Ashworth A: Identification of CDK10 as an important determinant of resistance to endocrine therapy for breast cancer. Cancer Cell. 13:91–104. 2008. View Article : Google Scholar : PubMed/NCBI

50 

Pathiraja TN, Nayak SR, Xi Y, Jiang S, Garee JP, Edwards DP, Lee AV, Chen J, Shea MJ, Santen RJ, et al: Epigenetic reprogramming of HOXC10 in endocrine-resistant breast cancer. Sci Transl Med. 6:229ra412014. View Article : Google Scholar : PubMed/NCBI

51 

Zhang Y, Zhang B, Fang J and Cao X: Hypomethylation of DNA-binding inhibitor 4 serves as a potential biomarker in distinguishing acquired tamoxifen-refractory breast cancer. Int J Clin Exp Pathol. 8:9500–9505. 2015.PubMed/NCBI

52 

Kim SJ, Kang HS, Jung SY, Min SY, Lee S, Kim SW, Kwon Y, Lee KS, Shin KH and Ro J: Methylation patterns of genes coding for drug-metabolizing enzymes in tamoxifen-resistant breast cancer tissues. J Mol Med. 88:1123–1131. 2010. View Article : Google Scholar : PubMed/NCBI

53 

Nimmrich I, Sieuwerts AM, Meijer-van Gelder ME, Schwope I, Bolt-de Vries J, Harbeck N, Koenig T, Hartmann O, Kluth A, Dietrich D, et al: DNA hypermethylation of PITX2 is a marker of poor prognosis in untreated lymph node-negative hormone receptor-positive breast cancer patients. Breast Cancer Res Treat. 111:429–437. 2008. View Article : Google Scholar : PubMed/NCBI

54 

Pathiraja TN, Shetty PB, Jelinek J, He R, Hartmaier R, Margossian AL, Hilsenbeck SG, Issa JP and Oesterreich S: Progesterone receptor isoform-specific promoter methylation: Association of PRA promoter methylation with worse outcome in breast cancer patients. Clin Cancer Res. 17:4177–4186. 2011. View Article : Google Scholar : PubMed/NCBI

55 

Martens JW, Nimmrich I, Koenig T, Look MP, Harbeck N, Model F, Kluth A, Bolt-de Vries J, Sieuwerts AM, Portengen H, et al: Association of DNA methylation of phosphoserine aminotransferase with response to endocrine therapy in patients with recurrent breast cancer. Cancer Res. 65:4101–4117. 2005. View Article : Google Scholar : PubMed/NCBI

56 

Ward A, Balwierz A, Zhang JD, Küblbeck M, Pawitan Y, Hielscher T, Wiemann S and Sahin Ö: Re-expression of microRNA-375 reverses both tamoxifen resistance and accompanying EMT-like properties in breast cancer. Oncogene. 32:1173–1182. 2013. View Article : Google Scholar : PubMed/NCBI

57 

Cui J, Yang Y, Li H, Leng Y, Qian K, Huang Q, Zhang C, Lu Z, Chen J, Sun T, et al: MiR-873 regulates ERα transcriptional activity and tamoxifen resistance via targeting CDK3 in breast cancer cells. Oncogene. 34:3895–3907. 2015. View Article : Google Scholar : PubMed/NCBI

58 

Lü M, Ding K, Zhang G, Yin M, Yao G, Tian H, Lian J, Liu L, Liang M, Zhu T, et al: MicroRNA-320a sensitizes tamoxifen-resistant breast cancer cells to tamoxifen by targeting ARPP-19 and ERRγ. Sci Rep. 4:87352015. View Article : Google Scholar

59 

Zhao Y, Deng C, Lu W, Xiao J, Ma D, Guo M, Recker RR, Gatalica Z, Wang Z and Xiao GG: let-7 microRNAs induce tamoxifen sensitivity by downregulation of estrogen receptor α signaling in breast cancer. Mol Med. 17:1233–1241. 2011. View Article : Google Scholar : PubMed/NCBI

60 

Bergamaschi A and Katzenellenbogen BS: Tamoxifen downregulation of miR-451 increases 14-3-3ζ and promotes breast cancer cell survival and endocrine resistance. Oncogene. 31:39–47. 2012. View Article : Google Scholar : PubMed/NCBI

61 

Yu Z, Xu Z, Disante G, Wright J, Wang M, Li Y, Zhao Q, Ren T, Ju X, Gutman E, et al: miR-17/20 sensitization of breast cancer cells to chemotherapy-induced apoptosis requires Akt1. Oncotarget. 5:1083–1090. 2014. View Article : Google Scholar : PubMed/NCBI

62 

Chen MJ, Cheng YM, Chen CC, Chen YC and Shen CJ: MiR-148a and miR-152 reduce tamoxifen resistance in ER+ breast cancer via downregulating ALCAM. Biochem Biophys Res Commun. 483:840–846. 2017. View Article : Google Scholar : PubMed/NCBI

63 

Manavalan TT, Teng Y, Litchfield LM, Muluhngwi P, Al-Rayyan N and Klinge CM: Reduced expression of miR-200 family members contributes to antiestrogen resistance in LY2 human breast cancer cells. PLoS One. 8:e623342013. View Article : Google Scholar : PubMed/NCBI

64 

Cittelly DM, Das PM, Salvo VA, Fonseca JP, Burow ME and Jones FE: Oncogenic HER2{Delta}16 suppresses miR-15a/16 and deregulates BCL-2 to promote endocrine resistance of breast tumors. Carcinogenesis. 31:2049–2057. 2010. View Article : Google Scholar : PubMed/NCBI

65 

Jansen MP, Reijm EA, Sieuwerts AM, Ruigrok-Ritstier K, Look MP, Rodríguez-González FG, Heine AA, Martens JW, Sleijfer S, Foekens JA, et al: High miR-26a and low CDC2 levels associate with decreased EZH2 expression and with favorable outcome on tamoxifen in metastatic breast cancer. Breast Cancer Res Treat. 133:937–947. 2012. View Article : Google Scholar : PubMed/NCBI

66 

Rodríguez-González FG, Sieuwerts AM, Smid M, Look MP, Meijer-van Gelder ME, de Weerd V, Sleijfer S, Martens JW and Foekens JA: MicroRNA-30c expression level is an independent predictor of clinical benefit of endocrine therapy in advanced estrogen receptor positive breast cancer. Breast Cancer Res Treat. 127:43–51. 2011. View Article : Google Scholar : PubMed/NCBI

67 

Hoppe R, Achinger-Kawecka J, Winter S, Fritz P, Lo WY, Schroth W and Brauch H: Increased expression of miR-126 and miR-10a predict prolonged relapse-free time of primary oestrogen receptor-positive breast cancer following tamoxifen treatment. Eur J Cancer. 49:3598–3608. 2013. View Article : Google Scholar : PubMed/NCBI

68 

Ahmad A, Ginnebaugh KR, Yin S, Bollig-Fischer A, Reddy KB and Sarkar FH: Functional role of miR-10b in tamoxifen resistance of ER-positive breast cancer cells through downregulation of HDAC4. BMC Cancer. 15:5402015. View Article : Google Scholar : PubMed/NCBI

69 

Wei Y, Lai X, Yu S, Chen S, Ma Y, Zhang Y, Li H, Zhu X, Yao L and Zhang J: Exosomal miR-221/222 enhances tamoxifen resistance in recipient ER-positive breast cancer cells. Breast Cancer Res Treat. 147:423–431. 2014. View Article : Google Scholar : PubMed/NCBI

70 

Shen R, Wang Y, Wang CX, Yin M, Liu HL, Chen JP, Han JQ and Wang WB: MiRNA-155 mediates TAM resistance by modulating SOCS6-STAT3 signalling pathway in breast cancer. Am J Transl Res. 7:2115–2126. 2015.PubMed/NCBI

71 

Rothé F, Ignatiadis M, Chaboteaux C, Haibe-Kains B, Kheddoumi N, Majjaj S, Badran B, Fayyad-Kazan H, Desmedt C, Harris AL, et al: Global microRNA expression profiling identifies MiR-210 associated with tumor proliferation, invasion and poor clinical outcome in breast cancer. PLoS One. 6:e209802011. View Article : Google Scholar : PubMed/NCBI

72 

Shibahara Y, Miki Y, Onodera Y, Hata S, Chan MS, Yiu CC, Loo TY, Nakamura Y, Akahira J, Ishida T, et al: Aromatase inhibitor treatment of breast cancer cells increases the expression of let-7f, a microRNA targeting CYP19A1. J Pathol. 227:357–366. 2012. View Article : Google Scholar : PubMed/NCBI

73 

Bailey ST, Westerling T and Brown M: Loss of estrogen-regulated microRNA expression increases HER2 signaling and is prognostic of poor outcome in luminal breast cancer. Cancer Res. 75:436–445. 2015. View Article : Google Scholar : PubMed/NCBI

74 

Masri S, Liu Z, Phung S, Wang E, Yuan YC and Chen S: The role of microRNA-128a in regulating TGFbeta signaling in letrozole-resistant breast cancer cells. Breast Cancer Res Treat. 124:89–99. 2010. View Article : Google Scholar : PubMed/NCBI

75 

Hayes EL and Lewis-Wambi JS: Mechanisms of endocrine resistance in breast cancer: An overview of the proposed roles of noncoding RNA. Breast Cancer Res. 17:402015. View Article : Google Scholar : PubMed/NCBI

76 

Zhang Z, Yamashita H, Toyama T, Sugiura H, Omoto Y, Ando Y, Mita K, Hamaguchi M, Hayashi S and Iwase H: HDAC6 expression is correlated with better survival in breast cancer. Clin Cancer Res. 10:6962–6968. 2004. View Article : Google Scholar : PubMed/NCBI

77 

Saji S, Kawakami M, Hayashi S, Yoshida N, Hirose M, Horiguchi S, Itoh A, Funata N, Schreiber SL, Yoshida M, et al: Significance of HDAC6 regulation via estrogen signaling for cell motility and prognosis in estrogen receptor-positive breast cancer. Oncogene. 24:4531–4539. 2005. View Article : Google Scholar : PubMed/NCBI

78 

Svotelis A, Bianco S, Madore J, Huppé G, Nordell-Markovits A, Mes-Masson AM and Gévry N: H3K27 demethylation by JMJD3 at a poised enhancer of anti-apoptotic gene BCL2 determines ERα ligand dependency. EMBO J. 30:3947–3961. 2011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2019
Volume 41 Issue 3

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Fontes‑Sousa M, Amorim M, Salta S, Palma De Sousa S, Henrique R and Jerónimo C: Predicting resistance to endocrine therapy in breast cancer: It's time for epigenetic biomarkers (Review). Oncol Rep 41: 1431-1438, 2019
APA
Fontes‑Sousa, M., Amorim, M., Salta, S., Palma De Sousa, S., Henrique, R., & Jerónimo, C. (2019). Predicting resistance to endocrine therapy in breast cancer: It's time for epigenetic biomarkers (Review). Oncology Reports, 41, 1431-1438. https://doi.org/10.3892/or.2019.6967
MLA
Fontes‑Sousa, M., Amorim, M., Salta, S., Palma De Sousa, S., Henrique, R., Jerónimo, C."Predicting resistance to endocrine therapy in breast cancer: It's time for epigenetic biomarkers (Review)". Oncology Reports 41.3 (2019): 1431-1438.
Chicago
Fontes‑Sousa, M., Amorim, M., Salta, S., Palma De Sousa, S., Henrique, R., Jerónimo, C."Predicting resistance to endocrine therapy in breast cancer: It's time for epigenetic biomarkers (Review)". Oncology Reports 41, no. 3 (2019): 1431-1438. https://doi.org/10.3892/or.2019.6967