Skip to main content
Epigenomics logoLink to Epigenomics
. 2015 Aug;7(5):847–862. doi: 10.2217/epi.15.10

Role of epigenetic modifications in luminal breast cancer

Hany A Abdel-Hafiz 1,1,*, Kathryn B Horwitz 1,1,2,2
PMCID: PMC4539290  NIHMSID: NIHMS671003  PMID: 25689414

Abstract

Luminal breast cancers represent approximately 75% of cases. Explanations into the causes of endocrine resistance are complex and are generally ascribed to genomic mechanisms. Recently, attention has been drawn to the role of epigenetic modifications in hormone resistance. We review these here. Epigenetic modifications are reversible, heritable and include changes in DNA methylation patterns, modification of histones and altered microRNA expression levels that target the receptors or their signaling pathways. Large-scale analyses indicate distinct epigenomic profiles that distinguish breast cancers from normal and benign tissues. Taking advantage of the reversibility of epigenetic modifications, drugs that target epigenetic modifiers, given in combination with chemotherapies or endocrine therapies, may represent promising approaches to restoration of therapy responsiveness in these cases.

Keywords: : breast cancer, epigenetic modifications, estrogen receptors, progesterone receptors


Breast cancer is the most common cancer and the second leading cause of cancer deaths among women worldwide. It is a heterogeneous disease, which presents significant challenges for treatment. Among other things, breast cancers are subclassified into various pathological categories and most recently into several molecular subtypes that include: luminal A, generally ER and/or PR positive and HER2 negative; luminal B, generally ER+ with low or no PR, exceptionally high proliferation rate and occasionally HER2 overexpressed; HER2+, ER-PR- and HER2 overexpressed; Basal-like, including triple-negative ER-PR-HER2-; and Claudin-low that also lack these markers and resemble cells undergoing epithelial-mesenchymal transition (EMT) [1]. These molecular subtypes differ significantly in incidence, survival and response to therapies. Over 70% of breast cancers fall into the two luminal categories. They tend to respond to ‘endocrine’ therapies that target ER, including the mixed antiestrogen tamoxifen or the pure antiestrogen fulvestrant, or estrogen (E) deprivation therapies like aromatase inhibitors. However, for reasons that are unclear, the clinical behavior of ER+ breast tumors and their response to therapies is highly variable even under conditions when ER levels are similar [2]. Indeed over 30% of ER+ tumors fail to respond to any endocrine therapies, exhibiting intrinsic (or de novo) resistance. An additional 30–40% of tumors that initially respond to therapies, eventually acquire resistance [2]. Restoring endocrine responsiveness to tumors with intrinsic or acquired resistance remains an important clinical goal.

Various explanations have been advanced for endocrine therapy resistance among ER+ tumors. Traditionally, these have invoked genetic mechanisms targeting E or ER signaling and proffering explanations such as: E-independent proliferation resulting from a constitutively active ER mutant; deregulation of ER-dependent transcription; alteration of E-dependent cell-cycle or cell-death controls; activation of alternative escape pathways such as upregulation of growth factor signaling. Each of these likely explains some cases of E-independent growth, tumor recurrence or endocrine resistance (reviewed in [3–5]). In addition, recent studies propose epigenetic mechanisms as important players in resistance (reviewed in [4,6]). Compared with genetic changes like ones involving mutations, epigenetic modifications are for the most part enzymatic and potentially reversible. Epigenetic modifications include methylation of DNA, acetylation of histone proteins and alteration of miRNA expression; all of which influence protein synthesis patterns [7]. Since epigenetic modifications can be reversed, they appear to be desirable targets for cancer therapies. Briefly (Figure 1), major epigenetic targets are:

  • DNA – Methylation, which adds a methyl group to cytosine (C) or adenine (A) nucleotides, alters the ability of DNA to be transcribed. This biochemical modification is carried out by a series of DNMTs charged with either maintenance methylation or de novo methylation. In cancers, attempts to modify DNA methylation patterns for therapeutic purposes have focused on DNMT inhibitors, including 5-Aza-2′-deoxycytidine (AZA; decitabine);

  • Chromatin – dsDNA is wrapped around core histone–protein complexes to form larger order nucleosomal structures whose position determines whether chromatin is ‘open’ and available for transcription or ‘closed’ and transcriptionally repressed. Acetylation/deacetylation controls nucleosome positioning by modifying lysine residues in the N-terminal tail of histones. These reactions are catalyzed by HAT or HDAC that are also thought to be important targets for cancer therapies. Valproic acid and suberanilohydroxamic acid (SAHA, vorinostat) are HDAC inhibitors that prevent inappropriate chromatin remodeling and are in clinical trials to restore hormone responsiveness (Tables 1 & 2);

  • MicroRNA – Just as methylation modifies DNA and its ability to be transcribed, it modifies miRNAs and their ability to regulate protein expression post-transcriptionally. For example, marks of epigenetic hypomethylation on miRNAs that regulate ER signaling are associated with deregulated ER function in breast cancers [8].

Figure 1. . Regulation of gene transcription by epigenetic modifications in breast cancers.

Figure 1. 

Methylation of histone H3 lysine 4 (H3K4), H3K36 or H3K79 is associated with open chromatin and active transcription. Methylation of H3K9, H3K20 or H3K27 is associated with closed chromatin and gene silencing. Histone methylation is regulated by several enzymes that are overexpressed in breast cancer, for example, LSD1, EZH2, G9a and SYMD3. DNA is methylated by DNMTs. MBDs bind to methylated cytosine and form a complex with SIN3A and HDAC leading to chromatin compaction and gene silencing. Histone acetylation is regulated by HDACs and HATs. HATs acetylate histone, relax chromatin and allow transcription factor binding and enhanced transcription. HDAC overexpression has been reported in breast cancers and the use of HDAC inhibitors, trichostatin A or vorinostat, may represent promising approaches to restoration of endocrine responsiveness. Examples of agents used in experimental or clinical trials with promising results are shown.

Ac: Acetylation; Me: Methylation.

Modified with permission from [9] © BioMed Central Ltd (2011).

Table 1. . Clinical trials using epigenetic drugs in breast cancer: published results.

Epigenetic agent Type Combined with Type Type of study Number of patients Response Ref.
Vorinostat
HDAC inhibitor
Decitabine
DNMT inhibitor
Phase I
22 advanced solid tumors
7 (32%) SD
[10]
Hydralazine + valproic acid
HDAC inhibitor
Doxorubicin
Chemotherapy
Phase I
15 advanced breast cancer
1 (6.6%) pathological CR
[11]
Hydralazine + valproic acid
HDAC inhibitor
Paclitaxel, Anastrozole
Chemotherapy, Endocrine therapy
Phase II
Three advanced solid tumors
1 (33%) SD
[12]
Vorinostat
HDAC inhibitor
None
 
Phase I
50 (solid tumors) two (breast cancer)
4 (8%) 0% (breast tumor)
[13]
Vorinostat
HDAC inhibitor
None
 
Phase II
14 metastatic breast cancer
4 (29%) SD
[14]
Vorinostat
HDAC inhibitor
Tamoxifen
Endocrine therapy
Phase II
43 ER+ metastatic breast cancer
11 (33%)
[15]
Entinostat
HDAC inhibitor
Aromatase inhibitor
Endocrine therapy
Phase II
27 ER+ advanced breast cancer
2 (7.5%)
[16]
Vorinostat
HDAC inhibitor
Paclitaxel
Chemotherapy
Phase I/II
44 Her2-negative
24 (55%)
[17]
Vorinostat
HDAC inhibitor
Trastuzumab
Her2-targeted therapy
Phase II
11 Her2+ advanced metastatic
No response
[18]
Panobinostat (LBH589)
HDAC inhibitor
Trastuzumab
Her2-targeted therapy
Phase I
Her2+ metastatic breast cancer
 
[19]
Panobinostat (LBH589)
HDAC inhibitor
Capecitabine, lapatinib
Chemotherapy, TK inhibitor
Phase I
metastatic breast cancer
 
[20]
Entinostat (E)
HDAC inhibitor
Exemestane (e)
Endocrine therapy
Phase II
130 ER+ advanced breast cancer
OS 28.1 m (Ee) vs 19.8 m (e)
[21]
Vorinostat HDAC inhibitor Doxorubicin Chemotherapy Phase I Five advanced breast cancer 1 (20%) PR [22]

CR: Complete response; OS: Overall survival; PR: Partial response; SD: Stable disease.

Table 2. . Clinical trials using epigenetic drugs in breast cancer: ongoing studies.

Epigenetic agent Type Combined with Type Type of study Number of patients Total response Identifier [23]
Entinostat
HDAC inhibitor
Fulvestrant
Endocrine therapy
Phase II
Not open for recruitment
 
NCT02115594
Vorinostat
HDAC inhibitor
Paclitaxel, trastuzumab, doxorubicin-cyclophosphamide
 
Phase I–II
Unknown
 
NCT00574587
Vorinostat
HDAC inhibitor
Tamoxifen
Endocrine therapy
Phase II
43
No published results
NCT00365599
Entinostat
HDAC inhibitor
Exemestane
Endocrine therapy
Phase III
Recruiting
 
NCT02115282
Entinostat
HDAC inhibitor
Azacitidine
DNMT inhibitor
Phase II
60
Ongoing study
NCT01349959
Entinostat
HDAC inhibitor
Lapatinib, trastuzumab
EGFR inhibitor
Phase I
Recruiting
 
NCT01434303
Vorinostat
HDAC inhibitor
None
 
Phase I
17
Ongoing study
NCT00788112
Panobinostat (LBH589)
HDAC inhibitor
Trastuzumab, paclitaxel
Her2-targeted therapy, chemotherapy
Phase I
15
No published results
NCT00788931
Panobinostat (LBH589)
HDAC inhibitor
Trastuzumab
Her2-targeted therapy
Phase I
67
No published results
NCT00567879
entinostat
HDAC inhibitor
None
 
Phase I
75
No published results
NCT00020579
Panobinostat (LHB589)
HDAC inhibitor
Decitabine, tamoxifen
DNMT inhibitor, Endocrine therapy
Phase I/II
Recruiting
 
NCT01194908
Valproic acid
HDAC inhibitor
None
 
Phase I
Recruiting
 
NCT01007695
Vorinostat
HDAC inhibitor
Paclitaxel, bevacizumab
Chemotherapy
Phase I/II
58
Ongoing study
NCT00368875
Vorinostat
HDAC inhibitor
None
 
Phase I/II
49
Ongoing study
NCT00416130
Vorinostat
HDAC inhibitor
Carboplatin, paclitaxel
Chemotherapy
Phase II
74
Ongoing study
NCT00616967
Vorinostat
HDAC inhibitor
Trastuzumab
Her2-targeted therapy
Phase I/II
16
 
NCT00258349
Vorinostat
HDAC inhibitor
None
 
Phase II
37
No published results
NCT00132002
Vorinostat
HDAC inhibitor
Tamoxifen
Endocrine therapy
Phase II
Terminated
 
NCT01194427
Vorinostat
HDAC inhibitor
Ixabepilone
chemotherapy
Phase I
56
Ongoing study
NCT01084057
Vorinostat
HDAC inhibitor
Lapatinib
 
Phase I/II
12
Terminated
NCT01118975
Vorinostat
HDAC inhibitor
Anastrozole, letrazole, exemestane
Endocrine therapy
Phase I
Recruiting (14)
 
NCT01720602
Vorinostat
HDAC inhibitor
Anastrozole, letrazole, exemestane
Endocrine therapy
Phase I
Eight
No published results
NCT01153672
Vorinostat
HDAC inhibitor
Carboplatin, paclitaxel
Chemotherapy
Phase I
Recruiting
 
NCT01249443
Valproic acid
HDAC inhibitor
None
 
Phase I
Recruiting
 
NCT01010958
Vorinostat HDAC inhibitor Radiation therapy   Phase I 17 Ongoing study NCT00838929

Below we summarize the epigenetic changes that are involved in breast cancer.

DNA methylation & breast cancer

DNA methylation is arguably the most important epigenetic modification in mammalian cells. It regulates gene expression associated with normal development and growth and is dysregulated in malignancies [24]. DNA methylation of CpG islands is catalyzed by DNMTs including DNMT1, DNMT3a and DNMT3b. DNMT1 is required for maintenance of methylation during DNA replication in mitosis of normal cells. Its deficiency may lead to global hypomethylation. DNMT3a and DNMT3b are implicated in the generation of de novo methylation patterns [25]. The expression levels of DNMT1, DNMT3a and DNMT3b has been shown to be elevated in breast cancer compared with normal breast tissue [26]. DNMT3b gene has shown the highest range of expression compared with DNMT1 and DNMT3a suggesting that DNTM3b is the main player in breast cancer [26]. Additionally, a family of MeCP-MBD binds to methylated cytosines on DNA and also modifies transcription [27]. For example, MeCP2 binds methylated DNA in vitro and in vivo. It contains a methyl-CpG-binding domain at its N-terminus and a transcription repression domain in its central region that interacts directly with maintenance DNMT1 forming a complex that recruits HDACs and may thus influence both DNA methylation, as well as histone acetylation and nucleosomal positioning (Figure 1). MBD1 and MBD2 also suppress transcription, but MBD4 is a DNA glycosylase involved in DNA mismatch repair. Last, NuRD chromatin remodeling complex are a group of proteins with chromatin remodeling and HDAC activity that interact with MBD2 to methylate DNA [28].

In the human genome approximately 70% of all CpG islands are methylated and located in tightly packed core regions of DNA where they control gene silencing and chromosomal stability. In contrast, CpG islands that remain unmethylated are found in relaxed, open, frequently promoter regions of DNA, which allows access to transcription factors and other regulatory proteins for the expression of housekeeping and regulatory genes. In normal cells, CpG islands in promoters of tumor-suppressor genes are usually unmethylated and therefore transcriptionally active. However CpG island hypermethylation in promoters of tumor-suppressor genes is common in malignancies. Over the past decade, multiple studies have identified and characterized DNA methylation patterns and their relationship to breast cancer development and progression [29]. A number of genes that have consistently been reported to be methylated and consequently silenced, include ones involved in cell cycle regulation (RASSF1A, CDKN2ACDKN1B and CCND2), DNA repair (BRCA1, MLH1 and MGMT), cell detoxification (GSTP1), apoptosis (HOXA5 and TMS1), cell adhesion and invasion (TWIST, CDH1 and TIMP3) and hormone receptors (ESR1 and PGR) [9].

Ordway et al. identified 220 differentially DNA methylated loci in malignancies, a subset of which appears to distinguish breast cancers from normal and benign tissues [30]. A recent genome-wide study by Fang et al. demonstrates that a coordinated pattern of hypermethylation at a large number of genes, referred to as ‘CpG island methylator phenotype’ exists in breast cancers [31]. This phenotype is protective and is characterized by a distinct epigenomic profile associated with low metastatic risk and survival. Its absence predicts high metastatic risk and death [31]. Other studies describe DNA methylation signatures that identify molecular breast cancer subtypes. For example, Holm et al. report that luminal B tumors are more frequently methylated than basal-like or triple-negative breast cancers [32]. In general, it appears that methylation plays a significant role in different subsets of breast cancers and it will be critical to understand the mechanism(s) that drive various methylation states in order to target them therapeutically [9]. It has been recently reported that the DNA methylation pattern of endocrine-resistant cancer could provide accurate biomarkers for detection and prediction of response to therapy [6]. Of importance is the fact that drugs specifically targeting various enzymes involved in epigenetic modifications are being designed and tested.

Histone modifications & breast cancer

Post-translational modifications of histone tails can involve phosphorylation, ubiquitination and SUMOylation, but acetylation/deacetylation and methylation are the best characterized in terms of their role in modifying gene expression. HDACs remove the acetyl groups from ϵ-amino groups of lysine residues in the N-terminal tails of core histones. This compacts chromatin into tightly ordered nucleosomes and prevents access of transcription factors to DNA. HATs acetylate the lysines, relaxing chromatin and allowing transcription factor binding (Figure 1). Histones can also be methylated, which generally turns genes ‘off,’ or demethylated, which turns them ‘on,’ by tightening or loosening or histone tails, respectively. This restricts or allows transcription factor loading onto DNA. HDACs and HATs are classified into several families that catalyze distinct cellular pathways [33].

Histone deacetylation & HDAC inhibitors

HDACs fall into two classes based on their structure: zinc-dependent class I, IIa, IIb and IV; and zinc-independent class III (also called sirtuins). Based on their chemical structure, HDAC inhibitors are divided into four groups: hydroxamic acids, cyclic peptides, short-chain fatty acids and benzamides. Some of these derepress silenced genes, slowing cancer cell growth and promoting apoptosis [34]. Several Phase I and II clinical trials are underway to evaluate vorinostat and other HDAC inhibitors such as entinostat and panobinostat (LBH-589) for the treatment of breast cancers, including their use in combination with standard cytotoxic (paclitaxel) and endocrine (tamoxifen) therapies; or in combination with therapies targeted at HER2 (Herceptin; trastuzumab) or VEGF (Avastin; bevacizumab) (Tables 1 & 2; [23]). Combination therapies using HDAC inhibitors plus DNMT inhibitors synergistically re-express silenced genes producing apoptosis in colon and lung cancer cell lines and decreasing tumor formation in lung cancer models [35]. Histone methylation effects on hormone-responsive breast cancers are reviewed below.

Histone acetylation & HAT inhibitors

HATs are divided into three classes based on their sequence homology: GNAT, MYST and orphan HATs that include p300/CBP and steroid receptor coactivators (SRCs). HAT inhibitors suppress the catalytic activity of the acetyl transferases. However, only a small number of HAT inhibitors have been described or investigated [36]. They are classified into bisulfate inhibitors, natural products and synthetic small molecules. Some of these prevent growth of cancer cells. Anacardic acid, isolated from the shells of cashew nuts, is a potent in vitro inhibitor of both p300 and PCAF's HAT activity [37]. Because cells are poorly permeable to anacardic acid, synthetic analogs are being analyzed for their HAT-inhibitory activity and effects on cancer cells [38]. Curcumin, a natural dietary product that modulates enzymatic activities of HATs is discussed below.

Histone methylation & demethylation

Histone methylation is restricted to lysine (K) and arginine (R) residues but is most common on lysines. It is reversed by lysine methytransferases and demethylases. Methylation of histone H3 lysine 4 (H3K4), H3K36 or H3K79 is associated with active transcription, whereas methylation of H3K9, H3K20 or H3K27 is associated with gene silencing (Figure 1) [39].

H3K27 is methylated by EZH2, a Polycomb group protein and highly conserved histone methyltransferase that functions as a transcriptional repressor. EZH2 overexpression is strongly associated with aggressive and metastatic breast cancers [40]. 3-Deazaneplanocin (DZNep), an inhibitor of the EZH2 induces apoptosis in breast cancer cells but not in normal cells [41]. Tanshindiols are small molecule inhibitors of EZH2 that also possess anticancer activity in several tumor cell lines [42]. Last, inhibitory EZH2 peptides have been designed among which one termed SQ037 has been validated and shown to have considerable anti-EZH2 potency [43]. These reagents demonstrate the specificity that can be engineered to generate drugs that precisely target epigenomic enzymes and would be expected to have the desired efficacy with minimal side effects.

H3K4 is targeted by the specific methyltransferase SMYD3, which is also overexpressed in several malignancies including breast cancers. Silencing of SMYD3 by small interfering RNAs inhibits growth of these cancer cells [44]. Similarly, Novobiocin decreases SMYD3 expression and inhibits the proliferation and migration of MDA-MB-231 breast cancer cells. Another potent H3K4 methylase is tranylcypromine. This small molecule demethylation inhibitor de-represses transcription of important target genes including the pluripotent stem cell marker Oct4 [45].

H3K4 is demethylated by LSD1, which also demethylates nonhistone proteins such as p53 and DNMT1, indicating that it has broad biological functions [46]. Overexpression of histone-modifying enzymes such as LSD1 and EZH2 silences critical genes, including tumor suppressor genes. Their inactivation is thought to play an important role in the genesis of breast and other cancers. On the other hand, LSD1 is highly expressed in ER- breast cancers where it is a biomarker of aggressiveness [47], so its regulation with regard to malignancies requires more study. A series of novel inhibitors of histone methylation or demethylation induces re-expression of silenced genes [9]. Potent pharmacologic inhibitors of LSD1 include biguanide and bisguanidine polyamine analogs. They promote re-expression of silenced genes of the GATA family of transcription factors that are important in the development of colon cancers [48]. These LSD1 inhibitors alter promoter activity of multiple genes in breast cancer cells and are postulated to have considerable therapeutic potential [49].

H3K9 is methylated by G9a (also called EHMT2), a methyltransferase that promotes cancer aggressiveness by silencing tumor suppressor and other genes [50]. Inhibition of G9a reduces the invasiveness and metastatic potential of human lung cancer cells [51] and inhibits the growth of prostate cancer cells [52]. Treatment of cancer cells with the G9a inhibitors BIX01294 or BRD4770 decreases cellular H3K9 methylation [53] and inhibits proliferation, motility and invasiveness of human neuroblastoma cells [54] and pancreatic cancer cell [55], respectively. In vitro knockdown of G9a inhibits migration and invasion of breast cancer cells [56].

microRNAs

miRNAs are short, naturally occurring noncoding RNA molecules averaging 18–22 nucleotides in length that epigenetically regulate gene expression by binding to the 3′ untranslated region (UTR) of target mRNAs and inhibit their translation. Compared with normal tissues, miRNA expression patterns are altered in malignancies. Using miRNA profiling, Iorio et al. defined a miRNA signature that is differentially expressed in breast cancers compared with normal mammary tissues. It includes miRNA-10b, miRNA-125b and miRNA-145, which are downregulated, and miRNA-21 and miRNA-155, which are upregulated, suggesting that at normal levels they act as tumor suppressors or oncogenes, respectively [57]. The same authors report that putative targets of miRNA-125b are the oncogenes YES, ETS1, TEL and AKT3; the FGFR2; and members of MAPK pathway (MAP3K10, MAP3K11 and MAPK14) [57]. On the other hand upregulated miRNA-21 targets the tumor suppressors PDCD4 and PTEN [58]. This study also showed that miRNA-30 expression is correlated with ER and PR levels, and that miRNA let-7 isoforms regulate PR status (let-7c), lymph node metastasis (let-7f-1, let-7a-3, let-7a-2) and proliferation indices (let-7c, let-7d). miRNA let-7 also appears to be a tumor suppressor that is downregulated in breast cancer stem cells (CSC) [59]. Because of these important regulatory functions it is not surprising that the scientific and medical communities are clamoring for reagents that specifically target select miRNAs for both experimental and clinical purposes (reviewed in [60]). Indeed, miRNA inhibitors based on anti-miRNA oligonucleotides are under development while miRNA induction can be achieved with modified miRNA mimetics; either introduced into cells as plasmids or encoded in lentiviral vectors [60]. Their ability to very specifically target a desired miRNA has enormous potential for therapies.

Estrogen receptors

Most HER2+ and all basal-like and Claudin-low tumors lack ER and are not candidates for endocrine therapies. Among luminal tumors, presence of ER is the main predictor of response to endocrine therapies and ER loss is a major mechanism for acquired resistance [5]. As discussed above, multiple genomic factors can explain ER loss including ER mutations; overexpression and signaling by EGFR, HER2 or other growth factors; or dysregulation or inactivation of ER at its promoter by tumor suppressors like pRb2/p130 or p53 [61]. For instance, with regard to ER, mutation of the ESR1 gene that encodes it is significantly higher (from 10 to >50% has been reported) in endocrine-resistant metastatic breast cancers compared with untreated primary tumors. Nearly all ESR1 mutations localize to the ligand-binding domain (LBD) and often result in constitutively active ER. Full discussion of ER mutations is outside the scope of this review. For details, the reader is referred to recent reviews by Fuqua et al. and Alluri et al. [62,63]. Epigenetic mechanisms including DNA methylation and histone deacetylation of the ER promoter are additional mechanisms for ER loss. This is clearly demonstrated by studies in which treatment of ER- human breast cancer cell lines with DNMT and/or HDAC inhibitors results in re-expression of ER and re-sensitizes them to endocrine therapies [64]. For example, inhibition of DNMT1-dependent methylation using an antisense oligonucleotide leads to re-expression of ER in ER- breast cancer cell lines [65]. Paradoxically, in ER+ human breast cancer cell lines, HDAC inhibitors promote transcriptional downregulation of ERs and their responsive genes, while in ER- cells, HDAC inhibitors can re-establish ER expression and resensitize them to antiestrogens. For example, ERs are restored to ER- MDA-MB-231 and MDA-MB-435 cells by the HDAC inhibitor LBH589 showing that chromatin reorganization plays a role in ER loss [66]. In xenografts of MCF-7 cells resistant to the aromatase inhibitor (AI) letrozole, the HDAC inhibitor entinostat restores letrozole sensitivity by modulating HER-2 expression and activity [67].

Alterations in methylation patterns not only influence ER responsiveness, but also alter responses of E-regulated genes. For instance, the developmental gene HOXC10 is E-regulated, and hypermethylation of its promoter is associated with resistance to AI therapies [68]. Prevention of AI-induced histone and DNA methylation may be beneficial in blocking or delaying AI resistance [68]. Another example is LSD1, which is highly expressed in ER- breast cancers [47]. Surprisingly, it also associates with most ER-promoter-specific targets and its binding is required for E-dependent activation of those genes at the same time that histone demethylation events are observed. It is postulated that this prevents constitutive activation by unliganded ER [69].

Some studies report that ER expression can be reactivated by combining the DNMT inhibitor AZA, and the HDAC inhibitor trichostatin A [70]. Combination therapies using HDAC inhibitors plus DNMT inhibitors synergistically increase expression of silenced genes and decrease tumor formation in several human cancer models [35,71]. In ER- breast cancers this combination results in synergistic re-expression of ER and restoration of response to endocrine therapy [72]. Compared with each alone, the combinations are also superior for ER re-expression and restoration of tamoxifen responsiveness in breast cancer models [72]. These data suggest that DNMT inhibitor plus HDAC inhibitor combinations provide a new treatment approach for patients with tamoxifen resistant and ER- disease.

ER levels in the context of breast cancers are also post-transcriptionally regulated by miRNAs, whose levels can oscillate between repression and stimulation in response to multiple factors. Micro RNAs that are upregulated in ER- states are thought to inhibit ER expression. Several have been described including miRNA-22, miRNA-206 and miRNA-221/222 [73]. Other miRNAs upregulated in ER- states include miRNA-185, miRNA-206, miRNA-212 and miRNA-520g [57]. Some miRNAs are downregulated in ER- states including miRNA-342–3p, miRNA-26a, miRNA-29b, miRNA-30a-5p, miRNA-30b, miRNA-30c, miRNA-30d and miRNA-191 [57,74]. These micRNAs offer multiple targets for therapeutics.

Clinically, epigenetic therapies for breast cancers are still in early stages. Several Phase I and II clinical trials have been performed using DNMT and/or HDAC inhibitors [23]. The HDAC inhibitors vorinostat, panobinostat and entinostat alone or in combination with other therapeutic agents such as endocrine therapy (the antiestrogen tamoxifen or aromatase inhibitors) or chemotherapy are being tested in various trials that are summarized in Tables 1 & 2.

Progesterone receptors

Progesterone Receptors have long been known as markers of endocrine responsiveness in ER+ breast cancers. It is clear that patients with ER+PR- tumors have a worse prognosis than ones with ER+PR+ tumors. Since PRs are E-regulated proteins, such fluctuations in PR levels are commonly ascribed to functional anomalies in ER-dependent transcription. However, both preclinical and clinical studies demonstrate that even when tumors are ER+, absence of PR is associated with resistance to tamoxifen and poor prognosis [75]. These and other data clearly suggest that besides their role as “markers” of E action, progesterone and its receptors have separate functional roles in breast cancers that remains to be explored in detail. Indeed, PRs are regulated not only by E and ER but also by other signaling pathways as well as by epigenetic mechanisms. Kinase signaling alters PR at both transcriptional and post-translational levels. For instance in MCF-7 cells, activation of the PI3K/AKT/mTOR pathway decreases PR mRNA and protein without altering ER levels or activity [76]. Similarly, PTEN loss, which activates PI3K, also decreases PR in a number of cell lines and human breast tumors [77]. With regard to epigenetic mechanisms of PR regulation, one study reports a lack of association between PR promoter methylation and PR expression levels [78]. In contrast, several studies demonstrate that PR loss is associated with promoter methylation [79]. Inhibition of HDAC activity in ER-;PR- breast cancer cells reactivates both ER and PR expression [80]. Similarly, the DNMT inhibitor zebularine induces ER and PR transcripts in ER-PR- MDA-MD-231 cells [81]. In both studies the effects on PR could theoretically be indirect, as a response to ER reactivation. However, in ER+ PR- T47D-Y breast cancer cells, treatment with AZA and trichostatin A restores PR; an effect that appears to be direct [Abdel-Hafiz H and Horwitz KB; Unpublished Data]. Clinically, methylation of the PR promoter is higher in PR- than in PR+ tumors [80] and PR methylation has been observed in endocrine-resistant tumors [2]. One study shows that this is correlated with HER2 overexpression [82]. This interesting relationship suggests a role for methylation in development of the small subset of luminal B tumors characterized by ER positivity, low or negative PR and elevated HER2.

Human PRs exist as two functionally different isoforms called PR-A and PR-B, which are the result of transcription from two alternative promoters, and initiation of translation at two different AUG codons [83]. In the normal adult human breast, PR-A and PR-B are co-expressed in equimolar ratios, and bind to DNA either as homodimers or heterodimers. This equimolarity is deregulated in malignancies [84]. Breast cancer patients with PR-A-rich tumors reportedly have poorer disease-free survival rates and more rapid recurrences after tamoxifen treatment than patients with PR-B-rich tumors [85]. Assuming that PR play a functional role in breast cancers beyond that of markers of estrogen action, then this is not surprising since pure (presumably homodimeric) progesterone-liganded PR-B regulate different gene sets and control different cellular responses than pure liganded PR-A [86]. It remains unclear which genes are involved in the putative beneficial effects of PR-B or harmful effects of PR-A.

With regard to mechanisms responsible for uniform reduction of both isoforms that lower total PR levels in luminal B tumors, this could be a consequence of equivalent methylation of each promoter [80]. However if methylation of the two promoters is dysregulated, expression of the two isoforms will become unequal [87]. Using tumor DNA from breast cancer patients treated with tamoxifen after surgery, Pathiraja and colleagues studied the relationship between PR-A and PR-B promoter methylation, and PR expression levels and clinical outcome. They found that along with the fact that low overall PR expression was associated with worse outcome, methylation of the PR-A but not PR-B promoter was responsible for tamoxifen resistance and a worse outcome [80]. They propose that this decreases the PR-A/PR-B ratio leading to PR-B dominance [80]. However, Gaudet et al. find no significant correlation between PR-A and PR-B methylation and protein expression levels in breast tumors [88].

Functionally, it remains unclear how dysregulation of the PRA/PRB ratio contributes to tamoxifen resistance in breast cancers since studies addressing this question have yielded contradictory results. PR-B are stronger transactivators than PR-A both by regulating more genes and, generally but not always, by having a more robust effect on genes co-regulated by PR-A. Additionally, some genes are regulated by one but not the other isoform [86]. One hypothesis holds that when PR-B are unopposed by PR-A, PR-B upregulate growth factor or other genes that heighten tumor aggressiveness [89]. Alternatively, PR-B may function indirectly by suppressing ER signaling. For example, unliganded PR-B, but not PR-A, have distinct antiestrogenic effects on E-induced cell growth, and ER interactions with estrogen response elements in promoters of ER-regulated genes [90]. We have shown that PR-A can suppress the ligand-dependent transcriptional activity of PR-B and other nuclear receptors, including ER. In this scenario, loss of PR-A would increase the activity of both liganded PR-B and ER [91]. How this contributes to endocrine resistance is unknown. In contrast to the above, Hopp et al. report that resistance to endocrine therapies targeting ER is associated with heightened PR-A/PR-B ratios [85]. Last, at present PR in breast cancers are in the main thought to be markers of ER function. They are rarely thought of as therapeutic targets; as with antiprogestins for instance. However, Wargon and coworkers have shown that acquired antiprogestin-resistant mammary gland tumors in mice have low PR-A levels, and conclude that PR-A might be direct targets of antiprogestins [92]. These authors subsequently showed that PR-A expression was silenced by DNA methylation in the resistant tumors and that treatment with the DNA methylase inhibitor AZA restores PR-A and reverses resistance [93]. In sum, much work remains to be done to understand whether in breast cancers, epigenetic mechanisms control the ratio of PR isoforms, and how their dysregulation influences estrogen signaling and outcome to hormone therapies.

Epigenetics & breast cancer stem cells

Cellular heterogeneity within breast cancers and the putative existence of breast cancer stem cells (CSCs) are also possible reasons for therapeutic failures. Assuming they exist, breast CSCs are predicted to be a minor (possibly <1%) nonproliferative precursor cell subpopulation within a tumor, able to give to rise to, and maintain, more differentiated downstream cell types within a tumor, but resistant to endocrine, radiation and chemotherapies that target the more differentiated cells [94–96]. If so, therapeutic approaches that kill CSCs may be the best hope for curing cancers. However, with regard to human breast CSCs, much uncertainty remains about their properties, markers that identify them and even whether there is a single definable breast CSC, or whether there are multiple CSCs that underlie breast cancer subtypes.

DNA methylation

Epigenetic modifications appear to be involved in the evolution, maintenance and function of breast CSCs although information remains sparse. With regard to DNA methylation our understanding is limited. Most published research focuses on CSCs of organs other than breast. DNA methylation has a mechanistic link to hematopoietic stem cells where it targets pluripotency factors; [97] and a link to colon, ovarian, blood, prostate and brain cancers where it targets CD133 [98–101]. The Polycomb repressive complex, which represses gene expression through histone modification and chromatin compaction modifies CSCs of the breast [102], prostate [103], ovary [104] and glioblastomas [105]. The association between DNA methylation and CSCs suggests that hypomethylating agents have the potential to induce CSC differentiation generating cells that would be sensitive to standard therapeutic agents [106]. A methyltransferase inhibitor, DZNep, can disrupt the Polycomb 2 complex and reduce CSCs in acute myeloid leukemia [107], hepatocellular carcinoma [108], glioblastoma [109] and prostate cancer [103].

Histones

In addition to targeting regulatory genes in differentiated cells, global histone modifications that alter nucleosomal positioning also play a role in CSC biology. As mentioned above, methylation of H3K27 represses transcription, while methylation of H3K4 stimulates transcription [97]. Inhibitor of LSD1, the enzyme responsible for demethylating H3K4, acts specifically on embryonal CSCs of pluripotent cancers such as teratocarcinomas, seminomas and embryonic carcinomas, suggesting that H3K4 demethylation is involved in formation of such tumors [110].

MicroRNAs

Several studies have identified unique gene expression profiles for putative breast CSCs compared with nontumorigenic cells, [111] including miRNAs – both oncomirs and tumor suppressors – that theoretically maintain malignancy. For example, Shimono et al. have shown that miRNA-200 isoforms a, b and c are downregulated in breast CSCs whose targets include stem cell self-renewal factor BMI1, and the transcriptional repressors of E-cadherin, ZEB1/ZEB2 [112]. Lin28, an RNA-binding protein that induces specific miRNA uridylation and blocks miRNA processing by Dicer, mediates downregulation of miRNA let-7, miRNA-200c and miRNA-107 [113]. Overexpression of the miRNA-103/107 family induces EMT and increases the risk of metastases in breast cancer patients [114]. EMT has been directly linked to generation of cells with CSC-like properties [115]. Other miRNAs reportedly downregulated in breast CSCs include miRNA-30, miRNA-128, miRNA-34c, miRNA-34a, and miRNA-16; and upregulated include miRNA-181 and miRNA-495 [111].

Histone acetylation reportedly plays a role in regulating CSC microRNAs. For example, miR-34a is downregulated in pancreatic CSCs. Treatment with the HDAC inhibitor vorinostat restores miR-34a levels and decreases CSC viability. In breast cancers, miR-34a suppresses HDAC1 and HDAC7 levels and its expression is inversely correlated with HDAC1 and HDAC7 activity, tumor grade and stage [116]. Low miR-34a expression combined with high HDAC1 and HDAC7 that deacetylate HSP70K 246 is thought to be a molecular marker of aggressive and therapy-resistant breast cancer [117].

Novel epigenetic therapeutics: dietary polyphenols

Cancers linked to epigenetic modifications can be influenced by environmental and dietary factors, drugs, pesticides and inorganic contaminants [118]. Dietary phytochemicals present in fruits, vegetables, beverages and spices may possess anticancer properties although in most cases their efficacy in humans is unproven. These include tea polyphenols (green and black tea), genistein (soybean), curcumin (turmeric), sulforaphane (cabbage), phenyl isothiocyanate (watercress), lycopene (tomato), resveratrol (red grape), quercetin (capers), indol-3-carbinol (broccoli), ellagitanin (respberry) and organosulfur compounds (garlic). Several mechanism(s) have been proposed for their beneficial effects including detoxification of enzymes, antioxidant effects, alterations in signaling such as that of the NFκB pathway and nuclear receptor signaling by acting as agonists or antagonists or by modifying ligands through their enzymatic actions [119]. Additionally, some phytochemicals target epigenetic modifying enzymes such as DNMTs and HDACs, as three examples below show. For more detailed discussions, the reader is referred to a recent review by Shankar et al. [118].

Epigallocatechin-3-galate (EGCG), the major catechin present in green and black teas, has the potential to reduce the risk of several diseases including cancer [120]. EGCG inhibits DNMT activity and reactivates methylation-silenced genes [121]. Additionally, EGCG inhibits DNA and RNA synthesis by suppressing dihydrofolate reductase activity. Several studies have reported a correlation between EGCG consumption and inhibition of cancers, including ones of the ovary, mouth, esophagus or stomach, breast, prostate, skin, colon/rectum, pancreas and head and neck [118].

Resveratrol is a stilbenoid polyphenol found in grape skins and other fruits. It has been linked to anticancer, anti-inflammatory and blood sugar lowering activity [122] and is thought to be a regulator of signaling pathways that control cell division, cell growth, apoptosis, angiogenesis and tumor metastasis [123]. The evidence for these benefits in humans is slim. Resveratrol is of interest in breast cancers because in vivo it increases endogenous testosterone production resulting from selective ER modulatory and aromatase inhibitor activities [124]. It appears to be a pan-HDAC inhibitor targeting class III HDAC, SIRT1, SIRT2, SIRT3 and p300 [125], and altering histone acetylation in hepatoblastomas. In MCF-7 breast cancer cells, resveratrol alone exhibits weak DNMT inhibitory activity that is however, insufficient to reverse the methylation of several tumor suppressor genes. On the other hand in combination with the adenosine analog 2-chloro-2′-deoxyadenosine, resveratrol inhibits methylation of the RARb2 gene promoter [126].

Curcumin is the principal curcuminoid of the spice turmeric, a member of the ginger family. The curcuminoids are natural phenols that reportedly modulate intracellular signaling pathways involved in inflammation, proliferation, invasion, survival and apoptosis [127]. Curcuminoids cause global hypomethylation in human leukemia MV4–11 cells [128]. They inhibit DNMT1 by covalently blocking the catalytic thiol group-binding site [129]. Curcumin is a potential modulator of histones and modulates enzymatic activities of both HATs and HDACs. Several studies demonstrate that curcumin inhibits expression of class I HDACs [130]. Other mechanisms for the anticancer activity of curcumin may include regulation of miRNAs involved in carcinogenesis [131].

Conclusion & future prespective

Because hormone-dependent breast cancers represent the majority of cases, intrinsic or acquired resistance to endocrine therapies are major challenges to physicians treating patients with this disease. Considerable research has focused on understanding mechanisms of hormone resistance, on identifying biomarkers of the resistant state, and on defining alternative molecular targets for therapeutics that bypass resistance pathways. Recent insights into the issue of resistance have described epigenetic modifications that characterize normal development and their changes in disease states including breast cancers. Indeed therapies targeting epigenetic processes appear to be very promising, and may offer alternatives or adjunct solutions to obstacles associated with endocrine resistance. For instance, HDAC and/or DNMT inhibitors have shown promising results in blocking the growth of breast cancer cells resistant to the antiestrogen tamoxifen. Additionally, Phase I and II clinical trials proffer preliminary evidence for disease stabilization or treatment response for patients in which epigenetic therapies are combined with standard therapies. Nevertheless at present no epigenetic drugs have moved into routine clinical practice. It is clear that rational patient selection coupled with appropriate drug combinations and sequencing may be critical determinants for restoring response in patients whose tumors have become resistant to conventional treatments. Such combinations should be considered for future clinical trials.

Executive summary.

Luminal breast cancer

  • Breast cancer is the most common cancer in women. It is a heterogeneous disease and presents significant challenges for treatment.

  • Luminal breast cancers, ER+ and/or PR+, represent approximately 75% of cases.

Role of epigenetic modifications in luminal breast cancer

  • Endocrine resistance is a consequence of genetic and epigenetic alterations that are valid clinical targets.

  • Epigenetic modifications are reversible, heritable and include changes in DNA methylation patterns, modification of histones and altered microRNA expression levels that target the steroid hormone receptors or their signaling pathways.

Epigenetic modifications could be targeted to restore endocrine responsiveness

  • Taking advantage of the reversibility of epigenetic modifications, epigenetic drugs, that target epigenetic modifiers, given in combination with chemotherapies or endocrine therapies, may represent promising approaches to restoration of therapy responsiveness in these cases.

Footnotes

Financial & competing interests disclosure

The authors are grateful for support from the NIH (RO1-CA026869-35), the Breast Cancer Research Foundation and the National Foundation for Cancer Research. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

No writing assistance was utilized in the production of this manuscript.

References

Papers of special note have been highlighted as: • of interest; •• of considerable interest

  • 1.Prat A, Karginova O, Parker JS, et al. Characterization of cell lines derived from breast cancers and normal mammary tissues for the study of the intrinsic molecular subtypes. Breast Cancer Res. Treat. 2013;142(2):237–255. doi: 10.1007/s10549-013-2743-3. [DOI] [PMC free article] [PubMed] [Google Scholar]; •• Data presented in this paper help to improve our understanding of the wide range of breast cancer cell line models through the appropriate pairing of cell lines with relevant in vivo tumor and normal cell counterpart.
  • 2.Badia E, Oliva J, Balaguer P, Cavailles V. Tamoxifen resistance and epigenetic modifications in breast cancer cell lines. Curr. Med. Chem. 2007;14(28):3035–3045. doi: 10.2174/092986707782794023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Milani A, Geuna E, Mittica G, Valabrega G. Overcoming endocrine resistance in metastatic breast cancer: current evidence and future directions. World J. Clin. Oncol. 2014;5(5):990–1001. doi: 10.5306/wjco.v5.i5.990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Bianco S, Gevry N. Endocrine resistance in breast cancer: from cellular signaling pathways to epigenetic mechanisms. Transcription. 2012;3(4):165–170. doi: 10.4161/trns.20496. [DOI] [PMC free article] [PubMed] [Google Scholar]; • Reviews signaling pathways implicated in endocrine resistance and try to merge them with recent epigenetic studies.
  • 5.Garcia-Becerra R, Santos N, Diaz L, Camacho J. Mechanisms of resistance to endocrine therapy in breast cancer: focus on signaling pathways, miRNAs and genetically based resistance. Int. J. Mol. Sci. 2013;14(1):108–145. doi: 10.3390/ijms14010108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Stone A, Valdes-Mora F, Clark SJ. Exploring and exploiting the aberrant DNA methylation profile of endocrine-resistant breast cancer. Epigenomics. 2013;5(6):595–598. doi: 10.2217/epi.13.70. [DOI] [PubMed] [Google Scholar]; • Exploring the DNA methylation profile of endocrine-resistant cancer could potentially provide clinically relevant biomarkers to guide new approaches to detection and therapy, as well as advance our understanding of the mechanisms underlining this disease phenotype.
  • 7.Maruyama R, Choudhury S, Kowalczyk A, et al. Epigenetic regulation of cell type-specific expression patterns in the human mammary epithelium. PLoS Genet. 2011;7(4):e1001369. doi: 10.1371/journal.pgen.1001369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.de Souza Rocha Simonini P, Breiling A, Gupta N, et al. Epigenetically deregulated microRNA-375 is involved in a positive feedback loop with estrogen receptor alpha in breast cancer cells. Cancer Res. 2010;70(22):9175–9184. doi: 10.1158/0008-5472.CAN-10-1318. [DOI] [PubMed] [Google Scholar]
  • 9.Huang Y, Nayak S, Jankowitz R, Davidson NE, Oesterreich S. Epigenetics in breast cancer: what's new? Breast Cancer Res. 2011;13(6):225. doi: 10.1186/bcr2925. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Stathis A, Hotte SJ, Chen EX, et al. Phase I study of decitabine in combination with vorinostat in patients with advanced solid tumors and non-Hodgkin's lymphomas. Clin. Cancer Res. 2009;27(15S) [PMC free article] [PubMed] [Google Scholar]
  • 11.Arce C, Perez-Plasencia C, Gonzalez-Fierro A, et al. A proof-of-principle study of epigenetic therapy added to neoadjuvant doxorubicin cyclophosphamide for locally advanced breast cancer. PLoS ONE. 2006;1:e98. doi: 10.1371/journal.pone.0000098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Candelaria M, Gallardo-Rincon D, Arce C, et al. A Phase II study of epigenetic therapy with hydralazine and magnesium valproate to overcome chemotherapy resistance in refractory solid tumors. Ann. Oncol. 2007;18(9):1529–1538. doi: 10.1093/annonc/mdm204. [DOI] [PubMed] [Google Scholar]
  • 13.Kelly WK, O'Connor OA, Krug LM, et al. Phase I study of an oral histone deacetylase inhibitor, suberoylanilide hydroxamic acid, in patients with advanced cancer. J. Clin. Oncol. 2005;23(17):3923–3931. doi: 10.1200/JCO.2005.14.167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Luu TH, Morgan RJ, Leong L, et al. A Phase II trial of vorinostat (suberoylanilide hydroxamic acid) in metastatic breast cancer: a California Cancer Consortium study. Clin. Cancer Res. 2008;14(21):7138–7142. doi: 10.1158/1078-0432.CCR-08-0122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Munster PN, Thurn KT, Thomas S, 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. 2011;104(12):1828–1835. doi: 10.1038/bjc.2011.156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Wardley A, McCaffery J, Crown J, et al. Phase II data for entinostat, a class 1 selective histone deacetylase inhibitor, in patients whose breast cancer is progressing on aromatase inhibitor therapy. J. Clin. Oncol. 2010;28(Suppl. 15):Abstract 1052. [Google Scholar]
  • 17.Ramaswamy B, Fiskus W, Cohen B, et al. Phase I–II study of vorinostat plus paclitaxel and bevacizumab in metastatic breast cancer: evidence for vorinostat-induced tubulin acetylation and Hsp90 inhibition in vivo. Breast Cancer Res. Treat. 2012;132(3):1063–1072. doi: 10.1007/s10549-011-1928-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Swaby R, Sparano JA, Bhalla K, et al. A Phase II study of the histone deacetylase inhibitor, vorinostat, in combination with trastuzumab in patients with advanced metastatic and/or local chest wall recurrent HER-2 amplified breast cancer resistant to transtuzumab- containing therapy: (E1104) a Trial of the Eastern Cooperative Oncology Group. Cancer Res. 2009;69(24 Suppl.):Abstract nr 5084. [Google Scholar]
  • 19.Conte P, Campone M, Pronzato P, et al. Phase I trial of panobinostat (LBH589) in combination with trastuzumab in pre-treated HER2-positive metastatic breast cancer (mBC): preliminary safety and tolerability results. J. Clin. Oncol. (Meeting Abstracts) 2009;27(15 Suppl.):1081. [Google Scholar]
  • 20.Peacock N, Jones SE, Yardley D, et al. A Phase I study of panobinostat (LBH589) with capecitabine with or without lapatinib. J. Clin. Oncol. 2010;28(15 Suppl.):Abstract 1115. [Google Scholar]
  • 21.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):2128–2135. doi: 10.1200/JCO.2012.43.7251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Munster PN, Marchion D, Thomas S, et al. Phase I trial of vorinostat and doxorubicin in solid tumours: histone deacetylase 2 expression as a predictive marker. Br. J. Cancer. 2009;101(7):1044–1050. doi: 10.1038/sj.bjc.6605293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.ClinicalTrails.gov. http://clinicaltrials.gov
  • 24.Jelinic P, Shaw P. Loss of imprinting and cancer. J. Pathol. 2007;211(3):261–268. doi: 10.1002/path.2116. [DOI] [PubMed] [Google Scholar]
  • 25.Portela A, Esteller M. Epigenetic modifications and human disease. Nat. Biotechnol. 2010;28(10):1057–1068. doi: 10.1038/nbt.1685. [DOI] [PubMed] [Google Scholar]
  • 26.Girault I, Tozlu S, Lidereau R, Bieche I. Expression analysis of DNA methyltransferases 1, 3A, and 3B in sporadic breast carcinomas. Clin. Cancer Res. 2003;9(12):4415–4422. [PubMed] [Google Scholar]
  • 27.Duthie SJ. Epigenetic modifications and human pathologies: cancer and CVD. Proc. Nutr. Soc. 2011;70(1):47–56. doi: 10.1017/S0029665110003952. [DOI] [PubMed] [Google Scholar]
  • 28.Lan J, Hua S, He X, Zhang Y. DNA methyltransferases and methyl-binding proteins of mammals. Acta Biochim. Biophys. Sin. (Shanghai) 2010;42(4):243–252. doi: 10.1093/abbs/gmq015. [DOI] [PubMed] [Google Scholar]
  • 29.Khan SI, Aumsuwan P, Khan IA, Walker LA, Dasmahapatra AK. Epigenetic events associated with breast cancer and their prevention by dietary components targeting the epigenome. Chem. Res. Toxicol. 2012;25(1):61–73. doi: 10.1021/tx200378c. [DOI] [PubMed] [Google Scholar]
  • 30.Ordway JM, Budiman MA, Korshunova Y, et al. Identification of novel high-frequency DNA methylation changes in breast cancer. PLoS ONE. 2007;2(12):e1314. doi: 10.1371/journal.pone.0001314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Fang F, Turcan S, Rimner A, et al. Breast cancer methylomes establish an epigenomic foundation for metastasis. Sci. Transl. Med. 2011;3(75):75ra25. doi: 10.1126/scitranslmed.3001875. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Holm K, Hegardt C, Staaf J, et al. Molecular subtypes of breast cancer are associated with characteristic DNA methylation patterns. Breast Cancer Res. 2010;12(3):R36. doi: 10.1186/bcr2590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Sawan C, Herceg Z. Histone modifications and cancer. Adv. Genet. 2010;70:57–85. doi: 10.1016/B978-0-12-380866-0.60003-4. [DOI] [PubMed] [Google Scholar]
  • 34.Marson CM. Histone deacetylase inhibitors: design, structure-activity relationships and therapeutic implications for cancer. Anticancer Agents Med. Chem. 2009;9(6):661–692. doi: 10.2174/187152009788679976. [DOI] [PubMed] [Google Scholar]
  • 35.Belinsky SA, Grimes MJ, Picchi MA, et al. Combination therapy with vidaza and entinostat suppresses tumor growth and reprograms the epigenome in an orthotopic lung cancer model. Cancer Res. 2011;71(2):454–462. doi: 10.1158/0008-5472.CAN-10-3184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Gao C, Bourke E, Scobie M, et al. Rational design and validation of a Tip60 histone acetyltransferase inhibitor. Sci. Rep. 2014;4:5372. doi: 10.1038/srep05372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Balasubramanyam K, Swaminathan V, Ranganathan A, Kundu TK. Small molecule modulators of histone acetyltransferase p300. J. Biol. Chem. 2003;278(21):19134–19140. doi: 10.1074/jbc.M301580200. [DOI] [PubMed] [Google Scholar]
  • 38.Eliseeva ED, Valkov V, Jung M, Jung MO. Characterization of novel inhibitors of histone acetyltransferases. Mol. Cancer Ther. 2007;6(9):2391–2398. doi: 10.1158/1535-7163.MCT-07-0159. [DOI] [PubMed] [Google Scholar]
  • 39.Jenuwein T, Allis CD. Translating the histone code. Science. 2001;293(5532):1074–1080. doi: 10.1126/science.1063127. [DOI] [PubMed] [Google Scholar]
  • 40.Collett K, Eide GE, Arnes J, et al. Expression of enhancer of zeste homologue 2 is significantly associated with increased tumor cell proliferation and is a marker of aggressive breast cancer. Clin. Cancer Res. 2006;12(4):1168–1174. doi: 10.1158/1078-0432.CCR-05-1533. [DOI] [PubMed] [Google Scholar]
  • 41.Tan J, Yang X, Zhuang L, et al. Pharmacologic disruption of Polycomb-repressive complex 2-mediated gene repression selectively induces apoptosis in cancer cells. Genes Dev. 2007;21(9):1050–1063. doi: 10.1101/gad.1524107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Woo J, Kim HY, Byun BJ, et al. Biological evaluation of tanshindiols as EZH2 histone methyltransferase inhibitors. Bioorg. Med. Chem. Lett. 2014;24(11):2486–2492. doi: 10.1016/j.bmcl.2014.04.010. [DOI] [PubMed] [Google Scholar]
  • 43.Smadbeck J, Peterson MB, Zee BM, et al. De novo peptide design and experimental validation of histone methyltransferase inhibitors. PLoS ONE. 2014;9(4):e95535. doi: 10.1371/journal.pone.0095535. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Hamamoto R, Silva FP, Tsuge M, et al. Enhanced SMYD3 expression is essential for the growth of breast cancer cells. Cancer Sci. 2006;97(2):113–118. doi: 10.1111/j.1349-7006.2006.00146.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Luo XG, Zou JN, Wang SZ, Zhang TC, Xi T. Novobiocin decreases SMYD3 expression and inhibits the migration of MDA-MB-231 human breast cancer cells. IUBMB Life. 2010;62(3):194–199. doi: 10.1002/iub.288. [DOI] [PubMed] [Google Scholar]
  • 46.Huang J, Sengupta R, Espejo AB, et al. p53 is regulated by the lysine demethylase LSD1. Nature. 2007;449(7158):105–108. doi: 10.1038/nature06092. [DOI] [PubMed] [Google Scholar]
  • 47.Lim S, Janzer A, Becker A, et al. Lysine-specific demethylase 1 (LSD1) is highly expressed in ER-negative breast cancers and a biomarker predicting aggressive biology. Carcinogenesis. 2010;31(3):512–520. doi: 10.1093/carcin/bgp324. [DOI] [PubMed] [Google Scholar]
  • 48.Huang Y, Greene E, Murray Stewart T, et al. Inhibition of lysine-specific demethylase 1 by polyamine analogues results in reexpression of aberrantly silenced genes. Proc. Natl Acad. Sci. USA. 2007;104(19):8023–8028. doi: 10.1073/pnas.0700720104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Zhu Q, Huang Y, Marton LJ, Woster PM, Davidson NE, Casero RA., Jr Polyamine analogs modulate gene expression by inhibiting lysine-specific demethylase 1 (LSD1) and altering chromatin structure in human breast cancer cells. Amino Acids. 2011;42(2–3):887–898. doi: 10.1007/s00726-011-1004-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Huang J, Dorsey J, Chuikov S, et al. G9a and Glp methylate lysine 373 in the tumor suppressor p53. J. Biol. Chem. 2010;285(13):9636–9641. doi: 10.1074/jbc.M109.062588. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Chen MW, Hua KT, Kao HJ, et al. H3K9 histone methyltransferase G9a promotes lung cancer invasion and metastasis by silencing the cell adhesion molecule Ep-CAM. Cancer Res. 2010;70(20):7830–7840. doi: 10.1158/0008-5472.CAN-10-0833. [DOI] [PubMed] [Google Scholar]
  • 52.Kondo Y, Shen L, Ahmed S, et al. Downregulation of histone H3 lysine 9 methyltransferase G9a induces centrosome disruption and chromosome instability in cancer cells. PLoS ONE. 2008;3(4):e2037. doi: 10.1371/journal.pone.0002037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Kubicek S, O'Sullivan RJ, August EM, et al. Reversal of H3K9me2 by a small-molecule inhibitor for the G9a histone methyltransferase. Mol. Cell. 2007;25(3):473–481. doi: 10.1016/j.molcel.2007.01.017. [DOI] [PubMed] [Google Scholar]
  • 54.Lu Z, Tian Y, Salwen HR, et al. Histone-lysine methyltransferase EHMT2 is involved in proliferation, apoptosis, cell invasion, and DNA methylation of human neuroblastoma cells. Anticancer Drugs. 2013;24(5):484–493. doi: 10.1097/CAD.0b013e32835ffdbb. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Yuan Y, Wang Q, Paulk J, et al. A small-molecule probe of the histone methyltransferase G9a induces cellular senescence in pancreatic adenocarcinoma. ACS Chem. Biol. 2012;7(7):1152–1157. doi: 10.1021/cb300139y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Dong C, Wu Y, Yao J, et al. G9a interacts with Snail and is critical for Snail-mediated E-cadherin repression in human breast cancer. J. Clin. Invest. 2012;122(4):1469–1486. doi: 10.1172/JCI57349. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Iorio MV, Ferracin M, Liu CG, et al. MicroRNA gene expression deregulation in human breast cancer. Cancer Res. 2005;65(16):7065–7070. doi: 10.1158/0008-5472.CAN-05-1783. [DOI] [PubMed] [Google Scholar]
  • 58.Frankel LB, Christoffersen NR, Jacobsen A, Lindow M, Krogh A, Lund AH. Programmed cell death 4 (PDCD4) is an important functional target of the microRNA miR-21 in breast cancer cells. J. Biol. Chem. 2008;283(2):1026–1033. doi: 10.1074/jbc.M707224200. [DOI] [PubMed] [Google Scholar]
  • 59.Iliopoulos D, Polytarchou C, Hatziapostolou M, et al. MicroRNAs differentially regulated by Akt isoforms control EMT and stem cell renewal in cancer cells. Sci. Signal. 2009;2(92):ra62. doi: 10.1126/scisignal.2000356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Piva R, Spandidos DA, Gambari R. From microRNA functions to microRNA therapeutics: novel targets and novel drugs in breast cancer research and treatment (Review) Int. J. Oncol. 2013;43(4):985–994. doi: 10.3892/ijo.2013.2059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Angeloni SV, Martin MB, Garcia-Morales P, Castro-Galache MD, Ferragut JA, Saceda M. Regulation of estrogen receptor-alpha expression by the tumor suppressor gene p53 in MCF-7 cells. J. Endocrinol. 2004;180(3):497–504. doi: 10.1677/joe.0.1800497. [DOI] [PubMed] [Google Scholar]
  • 62.Fuqua SA, Gu G, Rechoum Y. Estrogen receptor (ER) alpha mutations in breast cancer: hidden in plain sight. Breast Cancer Res. Treat. 2014;144(1):11–19. doi: 10.1007/s10549-014-2847-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Alluri P, Speers C, Chinnaiyan A. Estrogen receptor mutations and their role in breast cancer progression. Breast Cancer Research. 2014;16(6):494. doi: 10.1186/s13058-014-0494-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Lustberg MB, Ramaswamy B. Epigenetic therapy in breast cancer. Curr. Breast Cancer Rep. 2011;3(1):34–43. doi: 10.1007/s12609-010-0034-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Yan L, Nass SJ, Smith D, Nelson WG, Herman JG, Davidson NE. Specific inhibition of DNMT1 by antisense oligonucleotides induces re-expression of estrogen receptor-alpha (ER) in ER-negative human breast cancer cell lines. Cancer Biol. Ther. 2003;2(5):552–556. doi: 10.4161/cbt.2.5.469. [DOI] [PubMed] [Google Scholar]
  • 66.Zhou Q, Atadja P, Davidson NE. Histone deacetylase inhibitor LBH589 reactivates silenced estrogen receptor alpha (ER) gene expression without loss of DNA hypermethylation. Cancer Biol. Ther. 2007;6(1):64–69. doi: 10.4161/cbt.6.1.3549. [DOI] [PubMed] [Google Scholar]
  • 67.Sabnis GJ, Goloubeva OG, Kazi AA, Shah P, Brodie AH. HDAC inhibitor entinostat restores responsiveness of letrozole-resistant MCF-7Ca xenografts to aromatase inhibitors through modulation of Her-2. Mol. Cancer Ther. 2013;12(12):2804–2816. doi: 10.1158/1535-7163.MCT-13-0345. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Pathiraja TN, Nayak SR, Xi Y, et al. Epigenetic reprogramming of HOXC10 in endocrine-resistant breast cancer. Sci. Transl. Med. 2014;6(229):229ra241. doi: 10.1126/scitranslmed.3008326. [DOI] [PMC free article] [PubMed] [Google Scholar]; •• Long-term aromatase inhibitor (AI) treatment results in permanent repression of HOXC10 gene via methylation and confers resistance. Therapies aimed at inhibiting AI-induced histone and DNA methylation may be beneficial in blocking or delaying AI resistance.
  • 69.Garcia-Bassets I, Kwon YS, Telese F, et al. Histone methylation-dependent mechanisms impose ligand dependency for gene activation by nuclear receptors. Cell. 2007;128(3):505–518. doi: 10.1016/j.cell.2006.12.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Bovenzi V, Momparler RL. Antineoplastic action of 5-aza-2’-deoxycytidine and histone deacetylase inhibitor and their effect on the expression of retinoic acid receptor beta and estrogen receptor alpha genes in breast carcinoma cells. Cancer Chemother. Pharmacol. 2001;48(1):71–76. doi: 10.1007/s002800100294. [DOI] [PubMed] [Google Scholar]
  • 71.Belinsky SA, Klinge DM, Stidley CA, et al. Inhibition of DNA methylation and histone deacetylation prevents murine lung cancer. Cancer Res. 2003;63(21):7089–7093. [PubMed] [Google Scholar]
  • 72.Fan J, Yin WJ, Lu JS, et al. ER alpha negative breast cancer cells restore response to endocrine therapy by combination treatment with both HDAC inhibitor and DNMT inhibitor. J. Cancer Res. Clin. Oncol. 2008;134(8):883–890. doi: 10.1007/s00432-008-0354-x. [DOI] [PubMed] [Google Scholar]; • Showed that DNMT1 inhibitor 5-aza-2′-deoxycytidine and histone deacetylases inhibitor trichostatin A play important roles in restoring sensitivity of the ER-α-negative breast cancer cells to endocrine therapy in vitro and in vivo.
  • 73.Xiong J, Yu D, Wei N, et al. An estrogen receptor alpha suppressor, microRNA-22, is downregulated in estrogen receptor alpha-positive human breast cancer cell lines and clinical samples. FEBS J. 2010;277(7):1684–1694. doi: 10.1111/j.1742-4658.2010.07594.x. [DOI] [PubMed] [Google Scholar]
  • 74.Huynh KT, Chong KK, Greenberg ES, Hoon DS. Epigenetics of estrogen receptor-negative primary breast cancer. Expert Rev. Mol. Diagn. 2012;12(4):371–382. doi: 10.1586/erm.12.26. [DOI] [PubMed] [Google Scholar]
  • 75.Dowsett M, Cuzick J, Wale C, Howell T, Houghton J, Baum M. Retrospective analysis of time to recurrence in the ATAC trial according to hormone receptor status: an hypothesis-generating study. J. Clin. Oncol. 2005;23(30):7512–7517. doi: 10.1200/JCO.2005.01.4829. [DOI] [PubMed] [Google Scholar]
  • 76.Kim HJ, Cui X, Hilsenbeck SG, Lee AV. Progesterone receptor loss correlates with human epidermal growth factor receptor 2 overexpression in estrogen receptor-positive breast cancer. Clin. Cancer Res. 2006;12(3 Pt 2):s1013–s1018. doi: 10.1158/1078-0432.CCR-05-2128. [DOI] [PubMed] [Google Scholar]
  • 77.Saal LH, Holm K, Maurer M, et al. PIK3CA mutations correlate with hormone receptors, node metastasis, and ERBB2, and are mutually exclusive with PTEN loss in human breast carcinoma. Cancer Res. 2005;65(7):2554–2559. doi: 10.1158/0008-5472-CAN-04-3913. [DOI] [PubMed] [Google Scholar]
  • 78.Widschwendter M, Siegmund KD, Muller HM, et al. Association of breast cancer DNA methylation profiles with hormone receptor status and response to tamoxifen. Cancer Res. 2004;64(11):3807–3813. doi: 10.1158/0008-5472.CAN-03-3852. [DOI] [PubMed] [Google Scholar]
  • 79.Mirza S, Sharma G, Prasad CP, et al. Promoter hypermethylation of TMS1, BRCA1, ERalpha and PRB in serum and tumor DNA of invasive ductal breast carcinoma patients. Life Sci. 2007;81(4):280–287. doi: 10.1016/j.lfs.2007.05.012. [DOI] [PubMed] [Google Scholar]
  • 80.Pathiraja TN, Shetty PB, Jelinek J, et al. Progesterone receptor isoform-specific promoter methylation: association of PRA promoter methylation with worse outcome in breast cancer patients. Clin. Cancer Res. 2011;17(12):4177–4186. doi: 10.1158/1078-0432.CCR-10-2950. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Billam M, Sobolewski MD, Davidson NE. Effects of a novel DNA methyltransferase inhibitor zebularine on human breast cancer cells. Breast Cancer Res. Treat. 2009;120(3):581–592. doi: 10.1007/s10549-009-0420-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Fiegl H, Millinger S, Goebel G, et al. Breast cancer DNA methylation profiles in cancer cells and tumor stroma: association with HER-2/neu status in primary breast cancer. Cancer Res. 2006;66(1):29–33. doi: 10.1158/0008-5472.CAN-05-2508. [DOI] [PubMed] [Google Scholar]
  • 83.Kastner P, Krust A, Turcotte B, et al. Two distinct estrogen-regulated promoters generate transcripts encoding the two functionally different human progesterone receptor forms A and B. EMBO J. 1990;9(5):1603–1614. doi: 10.1002/j.1460-2075.1990.tb08280.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Mote PA, Bartow S, Tran N, Clarke CL. Loss of co-ordinate expression of progesterone receptors A and B is an early event in breast carcinogenesis. Breast Cancer Res. Treat. 2002;72(2):163–172. doi: 10.1023/a:1014820500738. [DOI] [PubMed] [Google Scholar]
  • 85.Hopp TA, Weiss HL, Hilsenbeck SG, et al. Breast cancer patients with progesterone receptor PR-A-rich tumors have poorer disease-free survival rates. Clin. Cancer Res. 2004;10(8):2751–2760. doi: 10.1158/1078-0432.ccr-03-0141. [DOI] [PubMed] [Google Scholar]
  • 86.Richer JK, Jacobsen BM, Manning NG, Abel MG, Wolf DM, Horwitz KB. Differential gene regulation by the two progesterone receptor isoforms in human breast cancer cells. J. Biol. Chem. 2002;277(7):5209–5218. doi: 10.1074/jbc.M110090200. [DOI] [PubMed] [Google Scholar]
  • 87.Pathiraja TN, Stearns V, Oesterreich S. Epigenetic regulation in estrogen receptor positive breast cancer–role in treatment response. J. Mammary Gland Biol. Neoplasia. 2010;15(1):35–47. doi: 10.1007/s10911-010-9166-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Gaudet MM, Campan M, Figueroa JD, et al. DNA hypermethylation of ESR1 and PGR in breast cancer: pathologic and epidemiologic associations. Cancer Epidemiol. Biomarkers Prev. 2009;18(11):3036–3043. doi: 10.1158/1055-9965.EPI-09-0678. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Faivre EJ, Lange CA. Progesterone receptors upregulate Wnt-1 to induce epidermal growth factor receptor transactivation and c-Src-dependent sustained activation of Erk1/2 mitogen-activated protein kinase in breast cancer cells. Mol. Cell Biol. 2007;27(2):466–480. doi: 10.1128/MCB.01539-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Zheng ZY, Zheng SM, Bay BH, Aw SE, V CLL. Anti-estrogenic mechanism of unliganded progesterone receptor isoform B in breast cancer cells. Breast Cancer Res. Treat. 2008;110(1):111–125. doi: 10.1007/s10549-007-9711-8. [DOI] [PubMed] [Google Scholar]
  • 91.Abdel-Hafiz H, Takimoto GS, Tung L, Horwitz KB. The inhibitory function in human progesterone receptor N termini binds SUMO-1 protein to regulate autoinhibition and transrepression. J. Biol. Chem. 2002;277(37):33950–33956. doi: 10.1074/jbc.M204573200. [DOI] [PubMed] [Google Scholar]
  • 92.Wargon V, Helguero LA, Bolado J, et al. Reversal of antiprogestin resistance and progesterone receptor isoform ratio in acquired resistant mammary carcinomas. Breast Cancer Res. Treat. 2009;116(3):449–460. doi: 10.1007/s10549-008-0150-y. [DOI] [PubMed] [Google Scholar]; •• The first in vivo study to show the relationship between progesterone receptor isoform expression and antiprogestin responsiveness.
  • 93.Wargon V, Fernandez SV, Goin M, Giulianelli S, Russo J, Lanari C. Hypermethylation of the progesterone receptor A in constitutive antiprogestin-resistant mouse mammary carcinomas. Breast Cancer Res. Treat. 2011;126(2):319–332. doi: 10.1007/s10549-010-0908-x. [DOI] [PubMed] [Google Scholar]
  • 94.Yin H, Glass J. The phenotypic radiation resistance of CD44+/CD24(-or low) breast cancer cells is mediated through the enhanced activation of ATM signaling. PLoS ONE. 2011;6(9):e24080. doi: 10.1371/journal.pone.0024080. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Maugeri-Sacca M, Vigneri P, De Maria R. Cancer stem cells and chemosensitivity. Clin. Cancer Res. 2011;17(15):4942–4947. doi: 10.1158/1078-0432.CCR-10-2538. [DOI] [PubMed] [Google Scholar]
  • 96.Balic M, Schwarzenbacher D, Stanzer S, et al. Genetic and epigenetic analysis of putative breast cancer stem cell models. BMC Cancer. 2013;13:358. doi: 10.1186/1471-2407-13-358. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.van Vlerken LE, Hurt EM, Hollingsworth RE. The role of epigenetic regulation in stem cell and cancer biology. J. Mol. Med. (Berl) 2012;90(7):791–801. doi: 10.1007/s00109-012-0917-9. [DOI] [PubMed] [Google Scholar]
  • 98.Baba T, Convery PA, Matsumura N, et al. Epigenetic regulation of CD133 and tumorigenicity of CD133 +ovarian cancer cells. Oncogene. 2009;28(2):209–218. doi: 10.1038/onc.2008.374. [DOI] [PubMed] [Google Scholar]
  • 99.Tabu K, Sasai K, Kimura T, et al. Promoter hypomethylation regulates CD133 expression in human gliomas. Cell Res. 2008;18(10):1037–1046. doi: 10.1038/cr.2008.270. [DOI] [PubMed] [Google Scholar]
  • 100.Yi JM, Tsai HC, Glockner SC, et al. Abnormal DNA methylation of CD133 in colorectal and glioblastoma tumors. Cancer Res. 2008;68(19):8094–8103. doi: 10.1158/0008-5472.CAN-07-6208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.You H, Ding W, Rountree CB. Epigenetic regulation of cancer stem cell marker CD133 by transforming growth factor-beta. Hepatology. 2010;51(5):1635–1644. doi: 10.1002/hep.23544. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.van Vlerken LE, Kiefer CM, Morehouse C, et al. EZH2 is required for breast and pancreatic cancer stem cell maintenance and can be used as a functional cancer stem cell reporter. Stem Cells Transl. Med. 2013;2(1):43–52. doi: 10.5966/sctm.2012-0036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Crea F, Hurt EM, Mathews LA, et al. Pharmacologic disruption of Polycomb repressive complex 2 inhibits tumorigenicity and tumor progression in prostate cancer. Mol. Cancer. 2011;10:40. doi: 10.1186/1476-4598-10-40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Rizzo S, Hersey JM, Mellor P, et al. Ovarian cancer stem cell-like side populations are enriched following chemotherapy and overexpress EZH2. Mol. Cancer Ther. 2011;10(2):325–335. doi: 10.1158/1535-7163.MCT-10-0788. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Crea F, Hurt EM, Farrar WL. Clinical significance of Polycomb gene expression in brain tumors. Mol. Cancer. 2010;9:265. doi: 10.1186/1476-4598-9-265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Fouse SD, Shen Y, Pellegrini M, et al. Promoter CpG methylation contributes to ES cell gene regulation in parallel with Oct4/Nanog, PcG complex, and histone H3 K4/K27 trimethylation. Cell Stem Cell. 2008;2(2):160–169. doi: 10.1016/j.stem.2007.12.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Zhou J, Bi C, Cheong LL, et al. The histone methyltransferase inhibitor, DZNep, up-regulates TXNIP, increases ROS production, and targets leukemia cells in AML. Blood. 2011;118(10):2830–2839. doi: 10.1182/blood-2010-07-294827. [DOI] [PubMed] [Google Scholar]
  • 108.Chiba T, Suzuki E, Negishi M, et al. 3-Deazaneplanocin A is a promising therapeutic agent for the eradication of tumor-initiating hepatocellular carcinoma cells. Int. J. Cancer. 2011;130(11):2557–2567. doi: 10.1002/ijc.26264. [DOI] [PubMed] [Google Scholar]
  • 109.Suva ML, Riggi N, Janiszewska M, et al. EZH2 is essential for glioblastoma cancer stem cell maintenance. Cancer Res. 2009;69(24):9211–9218. doi: 10.1158/0008-5472.CAN-09-1622. [DOI] [PubMed] [Google Scholar]
  • 110.Wang J, Lu F, Ren Q, et al. Novel histone demethylase LSD1 inhibitors selectively target cancer cells with pluripotent stem cell properties. Cancer Res. 2011;71(23):7238–7249. doi: 10.1158/0008-5472.CAN-11-0896. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Schwarzenbacher D, Balic M, Pichler M. The role of microRNAs in breast cancer stem cells. Int. J. Mol. Sci. 2013;14(7):14712–14723. doi: 10.3390/ijms140714712. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Park SM, Gaur AB, Lengyel E, Peter ME. The miR-200 family determines the epithelial phenotype of cancer cells by targeting the E-cadherin repressors ZEB1 and ZEB2. Genes Dev. 2008;22(7):894–907. doi: 10.1101/gad.1640608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Heo I, Joo C, Kim YK, et al. TUT4 in concert with Lin28 suppresses microRNA biogenesis through pre-microRNA uridylation. Cell. 2009;138(4):696–708. doi: 10.1016/j.cell.2009.08.002. [DOI] [PubMed] [Google Scholar]
  • 114.Martello G, Rosato A, Ferrari F, et al. A MicroRNA targeting dicer for metastasis control. Cell. 2010;141(7):1195–1207. doi: 10.1016/j.cell.2010.05.017. [DOI] [PubMed] [Google Scholar]
  • 115.Mani SA, Guo W, Liao MJ, et al. The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell. 2008;133(4):704–715. doi: 10.1016/j.cell.2008.03.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Nalls D, Tang SN, Rodova M, Srivastava RK, Shankar S. Targeting epigenetic regulation of miR-34a for treatment of pancreatic cancer by inhibition of pancreatic cancer stem cells. PLoS ONE. 2011;6(8):e24099. doi: 10.1371/journal.pone.0024099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Wu MY, Fu J, Xiao X, Wu J, Wu RC. MiR-34a regulates therapy resistance by targeting HDAC1 and HDAC7 in breast cancer. Cancer Lett. 2014;354(2):311–319. doi: 10.1016/j.canlet.2014.08.031. [DOI] [PubMed] [Google Scholar]; • Study not only identified the miR-34a-HDAC1/HDAC7-HSP70 K246 axis as a novel molecular signature predictive of therapy resistance but also a viable target for potential new anticancer therapies to reduce such resistance in breast cancer.
  • 118.Shankar S, Kumar D, Srivastava RK. Epigenetic modifications by dietary phytochemicals: implications for personalized nutrition. Pharmacol. Ther. 2013;138(1):1–17. doi: 10.1016/j.pharmthera.2012.11.002. [DOI] [PMC free article] [PubMed] [Google Scholar]; •• Discusses the effect of dietary phytochemicals in genetic and epigenetic modifications and how these modifications help to prevent various types of cancers and improve health outcomes.
  • 119.Lee JH, Khor TO, Shu L, Su ZY, Fuentes F, Kong AN. Dietary phytochemicals and cancer prevention: Nrf2 signaling, epigenetics, and cell death mechanisms in blocking cancer initiation and progression. Pharmacol. Ther. 2013;137(2):153–171. doi: 10.1016/j.pharmthera.2012.09.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Siddiqui IA, Adhami VM, Saleem M, Mukhtar H. Beneficial effects of tea and its polyphenols against prostate cancer. Mol. Nutr. Food Res. 2006;50(2):130–143. doi: 10.1002/mnfr.200500113. [DOI] [PubMed] [Google Scholar]
  • 121.Fang MZ, Wang Y, Ai N, et al. Tea polyphenol (-)-epigallocatechin-3-gallate inhibits DNA methyltransferase and reactivates methylation-silenced genes in cancer cell lines. Cancer Res. 2003;63(22):7563–7570. [PubMed] [Google Scholar]
  • 122.Savouret JF, Quesne M. Resveratrol and cancer: a review. Biomed. Pharmacother. 2002;56(2):84–87. doi: 10.1016/s0753-3322(01)00158-5. [DOI] [PubMed] [Google Scholar]
  • 123.Srivastava RK, Unterman TG, Shankar S. FOXO transcription factors and VEGF neutralizing antibody enhance antiangiogenic effects of resveratrol. Mol. Cell Biochem. 2010;337(1–2):201–212. doi: 10.1007/s11010-009-0300-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Wang Y, Lee KW, Chan FL, Chen S, Leung LK. The red wine polyphenol resveratrol displays bilevel inhibition on aromatase in breast cancer cells. Toxicol. Sci. 2006;92(1):71–77. doi: 10.1093/toxsci/kfj190. [DOI] [PubMed] [Google Scholar]
  • 125.Roy SK, Chen Q, Fu J, Shankar S, Srivastava RK. Resveratrol inhibits growth of orthotopic pancreatic tumors through activation of FOXO transcription factors. PLoS ONE. 2011;6(9):e25166. doi: 10.1371/journal.pone.0025166. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Stefanska B, Rudnicka K, Bednarek A, Fabianowska-Majewska K. Hypomethylation and induction of retinoic acid receptor beta 2 by concurrent action of adenosine analogues and natural compounds in breast cancer cells. Eur. J. Pharmacol. 2010;638(1–3):47–53. doi: 10.1016/j.ejphar.2010.04.032. [DOI] [PubMed] [Google Scholar]
  • 127.Teiten MH, Eifes S, Dicato M, Diederich M. Curcumin-the paradigm of a multi-target natural compound with applications in cancer prevention and treatment. Toxins (Basel) 2010;2(1):128–162. doi: 10.3390/toxins2010128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Liu Z, Xie Z, Jones W, et al. Curcumin is a potent DNA hypomethylation agent. Bioorg. Med. Chem. Lett. 2009;19(3):706–709. doi: 10.1016/j.bmcl.2008.12.041. [DOI] [PubMed] [Google Scholar]
  • 129.Medina-Franco JL, Lopez-Vallejo F, Kuck D, Lyko F. Natural products as DNA methyltransferase inhibitors: a computer-aided discovery approach. Mol. Divers. 2011;15(2):293–304. doi: 10.1007/s11030-010-9262-5. [DOI] [PubMed] [Google Scholar]
  • 130.Chen Y, Shu W, Chen W, Wu Q, Liu H, Cui G. Curcumin, both histone deacetylase and p300/CBP-specific inhibitor, represses the activity of nuclear factor kappa B and Notch 1 in Raji cells. Basic. Clin. Pharmacol. Toxicol. 2007;101(6):427–433. doi: 10.1111/j.1742-7843.2007.00142.x. [DOI] [PubMed] [Google Scholar]
  • 131.Yang J, Cao Y, Sun J, Zhang Y. Curcumin reduces the expression of Bcl-2 by upregulating miR-15a and miR-16 in MCF-7 cells. Med. Oncol. 2010;27(4):1114–1118. doi: 10.1007/s12032-009-9344-3. [DOI] [PubMed] [Google Scholar]

Articles from Epigenomics are provided here courtesy of Taylor & Francis

RESOURCES