Sie sind auf Seite 1von 14

Seminars in Cancer Biology 51 (2018) 22–35

Contents lists available at ScienceDirect

Seminars in Cancer Biology


journal homepage: www.elsevier.com/locate/semcancer

Review

Epigenetics of breast cancer: Biology and clinical implication in the era of T


precision medicine

Barbara Pasculli, Raffaela Barbano, Paola Parrella
Laboratory of Oncology, IRCCS “Casa Sollievo della Sofferenza”, 71013, San Giovanni Rotondo, FG, Italy

A R T I C L E I N F O A B S T R A C T

Keywords: In the last years, mortality from breast cancer has declined in western countries as a consequence of a more
Breast cancer widespread screening resulting in earlier detection, as well as an improved molecular classification and advances
DNA methylation in adjuvant treatment. Nevertheless, approximately one third of breast cancer patients will develop distant
Histones modifications metastases and eventually die for the disease. There is now a compelling body of evidence suggesting that
ncRNAs
epigenetic modifications comprising DNA methylation and chromatin remodeling play a pivotal role since the
Biomarkers
early stages of breast cancerogenesis. In addition, recently, increasing emphasis is being placed on the property
of ncRNAs to finely control gene expression at multiple levels by interacting with a wide array of molecules such
that they might be designated as epigenetic modifiers. In this review, we summarize the current knowledge
about the involvement of epigenetic modifications in breast cancer, and provide an overview of the significant
association of epigenetic traits with the breast cancer clinicopathological features, emphasizing the potentiality
of epigenetic marks to become biomarkers in the context of precision medicine.

1. Introduction novel and more effective therapeutic strategies.


A major milestone in this path has been the definition of five bio-
Breast cancer is the most frequent cancer in women and the second logically and clinically relevant intrinsic breast cancer subtypes based
most common cause of cancer related death. It has been estimated that on genome-wide expression and DNA copy number analyses: two oes-
approximately 1.5 million women worldwide are diagnosed with breast trogen receptor (ER) positive subtypes characterized by a relatively low
cancer each year [1]. Oestrogen receptor (ER) and progesterone re- (luminal A) and high (luminal B) expression of proliferation-related
ceptor (PgR) statuses have been used since the 1970s to identify a genes, a subtype enriched for HER2-amplified tumours (HER2-en-
breast cancer subgroup that is responsive to endocrine therapies re- riched), a subtype characterized by the absence of ER, PgR expression
presenting the first target therapy implemented into the clinics [2]. and HER2 amplification (Basal Like) and a subtype with an expression
More recently, the development of monoclonal antibodies targeting the profile similar to that of normal breast tissue (normal-like) [4–7]
HER2neu protein that is amplified in approximately 30% of breast (Table 1). This has led to the development of risk scores based on gene
cancers has significantly improved the outcome in both adjuvant setting expression signatures that are currently entering the clinical practice to
and metastatic disease [3]. Although these improvements together with identify low risk breast cancer patients in the attempt to spare un-
screening programs, and better education, have led to a significant necessary adjuvant treatment [8]. Nevertheless, even these classifica-
decline in breast cancer mortality in the past 15 years, about 25%–40% tion systems do not account for all the reported pathological and clin-
of breast cancer patients develop metastases and eventually die from ical heterogeneity of breast cancer.
the disease [1]. Therefore there is an urgent need to understand the
molecular bases of such a diverse behaviour to identifies the subset of
breast cancer patients at high risk of disease progression and investigate

Abbreviations: BC, breast Cancer; ER, estrogen Receptor; PgR,PR, progesteron receptor; HER-2, human epidermal growth factor receptor 2; TNBC, triple negative breast cancer; ncRNA,
non coding RNA; CpG(s), cytosine-phosphate-guanine dinucleotide(s); 5-me-C, 5-methyl-cytosine; DNMT(s), DNA-methyl transferase(s); DH, ductal hyperplasia; ADH, atypical hyper-
plasia; DCIS, in situ carcinoma; BRCA1, breast cancer 1 gene; PFS, progression-free survival; DSS, disease specific survival; OS, overall survival; DFS, disease-free survival; RFS, relapse-
free survival; DNMTi, DNMT inhibitor; PTM(s), post-translational modification(s); E2, estrogen; lncRNAs, long non coding RNAs; rRNA, ribosomal RNA; tRNA, transfer RNA; snRNA,
small nuclear RNA; snoRNA, small nucleolar RNA; siRNA, small interfering; piRNA, Piwi-associated RNA; miRNA, miR:microRNA; lincRNA, long intergenic non coding cRNA; EMT,
epithelial to mesenchymal transition

Corresponding author.
E-mail addresses: b.pasculli@operapadrepio.it (B. Pasculli), r.barbano@operapadrepio.it (R. Barbano), pparrella@operapadrepio.it (P. Parrella).

https://doi.org/10.1016/j.semcancer.2018.01.007
Received 28 April 2017; Received in revised form 15 December 2017; Accepted 11 January 2018
Available online 12 January 2018
1044-579X/ © 2018 Elsevier Ltd. All rights reserved.

Downloaded for Sitti Ayu Suhartina yahya Yahya (sitti.ayu.yahya@mail.ugm.ac.id) at Gadjah Mada University from ClinicalKey.com by Elsevier on October 23, 2019.
For personal use only. No other uses without permission. Copyright ©2019. Elsevier Inc. All rights reserved.
B. Pasculli et al. Seminars in Cancer Biology 51 (2018) 22–35

Table 1
Breast Cancer Intrinsic Molecular Subtypes and DNA methylation based classifications (Epitypes).

Intrinsic Subtypes Characteristics Surrogate TCGA[103] Stefansson et al Holm et al


classification* [104] [104]

Luminal A High expression of ER and oestrogen related genes. ER positive Group 1 Cluster 3 ET2
Low level of proliferation related genes. PgR positive Group 2 Cluster 1 ET3
Expression of luminal epithelial cytokeratins CK8 and CK18. HER2 negative Group 3 Cluster 2 ET4
Ki67low ET5
Luminal B HER2 neg Low expression of ER and oestrogen-regulated genes ER positive Group 3 Cluster 1
High expression of proliferation-related genes PgR positive/negative
High expression of growth receptor signaling genes HER2 negative ET2
Expression of luminal epithelial cytokeratins CK8 and CK18 Ki67high ET3
Luminal B HER2 pos High expression of proliferation-related genes ER positive ET4
High expression of growth receptor signaling genes PgR negative/positive ET5
Expression of luminal epithelial cytokeratins CK8 and CK18 HER2 positive
Overexpression/amplification of the HER2 oncogene
HER2 enriched HER2 amplification or high HER2 expression ER negative Group 1 Cluster 1 ET6
High expression of HER2 related genes and/or genes located within the PGR negative Group 2 Cluster 2
HER2 amplicon located in the 17q12 chromosome.
TP53 mutations HER2positive Cluster 3
Basal like ER negative ER negative Group 5 Cluster 2 ET7
PgR negative PgR negative
High expression of basal myoepithelial markers, (i.e. CK5, CK 14, CK 17 HER2 negative
and laminin),
High expression of P-cadherin, fascin, caveolins 1 and 2, alpha-beta
crystallin and epidermal growth factor receptor (EGFR).
TP53 mutation
inactivation of the (Rb) pathway.
Genetic instability

2. Epigenetic alterations in Breast cancer preserves the hemimethylated DNA generated during DNA replication
and is required for copying the DNA methylation pattern from the
Epigenetic mechanisms have emerged as fundamental players in template to the daughter DNA strand. Instead, the DNMT3A and B
breast cancer development and progression [9]. During the early stages enzymes are known as de novo methyltransferases and target un-
of carcinogenesis, alterations in chromatin structure through DNA methylated DNA [16]. DNA hypermethylation typically occurs at CpG
chemical modification (methyl CpG, 5-hydroxymethylcytosine, 5hmC) islands, which represent short interspersed DNA sequences that deviate
and post-translational modifications of DNA bound proteins including significantly from the average genomic pattern by being GC-rich, CpG-
histones, as result of genetic lesions or environmental hits, affect cel- rich, and predominantly not methylated. Approximately 70% of anno-
lular plasticity and favour the oncogenic reprogramming of tumour tated gene promoters are associated with CpG islands whose methyla-
progenitor cells promoting the acquisition of uncontrolled self-renewal tion status generally correlates with transcriptional activity [17].
properties. At later stages of cancer growth, additional epigenetic In breast cancer, promoter hypermethylation has been reported in
changes, together with subclonal mutations and signals from the mi- more than 100 genes [18]. Many of those genes play important roles in
croenvironment, modulate cancer cell phenotype and affect the meta- cell-cycle regulation (e.g. CCND2, CDKN2A), DNA repair (e.g. BRCA1,
static propensity of the tumour [10–13]. More recently, beyond the GSTP1) apoptosis (e.g. BCL2, DAPK), tissue invasion and metastasis
classical epigenetic mechanisms, an increasingly recognized role as (e.g. RASSF1A, RARβ, TWIST, HIN1), regulation of cell transcription
epigenetic modifiers is being given to non-coding RNAs (ncRNAs), (e.g. HOXA5), cell adhesion (e.g. CDH1), and hormone-mediated cell
especially to miRNAs and lncRNAs [14]. Indeed, while it has been signaling (ERα, ERβ and THRβ). In addition, genome wide hypo-
shown that, similar to protein-coding genes, ncRNAs are susceptible to methylation also represents a common feature of breast tumours and
epigenetic regulation at transcriptional level, they regulate in turn gene frequently occurs in regions of segmental duplications [19]. Moreover,
expression through acting by both themselves and/or their interactions a number of studies also identified hypomethylated genes in primary
with DNA and/or proteins. This enables ncRNAs to influence several breast tumours including IL10 [20], MDR1 [21], FEN1 [22], NAT1 [23],
genes at the same time hence it is not surprising that the documented CDH3 [24], urokinase [25], synuclein [26], JAGGED1 and NOTCH1
deregulation of ncRNAs in human cancers affects every step (hallmark) [27].
of cancerogenesis and progression till metastatic spread to distant or- A growing number of evidences supports a model where epigenetic
gans. In light of this, epigenome-wide analyses may offer the opportu- reprogramming of breast cancer initiating cells represent a key step in
nity to identify novel, typical traits of cancer for improving cancer breast carcinogenesis and in the subclonal evolution that ultimately
prognostication and individualized treatments. leads to the pathological and clinical heterogeneity of breast tumours
[28] (Fig. 1). Indeed, by using a genome wide promoter microarray,
Helou et al. [13] evaluated the DNA methylation profile of ALDE-
2.1. DNA methylation FLUOR-positive breast cancer stem cells (bCSCs) and non-bCSCs. They
found that bCSCs have a distinct DNA methylation landscape with TGFβ
In eukaryotes, DNA methylation occurs mainly at cytosines con- signaling as a key epigenetic regulator of their differentiation. Inter-
tained within cytosine-phosphate-guanine (CpG) dinucleotides which estingly, the analysis of peripheral blood lymphocytes isolated from
are converted by the addition of a methyl (CH3) group to the 5th carbon breast cancer patients and healthy controls by Severi et al. [29] found
of the pyrimidine ring of cytosine resulting in a formation of 5-methyl- that the epigenome-wide DNA methylation profile of CpGs within
cytosine (5-me-C) [15]. This enzymatic reaction is catalyzed by a family functional promoters was associated with an increased breast cancer
of enzymes named DNA-Methyl Transferases (DNMTs), which catalyze risk, whereas DNA methylation of genomic regions outside promoters
the transfer from the methyl group of the S-adenosyl methionine donor. was associated with decreased risk, thus suggesting that constitutive
There are five known types of DNMT enzymes, but only DNMT1

23

Downloaded for Sitti Ayu Suhartina yahya Yahya (sitti.ayu.yahya@mail.ugm.ac.id) at Gadjah Mada University from ClinicalKey.com by Elsevier on October 23, 2019.
For personal use only. No other uses without permission. Copyright ©2019. Elsevier Inc. All rights reserved.
B. Pasculli et al. Seminars in Cancer Biology 51 (2018) 22–35

Fig. 1. CpG DNA methylation changes contributing


to breast cancerogenesis. The most common type of
breast tumour is the invasive ductal carcinoma (IDC,
or No Special Type, NST, according to WHO 2017).
During progression from no-obliged pre-invasive le-
sions (Atypical Ductal Hyperplasia, ADH), and/or
precursor Ductal Carcinoma in situ (DCIS) to in-
vasive and metastatic disease, the epigenetic repro-
gramming of cancer cells leads to an increase in CpG
island promoter methylation, associated with de-
methylation of gene bodies and repetitive sequences.

epigenetic traits play a role in breast cancer development. homologous protein H5 are involved in higher-order structures of
Early studies that used a candidate gene approach already found chromatin. The other four types of histones associate with DNA to form
precise patterns of methylation during the transition from ductal hy- nucleosomes [38]. A major component of epigenetic regulations is re-
perplasia (DH), atypical hyperplasia (ADH) to in situ carcinoma (DCIS) presented by the post-translational modifications (PTMs) of histone
and invasive carcinoma [30–33]. A study assessing methylation levels tails. These modifications affect chromatin structure by destabilizing
of nine genes on synchronous preinvasive lesions, (ADH and/or DCIS) histone interactions with DNA, altering nucleosome contacts, and
invasive ductal breast carcinomas (IDC) and normal breasts demon- forming binding sites for transcriptional regulators[39]. Overall, 60
strated differential methylation patterns for APC, CDH1, and CTNNB1 distinct modification sites have so far been identified within histones
gene promoter regions in pathological samples as compared with [40]. Among the common PTMs, acetylation of lysine 9 of histone H3
normal tissues [34]. Similarly, the analysis of RARβ2, RASSF1A, (H3K9Ac) has been classically associated with open and accessible
MINT17, and MINT13 throughout the key steps of breast cancer de- chromatin regions [41,42]. By contrast, histone methylation can be
velopment, i.e. transitions from DH to ADH to DCIS, identified the hy- associated with either open or compacted chromatin regions, de-
permethylation of RARβ2 and RASSF1A promoters as an early epige- pending on the specific histone aminoacid that is methylated. For ex-
netic event in BC, representing a putative predictor of malignant ample, mono- or trimethylation of lysine 4 on histone H3 (H3K4me1 or
potential [35]. By using a quantitative PCR method for the assessment H3K4me3) and trimethylation of histone H3 on lysine 36 (H3K36me3)
of methylated alleles (AQAMA), van Hoesel et al. [36] made a com- are associated with open chromatin [41–43]. Whereas, trimethylation
parative analysis of long interspersed nucleotide element-1 (LINE1) of lysines 9 and 27 on histone H3 (H3K9me3 and H3K27me3, respec-
methylation levels in breast-cancer patients, with available normal tively) is associated with compacted chromatin regions resulting in
breast tissue, ductal hyperplasia, atypical ductal hyperplasia, ductal repression of target genes [44].
carcinoma in situ (DCIS), and invasive carcinoma. They found that In recent years, the evaluation of genome-wide pattern of histone
LINE1 was hypermethylated in the benign lesions but became hypo- modifications has made increasingly clear that different combinations
methylated in the DCIS and invasive tumours [36]. of histone marks can provide more detailed information about cell
More recently, Teschendorff et al. have shed new light into the early identity and disease state [28,45]. For example, the presence of both
epigenetic events that govern breast cancer development [37]. By using the open chromatin mark H3K4me3 and the compacted chromatin
the Illumina 450k bead chip array, they analysed 42 matched normal- mark H3K9me3 at a promoter can identify imprinted genes [46]. Thus,
adjacent and cancer tissues and 50 normal tissues from healthy women, the variety and complexity of histone PTMs have led to the “histone
demonstrating the presence of tens to thousands epigenetic alterations code hypothesis” which proposes that histone PTMs represent an es-
in normal tissue adjacent to cancer, supporting the existence of an sential mechanism regulating chromatin-template processes, ultimately
epigenetic field defect [37]. These field defects were also present in a influencing physiological and pathological cellular responses [47].
cohort of 15 normal adjacent and 40 DCISs, with a substantial DNA Changes in histone marks have been associated with malignant
methylation increase in DCISs compared with normal adjacent samples. transformation and metastatic behaviour in in vitro studies. For in-
The distribution of the recognized methylation patterns was strongly stance, by combining biochemical and epigenomic approaches in a cell-
enriched in binding sites for transcription factors associated with based model mimicking the transformation from human primary cells
maintaining chromatin structure, including two Polycomb Repressive to tumour cells, Zhao et al. found a decrease in histones H3K9me2 and
Complex 2 (PRC2) members (EZH2 and SUZ12) as well as RBBP5, CTCF H3K9me3 during transformation. In addition, they identified KDM3A/
and RAD21. System-level gene promoter analysis also confirmed that JMJD1A, an H3K9me2 demethylase as responsible for H3K9me2 de-
epigenetic field defects target specific signaling pathways, notably WNT crease [48]. In another study, Messier et al. [49] used a genome-wide
signaling, a stem-cell differentiation pathway, and FGF signaling [37]. ChIP-Seq approach to evaluate the relative roles of histone H3 methy-
lation (H3K4me3) and acetylation (H3K4ac) in three breast cancer cell
lines mimicking the normal (MCF-10A), tumorigenic (MCF-7) and pro-
2.2. Histone modifications metastatic (MDA-MB-231) phenotypes. In both breast cancer cell lines,
there was an increase in H3K4ac marks as compared to the normal-like
Histones are nuclear proteins closely associated with DNA mole- MCF-10A, suggesting that this alteration is required for malignant
cules responsible for the structure of chromatin, and play important transformation. By contrast, the global increase in H3K4me3 at pro-
roles in the regulation of gene expression. Five types of histones have moters was primarily observed in the triple negative MDA-MB-231 cell
been identified: H1 (or H5), H2A, H2B, H3 and H4. H1 and its

24

Downloaded for Sitti Ayu Suhartina yahya Yahya (sitti.ayu.yahya@mail.ugm.ac.id) at Gadjah Mada University from ClinicalKey.com by Elsevier on October 23, 2019.
For personal use only. No other uses without permission. Copyright ©2019. Elsevier Inc. All rights reserved.
B. Pasculli et al. Seminars in Cancer Biology 51 (2018) 22–35

line, suggesting that it may be linked to an increased metastatic po- charged DNA. This increases accessibility for the transcriptional ma-
tential. For all the cell lines, gene promoters are differentially marked, chinery, resulting in transcriptional activation and can be reversed by
with the normal-like MCF-10A having fewer marked promoters com- deacetylation by HDACs [55]. Thus, the epigenetic changes caused by
pared to the cancer cell lines. In addition, MCF-7 showed an increase in imbalances between HATs and HDACs heavily affect global transcrip-
the number of promoters marked by acetylation whereas MDA-MB-231 tional profiles and contribute to tumour development and progression.
cells displayed an increase in H3K4 tri-methylation. In MCF-7 cells, The eighteen human HDAC enzymes are classified into four groups
H3K4ac was found to play a role in the regulation of ESR response based on their homology with yeast HDACs [56,57]. Class I HDACs
pathway, whereas H3K4ac, together with H3K4me3, was associated comprise HDACs 1, 2, 3 and 8 they are localized in the nucleus and are
with the EMT response pathway in the MDA-MB-231 cell line [49]. the most abundant and ubiquitously-expressed HDACs. Class II HDACs
Altogether, these data indicate that deregulation of histone PTMs play a can be further subdivided in class IIa (HDACs 4, 5, 7, and 9) and Class
pivotal role in the development and progression of breast cancer and IIb (HDACs 6 and 8), based on their sequence homology and domain
highlight their possible exploitation as prognostic and predictive bio- organization. These HDACs can shuttle between the nucleus and the
markers as well as therapeutic targets. cytoplasm and their expression is tissue-specific [55]. Class III HDACs
Recently, the genome-wide binding patterns of activating histone are structurally homologous with the yeast Sir2 protein but their role in
H3 lysine 4 trimethylation (H3K4me3) and repressive histone H3 lysine tumorigenesis is still debated because some of them have dual roles as
27 trimethylation (H3K27me3) was compared by ChIP-seq analysis in oncoproteins and tumour-suppressors [103,104]. Last, HDAC11 is the
Human Mammary Epithelial Cells (HMECs), and three breast cancer sole member of class IV [58].
cell lines representing the luminal (ZR-75-1), HER2 amplified (SK-BR-3) Only few studies have evaluated the expression of writers and
and Triple negative/basal (MDA-MB-436) phenotypes. Although the readers in breast cancer. The histone acetyltransferase (HAT) hMOF
genomic distributions and the overall detected peaks of H3K4me3 and (human males absent on the first), which acetylates H4K20, was found
H3K27me3 were comparable, thousands of locus-specific binding to be significantly reduced [58]. Conversely, the Eraser LSD1 (Lysine-
events specific for each of the breast cancer phenotypes were char- specific histone demethylase 1) was highly expressed in oestrogen re-
acterized. Some of these peaks were shared among all cell types sug- ceptor negative breast cancer and was associated with a more ag-
gesting a role in normal cell development. Other peaks were shared by gressive behaviour. Finally, reduced expression of LSD1 resulted in
the three cancer cell lines but exclusive to HMEC, suggesting that they growth inhibition in vitro [59]. The ectopic expression of H4K20me3-
are important in cancer initiation and development. Finally, peaks specific histone methyltransferases SUV420H1 and SUV420H2 in breast
unique to different cancer cell lines suggested a specific role in cell fate cancer cell lines was able to suppress cell invasiveness. On the contrary,
commitment during breast cancer initiation and development. In par- knockdown of SUV420H2 in the immortalized but not tumorigenic
ticular, the number of H3K4me3 peaks between normal and each of the MCF-10A cell line was able to activate invasion in vitro [60].
cancer cell lines were comparable while H3K27me3 peaks varied re- More data are instead available about the role of HDACs in breast
markably among cell types. By analysing the unique genes marked by tumorigenesis. Indeed, a significant correlation between HDAC1 higher
H3K27me3 in each cell line, distinct biological pathways were found to expression and positive hormone receptor status [61,62] has been
be repressed. The expression patterns of the genes regulated by these found. In addition, Muller et al. [63] found that HDAC2 and HDAC3
histone marks in each subtype were validated in three independent expression was significantly higher in poorly differentiated and hor-
gene expression datasets (GSE60785, GSE45827, and GSE69986) and in mone receptor negative tumours. For HDAC2, a significant correlation
a small cohort of fresh frozen breast cancer primary tumours finding a was also found with HER2 overexpression [63].
correlation between histone mark-based gene classifiers and patients
outcome [50]. 2.3. Non coding RNAs
While the role of core histones has been widely investigated in
breast cancer, very little is known about the putative role of linker Based on their size, non coding RNAs are conventionally grouped
histone H1 in breast cancerogenesis. In a study evaluating the expres- into two major categories: small ncRNAs (< 200 nt) and long ncRNAs
sion of 6 out of the 7 H1 somatic variants in BC cell lines, the H1.2 (lncRNAs, > 200 bp, up to 100 kb). Alternatively, ncRNAs can be
variant was found less abundant than other variants at the transcription classified according to their function, namely housekeeping molecules
start sites of inactive genes, and promoters enriched in H1.2 tended to such as ribosomal (rRNA), transfer (tRNA), small nuclear (snRNA) and
be repressed [51]. In another study [52], the H1 phosphorylation small nucleolar (snoRNA) RNAs, that are usually constitutively ex-
profiles was evaluated in two breast cancer cell lines (MCF-7 and MDA- pressed, and regulatory molecules such as small interfering (siRNAs),
MB-231) and in the non-neoplastic MCF-10A cell line. H1 phosphor- Piwi-associated (piRNAs) RNAs, microRNAs (miRNA, miRs) [64,65]
ylation showed a different pattern in the different cell lines. In parti- and long non coding RNAs (lncRNAs) [66,67].
cular, in metastatic MDA-MB-231 cell line, the phosphorylation at Considering ncRNAs in the framework of epigenetic regulation of
threonine 146 was found on both histones H1.2 and H1.4 [51]. Histone gene expression comes from the fact that molecular intermediaries of
H1 phosphorylation could increase and decrease in response to estra- the epigenetic modifications such as DNMTs, HDACs, and Polycomb
diol in MCF-7 and to a PI3 K inhibitor in MDA-MB-231 cells. When proteins have been demonstrated experimentally to function as targets
primary breast tissues were stained for the histone H1 phosphorylation of miRNAs or co-factors of lncRNAs [14], thus ncRNAs may be referred
at threonine 146, variable staining patterns were observed with pT146 to as epigenetic modifiers. On the other hand, since the mechanisms by
labelling correlating with tumour grade [52]. Overall, these data sug- which both molecules exert gene expression regulation do not affect
gest critical roles for specific PTM signatures in directing the organi- DNA sequence, the ncRNAs control of gene expression can be con-
zation of distinct breast cancer epigenomes and regulating oncogenic sidered an epigenetic phenomenon itself, whose aberrations have se-
signaling pathways resulting in the widespread heterogeneity of breast vere downstream effects and virtually affect every step of cancerogen-
tumours. esis.
Three categories of histone modification enzymes are currently re- In the cancer setting, both miRNAs and lncRNAs behave as onco-
cognized. “Writers” such as histone acetyltransferases (HAT) which add genes or tumour suppressor genes as they can be found upregulated or
modifications, “Erasers” such as histone deacetlylases (HDAC) which downregulated. In this regard, while a global downregulation has been
remove PTMs and “Readers” such as 53BP1 which recognize chromatin observed for miRNAs in human tumours compared to healthy tissues
bound PTM marks through specific PTM-specific binding domains [68], the majority of lncRNAs showed to be upregulated owing to their
[53,54]. The acetylation of histones by HAT changes their charge from low expression under physiological conditions [69]. Noteworthy, it has
positive to negative, which reduces their interaction with negatively- been documented that dysregulation of ncRNAs in breast cancer can

25

Downloaded for Sitti Ayu Suhartina yahya Yahya (sitti.ayu.yahya@mail.ugm.ac.id) at Gadjah Mada University from ClinicalKey.com by Elsevier on October 23, 2019.
For personal use only. No other uses without permission. Copyright ©2019. Elsevier Inc. All rights reserved.
B. Pasculli et al. Seminars in Cancer Biology 51 (2018) 22–35

derive from aberrant epigenetic regulation of their gene promoters. are able to fold into more complex secondary or tertiary structures to
Indeed, the analysis of published sequencing-based methylome data by serve their function and to interact with different types of molecules,
Li Y et al. [70] found that aberrant methylation at the promoters of i.e. DNA, RNA and proteins, thereby participating to multiple reg-
ncRNAs was more frequent than that at the promoters of protein-coding ulatory networks represented by chromatin remodeling, alternative
genes in the breast cancer setting, with the intergenic ncRNAs being the splicing, transcriptional and post-transcriptional gene regulation, mo-
majority of the aberrantly methylated ncRNAs. Moreover, they dis- lecular decoys for miRNAs [77].
tinguished five patterns of aberrant ncRNA promoter methylation in the Like miRNAs, lncRNAs have been found to be differentially ex-
context of genomic CpG islands (CpGs), in which aberrant methylation pressed between breast cancers and normal mammary tissues sup-
occurred not only on CpGs, but also in regions flanking CpG and in CpG- porting their role in cancerogenesis [78] HOTAIR (HOX antisense in-
lacking promoters. Integration with transcriptional datasets confirmed tergenic RNA) was the first lncRNA observed deregulated in breast
the association between ncRNA promoter methylation events and cor- cancer where it showed higher levels in primary tumours compared to
respondent transcriptional changes which were in turn functionally adjacent tissue [79]. Noteworthy, this lncRNA has been recognized as
responsible for deregulation of key intracellular signaling pathway such involved in the transcriptional regulation of HOXD10, the well-known
as the MAPK signaling pathway. target of the metastasis-promoting miR-10b, and has also been shown to
miRNAs are short (18–25 nucleotides), highly conserved non-coding promote breast cancer metastasis [80]. Indeed, HOTAIR is involved in
RNAs, that induce translation inhibition or degradation of protein reprogramming the chromatin state, causing increased Polycomb re-
coding mRNAs via imperfect base-pairing to complementary sites on pressive complex-2 occupancy on promoters of genes that inhibit breast
target mRNAs (MREs, miRNA Response Elements). Breast cancer is cancer progression, including HOXD10 [79,81]. Similarly, metastasis-
among the first tumours for which miRNA deregulation was docu- associated lung adenocarcinoma transcript 1 (MALAT1) upregulation
mented through a comparison of 76 cancerous and 10 normal mam- was found in primary BC and its levels were further increased in the
mary tissues by microarray analysis [71]. In their work, Iorio et al. correspondent metastases [58]. The findings from this study suggest
identified a signature of 29 differentially expressed miRNAs able to MALAT1 being a co-transcriptional splicing scaffold that cooperates to
predict the nature of the samples (i.e., tumour or normal breast tissue) the transcription and splicing of pro-tumorigenic genes such as in-
[71]. Among them, some (e.g. miR-21 and miR-155) were upregulated tegrins, ECM proteins, and genes involved in migration and metastasis
whereas others (e.g. miR-10b and miR-145) were downregulated in BC, throughout the tumour progression. Interestingly, alternative spliced
suggesting that each miRNA plays a differential role in the pathology of variants of MALAT1 could be differentially expressed with regard to
breast cancer (few examples are reported in Table 2). Soon after, other full-length transcript and showed independent prognostic performance
miRNA signatures such as the 46-miRNA signature found by De Ri- for MFS [82], underlying the complexity of the mechanisms by which
naldis et al. [72], including 13 highly expressed miRNAs in basal-like, lncRNAs regulate changes in the tumour phenotype.
23 highly expressed in luminal tumours (A or B), 6 in normal-like and 4
in HER2 tumours, and others demonstrated the power of miRNAs to
even stratify BC subtypes [73,74]. 3. Epigenetics traits in the era of precision medicine
LncRNAs represent the largest family of non-coding transcripts, in-
cluding long intergenic ncRNAs (lincRNAs), enhancer ncRNAs (eRNAs), According to the National Institutes of Health (NIH), precision
natural antisense transcripts (NATs), and others [66,67], and the me- medicine is &ldquo;an emerging approach for disease treatment and
chanisms by which they may operate in tumours are emerging as dis- prevention that takes into account individual variability in genes, en-
parate and finely orchestrated [75]. Like protein-coding mRNAs, they vironment, and lifestyle for each person&rdquo;. This new concept of
are frequently transcribed by RNA polymerase II and undergo co- medicine contrasts with the previously &ldquo;one-size-fits-all&rdquo;
transcriptional and post-transcriptional processing events [76], al- approach, in which disease treatment and prevention strategies are
though they appear not to possess coding potentials. Importantly, targeted to the average person, without considering individual differ-
compared to miRNAs whose molecular targeting is based on their pri- ences [83]. As we discussed above, there is now a compelling body of
mary sequence specificity for target sites within target mRNAs, lncRNAs evidence sustaining the pivotal role of epigenetic mechanisms in the
development and progression of breast cancer. The point is how

Table 2
Deregulated miRNAs and associated cancer hallmarks in breast cancers.

microRNA Expression level Main Targets Downstream effect Cancer Hallmark Refs
in BC

miR-155 Increased SOCS1 Induction of STAT3 mediated inflammatory signalling Tumor-promoting inflammation [183–186]
SOCS1 Induction of HK2 Deregulating cellular energetics
C/EBPβ
RHOA E-cadherin expression inhibition Activating Invasion and metastasis
miR-26b, miR- Decreased CDK8 Evading cell cycle arrest at the G0/G1 phase Sustaining proliferative signaling [187, 188]
107
miR-21 Increased Bcl2 Blockade of apoptosis and activation of cyclin-dependent Evading growth suppressors, Resisting [189–191]
PDCD4 kinase inhibitors (e.g. p21), and deregulation of anchorage- cell death, activating invasion and
TIMP3 independent growth. metastasis
TPM1
SERPINB5
miR-10b Increased HOXD10 Activation of RhoC, urokinase plasminogen activator Activating Invasion and metastasis [192, 193]
KLF4 receptor, α3-integrin, MT1-MMP
Neurofibromin
miR-200 family Decreased ZEB1/ZEB2 Loss of epithelial marker (e.g. E-cadherin) Activating Invasion and metastasis [194–197]
FOG2, Fibronectin,
Moesin, BMI-1
miR-205 Decreased ZEB1/ZEB2 Activation of HER2 mitogenic pathway Activating Invasion and metastasis [198]
HER3
VEGF-A

26

Downloaded for Sitti Ayu Suhartina yahya Yahya (sitti.ayu.yahya@mail.ugm.ac.id) at Gadjah Mada University from ClinicalKey.com by Elsevier on October 23, 2019.
For personal use only. No other uses without permission. Copyright ©2019. Elsevier Inc. All rights reserved.
B. Pasculli et al. Seminars in Cancer Biology 51 (2018) 22–35

epigenetic traits can be exploited to improve management of breast evaluated 1452 CpG sites (corresponding to 803 genes) in 189 paired
cancer patients. Indeed, due to their fundamental biological role, epi- primary breast tumours and normal samples. This study found that
genetic traits represent suitable biomarkers for early detection, prog- basal-like, luminal A and luminal B breast cancer subtypes harbor
nosis or therapy response predictors. specific methylation profiles [102]. In particular, in the basal like/
Triple-negative BC subgroup, the methylation pattern was defined by
3.1. Diagnostic biomarkers the methylation of five genes (CDKN2B, CD44, MGMT, RB and P73) and
hypomethylation of 11 genes (GSTP1, PMS2, MSH2, MLH1, MSH3,
The detection of breast cancer in the early stages of development is MSH6, DLC1, CACNA1A, CACNA1G, TWIST1 and ID4) [102].
the key for a successful treatment of the disease. Methylated DNA can Subsequently, a number of studies have combined DNA methylation
be detected at a very high degree of specificity even in the presence of profile with expression analyses to identify molecular signatures that
an excess of unmethylated DNA. Aberrant promoter methylation has may better account for the pathological and clinical heterogeneity of
been detected with a high degree of concordance in Fine Needle breast cancer.
Aspirate (FNA) and primary tumours, but not always showed a better Initially, the comprehensive molecular analysis of breast cancer
sensitivity and specificity as compared with cytological examination performed by the Tumour Cancer Genome Atlas Network (TCGA)
[84,85]. Atypical and malignant cancer cells can be also found in ductal identified 5 epitypes. Of those, Group 5 showed the lowest level of DNA
lavages or spontaneous produced ductal fluid (Nipple Aspirate). Since methylation and overlapped with the basal-like subtype, whereas
cytomorphological analysis of these specimens is often unsatisfactory, Group 3 showed the highest levels of DNA methylation and was sig-
previous studies evaluated the possibility to detect tumour in ductal nificantly enriched for Luminal B subtype [103]. Stefansson et al.
lavages by analysing promoter methylation of several genes. However, identified three DNA methylation-based subtypes. In particular, cluster
promoter methylation was also detected in healthy individuals and was 1 was enriched in luminal B tumours (Epi-LumB) and showed CpG is-
unable to distinguish between pre-invasive and invasive lesions land promoter hypermethylation; while cluster 3 was enriched in basal
[86,87]. tumours (Epi-Basal) and characterized by gene body hypermethylation.
It is now well established that serum and plasma carry significant By contrast, HER2 and Luminal A breast tumours were more hetero-
amount of cell-free DNA that can be analysed to detect genetic ab- geneous, and only a small subset of the Luminal A showed evidence of a
normalities present in the tumour. Indeed, cell-free DNA from plasma distinctive pattern of DNA methylation changes (Cluster 3) [104]. More
was used in DNA methylation analysis of APC, GSTP1, RAR-β, and recently, another study from Holm et al. [105] identified 7 breast
RASSF1A for early cancer detection [88]. This study showed that the cancer subgroups. Epitypes (ET7) was distinctly associated with basal-
methylation of the selected genes increased with tumour progression like tumours and presence of BRCA1 mutations, two epitypes were
and that additional breast cancer–specific genes could ameliorate the enriched for HER2-amplified tumours (ET6), and one epitype displayed
sensitivity and specificity of this approach. To date, alongside circu- a methylation profile similar to normal epithelial cells (ET1). Luminal
lating DNA, the documented stability of ncRNAs, especially miRNAs, in tumours were stratified into the remaining four epitypes (ET2, ET3,
bodily fluids [89–95] has unveiled the potentials of circulating ncRNAs ET4, ET5). Two dominant patterns of aberrant methylation were
to help diagnose breast cancer. For instance, the first report found that identified. The first, constitutively methylated in both basal-like and
miR-195 and let-7a expression were significantly elevated in BC pa- luminal breast cancer was associated to genes with promoters in a
tients compared to healthy controls [96], and these levels diminished Polycomb-repressed state in normal epithelial cells, and the second
after the surgical resection of the tumour mass. High levels of circu- pattern correlated with gene expression levels and was associated with
lating miR-21 and miR-10b were found to be associated with ER ne- methylation in luminal tumours and genes with active promoters in
gativity, whereas high miR-155 was associated with PR positivity [97]. normal epithelial cells [105]. Overall, these studies agree in identifying
Upregulation of miR-155 in BC patients sera was validated by addi- specific methylation clusters (epitypes) for basal like and luminal B
tional studies [93,98] and, more interestingly, while miR-155 was breast tumours, whereas HER2 enriched and luminal A breast cancers
likely to distinguish BC patients with both primary and metastatic do not show specific methylation patterns (Table 1).
disease from controls, miR-10b and miR-34a were significantly differ- A few studies have analysed the methylation profile of specific
entially expressed only in patients with metastatic disease [93,98]. Of breast cancer subtypes. In particular, the methylome analysis in a large
note, cell-free miR-181a-5p represents one of the most reliable candi- cohort of TNBCs was able to stratify the population into high, medium
date biomarker as its decreased levels of expression in the serum of BC and low risk groups and to identify 17 differentially methylated regions
patients compared to healthy controls were confirmed in more than one significantly associated with patient&rsquo;s survival in both univari-
study [99]. Circulating miRNA signatures were also proposed as diag- able and multivariable analyses [106]. In luminal breast cancers, by
nostic markers in large cohorts of BC patients [100]. Additional studies applying a powerful integrative network algorithm to match DNA me-
demonstrated that also lncRNAs can be detected in bodily fluids. For thylation and RNA-Seq data from the TCGA, Gao et al. identified two
instance, the serum expression levels of circulating lncRNA RP11- methylation clusters which correlated with the luminal A and luminal B
445H22.4 were found significantly increased in BC patients, identifying subtypes. Indeed, luminal A and luminal B tumours shared the same
BC cases with 92% sensitivity and 74% specificity [94]. Noteworthy, epigenetically deregulated traits, but luminal B cancers were char-
lncRNA prostate cancer gene 3 (PCA3) has been found to be strongly acterized by a higher level of methylation than normal tissues [107].
associated to prostate cancer, and it is the first lncRNA that has entered The putative use of histone PMTs as classifiers has been evaluated
the routine clinical practice to identify prostate cancer risk in urine by Elsheikh et al. [108] using immunohistochemistry (IHC). Histones 3
samples (Progensa PCA3 urine test), thereby avoiding invasive prostate and 4 PTMs were evaluated on a large series of breast cancer cases
biopsies[101]. This provides evidences of ncRNAs potentiality to be- (n = 880). In particular, histone lysine acetylation (H3K9ac, H3K18ac,
come of real utility and use in the patient&rsquo;s management and H4K12ac, and H4K16ac), lysine methylation (H3K4me2 and
care. H4K20me3), and arginine methylation (H4R3me2) marks were eval-
uated. High relative levels of global histone acetylation and methyla-
3.2. Prognostic biomarkers tion were associated almost exclusively with the luminal-like breast
cancer subset. By contrast, moderate to low levels of lysine acetylation
DNA methylation profiling and core histone modifications have (H3K9ac, H3K18ac, and H4K12ac), lysine (H3K4me2 and H4K20me3),
been evaluated in breast cancer tissues with the aim to better refine the and arginine methylation (H4R3me2) were associated with Triple ne-
molecular classification and improve the prognostic and predictive gative/basal-like and HER2 amplified breast cancer subgroups. Indeed,
abilities in the clinical setting. In a pioneering study, Holm et al. loss of histone H4 lysine 20 trimethylation (H4K20me3) was associated

27

Downloaded for Sitti Ayu Suhartina yahya Yahya (sitti.ayu.yahya@mail.ugm.ac.id) at Gadjah Mada University from ClinicalKey.com by Elsevier on October 23, 2019.
For personal use only. No other uses without permission. Copyright ©2019. Elsevier Inc. All rights reserved.
B. Pasculli et al. Seminars in Cancer Biology 51 (2018) 22–35

with luminal subtypes, and was independently associated with worse downregulated [119]. Furthermore, HOTAIR was the most up-regu-
disease free survival in multivariable analysis [108]. lated lncRNA of the HER2+ signature.
In the context of prognostic evaluation, also a discrete number of
miRNAs have been associated with prognosis in breast cancer and a few
have been validated in large cohorts of patients [109]. For instance, 3.3. Predictive biomarkers
along with its diagnostic potential, high miR-21 levels likely associated
with decreased overall survival (OS) and disease-free survival (DFS) in In Early Breast Cancer, breast conserving surgery or mastectomy
early-stage breast cancer patients (stage I/II), with miR-21 expression followed by radiotherapy represent the standard of care [121–123].
in this cohort correlating strongly with histological grade[110]. Simi- Decision to treat these patients with adjuvant systemic therapy is
larly, miR-10b was found significantly correlated to DFS, distant me- guided by clinical and pathological features of the tumour. Without
tastases and worse OS independently from the prognostic factors used adjuvant therapy, 12–58% of women will experience disease relapse
routinely to stratify patients according to their risk to progress [111]. within 5 years [124–126], thus the majority of breast cancer patients
However, as expected, the statistical power of miRNA panels over a (approximately 75–92%) will receive adjuvant therapy [126,127]. En-
miRNA alone has been demonstrated by the integrated miRNA/gene docrine therapy is considered for all ER positive breast cancer patients,
signature in a group of 466 patients with breast cancer from The Cancer as well as anti-HER2neu treatments is the standard of care in HER2
Genome Atlas (TCGA) [112]. The final signature, including 30 mRNAs amplified tumours. In locally advanced breast cancer, systemic treat-
and 7 miRNAs (miR-93, miR-103, miR-148b, miR-328, miR-484, miR- ment including hormone therapy and/or anti-HER2 and chemotherapy
874, and miR-1307), proved to significantly predict OS in a multi- are often used prior to surgery to reduce tumour burden. The most
variable model independent of other clinical pathological character- frequently used chemotherapy regimens contain anthracyclines and/or
istics, and showed the highest prognostic value of distant relapse-free taxanes [122,123,128]. Large-scale methylation analyses of breast-
survival in patients with early stage I and II tumours. Furthermore, the cancer tumours have started to investigate the role of global methyla-
validation of this signature on 8 independent breast cancer cohorts, tion differences in association with breast-cancer subtypes and clinical
comprising a total of 2399 patients, demonstrated its superior perfor- features. These advances in understanding the key molecular mechan-
mance for risk stratification with respect to other RNA predictors, in- isms underlying cancer behaviour holds great potential in identifying
cluding those comprised in the MammaPrint and Oncotype DX panels. individuals who might benefit from improved modalities and treatment
Single miRNAs and other miRNA panels associated with prognosis are strategies for breast cancer in line with the ambition of precision
summarized in Table 3. medicine.
Among lncRNAs, HOTAIR expression was shown to independently The standard prognostic and predictive biomarkers that are cur-
predict the risk of metastasis in 164 ER positive BC patients [113]. rently used in clinical practice are based on the recommendations of
Whereas, alternative spliced variants of MALAT1 were found differen- multidisciplinary consensus panels and include the evaluation of ER,
tially expressed with regard to full-length transcript and showed in- PgR, HER2, and Ki67 immunoreactivity, together with clin-
dependent prognostic performance for MFS [82]. Other lncRNAs with icopathological variables including tumour size, lymph node status,
prognostic potentials include both individual transcripts, such as SPRY4 grade, [8,121–123,128]. Martens et al. [129] profiled 200 receptor
intronic transcript 1 (SPRY4-IT1) [114], LINC00472 [115], and positive BCs by using the MethyLight assay and identified 10 genes for
LINC00978 [116], and lncRNAs signatures [78,117]. In particular, Van which DNA methylation correlated with resistance to endocrine
Grembergen et al. [118] have performed a genome-wide transcriptional therapies [129]. In another study, methylation of the Paired-like
survey to explore the lncRNA landscape across 835 breast tissue sam- homeodomain transcription factor 2 gene (PITX2) has been associated
ples and 172 normal breast tissues and identified 215 aberrantly ex- with poor prognosis in ER positive, lymph node negative patients
pressed lncRNAs in breast tumours as compared to normal samples. In treated only with hormone therapy [130,131]. Furthermore, methyla-
particular, lncRNA profiling was able to distinguish ER+ from ER− tion of the PITX2 gene promoter was also associated with poor patient
tumours and allowed stratification into different molecular subtypes, outcome in lymph node positive, ER positive, HER-2/neu-negative
thus supporting the potential of being prognostic. For instance, the breast cancer patients treated with anthracycline-based adjuvant che-
lncRNA H19, the first identified imprinted long non coding transcript motherapy[132]. The PITX2 gene encodes a member of the RIEG
[119], involved in EMT and regulating different miRNAs [120], was up- (Rieger Syndrome)/PITX homeobox family, and acts as a transcription
regulated in luminal A breast cancers. In the same report, an overall factor. Although its role in breast cancer is not clear, PITX2 is involved
downregulation of lncRNAs compared to the other subtypes was ob- in basal and hormone-regulated activity of prolactin thus its reduced
served In the basal-like subtype, with LINC00993 being the most expression may influence prolactin levels within breast cancer tissues
and affect cellular sensitivity to anti-estrogen therapies [132,133].

Table 3
Examples of miRNAs and miRNA panels with potentials of prognostic biomarkers in breast cancers.

microRNAs Patients’ cohort Expression level Clinical Association Refs

Single marker
miR-21 84 early BCs (stage I/II) vs 13 controls Increased according to Grade Decreased OS, DFS [110]
miR-10b-5p 101 BCs vs paired normal tissues Increased according to Metastasis Decreased OS, DFS [111]
miR-155 92 and 231 BCs Increased according to grade, TNM, TNBC subtype Decreased OS [199, 200]
miRNA signatures
miR-128a, miR-767-3p, miR -769-3p 207 early BCs Increased in ER positive Decreased OS [201]
miR-135a Increased in ER positive Increased OS
miR-27b, miR-144, miR-210 Increased in ER negative Decreased OS
miR-342, miR-150, miR-30c Increased in ER negative Increased OS
Let-7c/miR99a/miR125b cluster 285 ER positive BCs (from TCGA dataset) Increased in Luminal A BCs Increased OS [202]
Decreased in Luminal B and a subset of luminal A BCs Decreased OS
miR-493, miR-155 173 TNBC Increased in TNBC Increased OS [203]
miR-30e, miR-27a Decreased in TNBC Decreased OS
miR-200c 172 invasive BCs Increased in PR- tumors Decreased OS [204]
Decreased in PR+ tumors Decreased OS

28

Downloaded for Sitti Ayu Suhartina yahya Yahya (sitti.ayu.yahya@mail.ugm.ac.id) at Gadjah Mada University from ClinicalKey.com by Elsevier on October 23, 2019.
For personal use only. No other uses without permission. Copyright ©2019. Elsevier Inc. All rights reserved.
B. Pasculli et al. Seminars in Cancer Biology 51 (2018) 22–35

An integrated approach including chromatin immunoprecipitation, histological grade, and hormone receptor positive status. Accordingly,
gene expression analysis and whole genome mapping of transcription patients expressing high HDAC6 mRNA and protein levels had a better
factor-binding sites combined with gene expression profiling was im- disease free survival as compared with those expressing low levels.
plemented to identify genes involved in the proliferative response to However, multivariable analysis failed to show that HDAC6 mRNA and
oestrogen (E2). These analyses identified the increased expression of protein are independent prognostic factors [148].
the E2-inducible histone variant H2A.Z as significantly associated with The association between histone protein expression and resistance
lymph node metastasis and worse overall prognosis [134]. to anthracyclines was investigated by using a panel of cell lines re-
The constitutive activation through serine 139 phosphorylation of presentative of the major breast cancer subtypes [149]. Upregulation of
histone H2AX (γ-H2AX) has been associated in breast cancer cell lines histones H2A and H2 B was a common event in all epirubicin resistant
with a triple negative/basal like phenotype and/or with the presence of cell lines as compared with their sensitive counterpart. Moreover,
BRCA1 mutations. The analysis of a breast cancer cohort including epirubicin resistance could be reversed by histone deacetylase small
lymph node negative and triple negative cases (n = 122) demonstrated molecules. The involvement of histones in anthracycline resistance was
an association between H2AX immunostaining and worse prognosis in also evaluated in tumour samples from patients enrolled in the BR9601
multivariable analysis [135]. Notably, upon DNA damage, the phos- adjuvant clinical trial by using gene expression analysis. In particular,
phorylation at serine 139 of the histone H2AX is one of the first events patients showing low expression of an 18-genes histone module,
leading to the activation of DNA Double Strands Breaks (DSBs) repair benefited from anthracycline treatment more than those with high ex-
pathway, thus the constitutive activation of H2AX in Triple negative/ pression [149].
basal like breast cancer is likely to affect sensitivity to conventional Last, candidate predictive biomarkers have been investigated
chemo and radiotherapy treatments [135]. among ncRNAs but only a few have shown to associate with treatment
While the majority of HER2-amplified breast cancers are success- response in large patient cohorts. For example, higher miR-126 was
fully treated with specific monoclonal antibodies, 30% of patients identified as being associated with increased MFS in a small cohort of
eventually become resistant. A high frequency of PGR, HSD17B4, and breast cancer patients [150], and more recently was associated with
CDH13 methylation has been associated with HER-2 positive breast increased RFS (Relapse Free Survival) in a cohort of tamoxifen treated
tumours [136]. Interestingly, Fujii et al. found an association between BC patients [151], probably due to the coordinated targeting of the pro-
hypermethylation of the HSD17B4 gene promoter and complete re- apoptotic genes IGFBP2, PITPNC1 and MERTK [152,153]. Interestingly,
sponse to anti-HER2 treatment associated with chemotherapy [137]. miR-342 and miR-30c were associated with decreased distant relapse in
DNA promoter methylation of the breast cancer 1 (BRCA1) gene has ER-negative breast cancers [151], while both were found to associate
been associated to chemosensitivity in TNBC. BRCA1 germline muta- with clinical benefit in tamoxifen-treated patients [154,155]. Further-
tions account for 10–55% of familial breast cancers and 2% of breast more, miR-301 was shown to mediate resistance to tamoxifen and as-
cancers overall; nevertheless, 13–40% of sporadic breast cancers bear sociate with increased risk of relapse [156]. In this regard, the precise
BRCA1 gene promoter methylation [30, 138–140]. In a subgroup of 167 mechanisms of regulation of tamoxifen resistance remain largely un-
TNBC patients who received adjuvant chemotherapy, BRCA1 methy- known but the molecular targeting of PTEN by miR-301 in human
lation was an independent favorable predictor of 10 years disease breast cancer suggests the involvement of the miR-301/PTEN/Akt axis
specific survival (DSS) in multivariable analysis. By contrast, in 675 in the development of tamoxifen resistance. As concerns resistance to
non-triple-negative breast cancer patients who received adjuvant che- other therapies, miR-128 was found associated with sensitivity to
motherapy, BRCA1 methylation was predictor of worse survival in chemotherapeutics via Bmi-1 and ABCC5 regulation [157]; plasma
univariable analysis [140]. Thus, BRCA1 methylation could represent a miR-221 correlated with overall response rate in a cohort of che-
useful biomarker of chemotherapy sensitivity in triple negative tu- motherapy-treated patients [158], whereas circulating miR-125b pre-
mours. Preclinical studies and breast and ovarian cancer clinical trials dicted chemotherapy resistance [159]. Yet, high plasma levels of miR-
have shown the poly(adenosine diphosphate)-ribose polymerase 210 were associated with resistance to anti-HER2 Trastuzumab therapy
(PARP) inhibitors efficacy in BRCA1- and BRCA2-mutant patients [160] whereas high levels of miR-155 sensitized breast cancer to
[141,142]. Since BRCA1 promoter hypermethylated tumours show the radiotherapy [161].
same molecular phenotype than BRCA mutated cancers (BRCAness) Among lncRNAs, BC anti-estrogen resistance 4 (BCAR4) has been
[143] it has been proposed that BRCA1 hypermethylation might predict shown to predict tamoxifen resistance [162]. On the other hand,
response to PARP inhibitors. In an in vitro study, BRCA1 hypermethy- LINC00160 and LINC01016 were found to be downregulated in ER+
lation was able to confer the same degree of sensitivity to the three compared to ER- tumours and were early predictors of ET response
PARP inhibitors as the BRCA1 mutation did [144]. However, there are [163]. Increased levels of lncRNA-ATB (lncRNA activated by TGF-β)
no clinical data currently supporting the role of promoter methylation have been associated with Trastuzumab resistance in breast cancer by a
in the response to PARP1 inhibitors in non BRCA1 mutated breast mechanism of endogenous competition for the binding to miR-200c
cancer patients. that led to up-regulation of ZEB1 and ZNF-217, and induced EMT
Adding a taxane to chemotherapy improves outcome in breast [164]. Rather, CCAT2 (colon cancer associated transcript 2) down-
cancer patients [145,146]. Interestingly, in a cohort of 102 patients we regulation identified a subset of BC patients that might benefit from
demonstrated that aberrant promoter methylation of the KEAP1 (Kelch- cyclophosphamide, methotrexate and fluorouracil (CMF) based ad-
like ECH-associated protein 1) gene was associated in both univariable juvant chemotherapy [165]. Finally, Chen et al. [166] demonstrated
and multivariable analyses with a better overall survival (OS) in the that overexpression of lincRNA Regulator of Reprogramming (ROR) is
patients&rsquo; group receiving sequential therapy with anthracyclines associated chemotherapy tolerance [166].
and cyclophosphamide followed by taxanes, whereas no association To date, to the best of our knowledge, the measurement of miRNAs
was found in the group receiving anthracyclines and cyclophosphamide and evaluation of their biomarker potentialities have been included in
[147]. several on-going clinical trials as primary or secondary endpoints
The putative role of HDAC as prognostic factor in breast cancer is [167].
controversial. Increased expression HDAC1 but not HDAC3 as de-
termined by IHC was able to predict significantly a better disease free 4. Epigenetic therapies in breast cancer
survival in both univariable and multivariable analyses [107], whereas
Muller et al. [63] did not find any association between HDAC1 or The intense interest in epigenetics as therapeutic target stems from
HDAC2 and prognosis. Higher levels of HDAC6 expression in breast the fact that unlike gene alterations, aberrant hypermethylation as well
cancer were significantly associated with small tumours, low as histone PTMs are potentially reversible. Currently, there are two

29

Downloaded for Sitti Ayu Suhartina yahya Yahya (sitti.ayu.yahya@mail.ugm.ac.id) at Gadjah Mada University from ClinicalKey.com by Elsevier on October 23, 2019.
For personal use only. No other uses without permission. Copyright ©2019. Elsevier Inc. All rights reserved.
B. Pasculli et al. Seminars in Cancer Biology 51 (2018) 22–35

Table 4
Phase I/II clinical trials evaluating the efficacy of epigenetic therapies in breast cancer.

Therapeutical scheme N° patientsa Trial Phase Efficacyb Refs

Hydralazine 16 locally advanced or metastatic Phase II CR 31%; PR 50%, ORR 81%. [172]
+Doxorubicin BC)
+Cyclophosphamide
Vorinostat 14 BC Phase II No clinical benefits [173]
Vorinostat 43 Metastases on AI Phase II ORR 19% [174]
+Tamoxifen SD ≥ 24 months 21%
Vorinostat 55 Locally advanced or metastatic Phase I/II pCR Her2-positive patients 55% (95%CI 35–72 %), TNBC patients 28% (95%CI [176]
+Doxorubicin BC 11–52 %); ER-positive, Her2/neu negative 0%)
+Cyclophosphamide
Vorinostat 44 metastatic BC Phase I/II ORR 55% 95%CI: 39%–70% [175]
+Paclitaxel
+Bevacizumab
Entinostat 130 (EE 64; EP 66) Phase II randomized PFS 4.3 months EE versus 2.3 months for the EP (HR 0.73 (95%CI, 0.50–1.07; one- [179]
sided P = 0 .055; two-sided P = 0.11)
+Examestane OS 28.1 months EE versus 19.8 months EP (HR, 0.59; 95%CI, 0.36–0.97; P = 0.036)
Adverse effect 11% in EE vs 2% in EP
Entinostat 40 metastatic BC; 13 TNBC; 27 HT Phase II TNBC no clinical benefit; HT resistant PR 1 out of 27 ORR of 4% (95% CI 0–19). [180]
+5-Azacytidine resistant
Panobinostat 12 post-menopause metastatic BC Phase I PR 2 out of 12 [180]
+Letrozole SD 4 out of 12

a
BC Breast Cancer, AI Aromatase Inhibitors; EE Entinostat + Examestane; EP Eamestane + Placebo; TNBC Triple Negative Breast Cancer; HT, Hormone Therapy.
b
CR complete response; PR Partial Response; ORR Objective Response Rate; SD Stable Disease; CI Confidence Interval; PFS Progression Free Survival; OS Overall Survival.

types of epigenetic drugs approved in the clinics: DNMT inhibitors positive disease, in 4 of 15 patients with triple negative disease and
(DNMTi) and histone deacetylase small molecules inhibitors (HDACi) none of 12 patients with ER-positive, Her2/neu negative disease. The
[168,169]. The DNA methylation inhibitors 5-Azacytidine (Azacyti- efficacy of Vorinostat in combination with Paclitaxel and Bevacizumab
dine), 5-aza-2′-deoxycytidine (Decitabine) and hydralazine inhibit was evaluated in 54 metastatic breast cancer patients. In the 44 eva-
DNMTs by forming a covalent bond with the enzyme after incorpora- luable patients treated at the recommended phase II dose, there were 24
tion into DNA. HDAC inhibitors enhance the acetylation of cellular objective responses (55%, 95% CI 39–70%). In an intention-to-treat
proteins by blocking HDAC activity [55]. HDAC inhibition affects tu- analysis including all the 53 treated and eligible patients from the phase
mour growth, and apoptosis of cancer cells, whereas normal tissue is I and II portion of the trial, 26 (49%, 95%C.I. 37%-60%) exhibited an
less affected. HDACi are not only able to decompose and condense the objective response (2 CR and 24 confirmed PR), and 16 (30%) patients
histone–DNA complex, but also the acetylation status of non-histone had stable disease for more than 24 weeks.
proteins [55]. In a recent Phase I clinical trial, the pan-HDCA Panobinostat also
DNMT inhibitors and HDAC inhibitors have demonstrated anti- showed encouraging results in post-menopause ER positive metastatic
tumor activity in several tumour types and some of them have been disease when administered in combination with Letrozole [177].
recently approved by drug regulatory agencies for the clinical use in The more promising results in breast cancer have been obtained
haematological malignancies [168,169]. In breast cancer, both DNMTi with Entinostat, an HDAC inhibitor which potently and selectively in-
and HDCAi have demonstrated antitumor activity in preclinal studies hibits class I and IV HDAC enzymes and has been demonstrated to re-
[170,171], however the putative clinical benefits of these molecules are verse EMT phenotype in breast cancer cell lines [178]. An ECOG-ACRIN
still under investigation in clinical trials (Table 4). Phase III registration study is currently ongoing in advanced breast
In a proof of concept study, Arce et al. [171] evaluated 16 locally cancer (E2112, NCT02115282) to confirm the overall survival ad-
advanced or metastatic breast cancer patients treated preoperatory with vantage observed in combination with Exemestane in the Phase II set-
hydralazine and magnesium valproate, followed by doxorubicin and ting (ENCORE301 trial), that is a randomized, double-blind, placebo-
cyclophosphamide. A complete response (CR) was reached in 5 out of controlled study on locally advanced or metastatic BC whose disease
the 16 breast cancer patients (31%) and partial response in 8 out of the progressed on aromatase inhibitors. In this study, the administration of
16 patients (PR), thus the objective response rate (ORR) was 81%. A Entinostat in combination with Exemestane demonstrated an increase
statistically significant decrease in global 5mC content and HDAC ac- in median PFS in the group treated with Entinostat and Examestane (EE
tivity was demonstrated after treatment as compared to pre-treatment 4.3 months) as compared with the group treated with Examestane and
core biopsies [172]. placebo (EP 2.3 months) (HR of 0.73 95% CI 0.50-1.07; one-sided
In phase I/II clinical trials, the pan-HDAC inhibitor Vorinostat failed P = 0.055). Indeed, Median Overall Survival (OS) was 28.1 months for
to reach response criteria for single-agent activity [173] but showed the EE group and 19.8 months for the EP group (HR, 0.59; 95% CI,
clinical benefits in combination with anti-estrogen treatment [174] or 0.36–0.97; P = 0.036) [179]. Nevertheless, when the efficacy of com-
chemotherapeutic agents [175,176]. In the first case, Vorinostat was bining an epigenetic therapy with Entinostat and the DNA methyl-
administered in association to Tamoxifen in 43 women affected by ER- transferase inhibitor 5-azacitidine (AZA) was evaluated in a phase II
positive metastatic breast cancer progressing on aromatase inhibitors. study in women with advanced hormone-resistant or triple-negative
Confirmed objective responses according to RECIST criteria were de- breast cancer (TNBC) with the intention to re-express ER, no significant
monstrated in 8 out of 43 (19%) patients and stable disease for more clinical benefits were observed [180].
than 24 weeks in 9 out of 43 (21%) patients. The objective response rate
(ORR) was 19% and the clinical benefit rate determined as response or
stable disease after 24 weeks was 40% [174]. Yet, Tu et al. [176] 5. Concluding remarks
evaluated the association of Vorinostat with doxorubicin and cyclo-
phosphamide in 55 metastatic breast cancer patients. Pathological In closing, we can state that much is known about the pivotal role
complete response occurred in 13 of 24 evaluable patients with Her2- that epigenetic marks and ncRNAs play in human cancers, and beside
an in depth mechanistic investigation of their orchestrated networks

30

Downloaded for Sitti Ayu Suhartina yahya Yahya (sitti.ayu.yahya@mail.ugm.ac.id) at Gadjah Mada University from ClinicalKey.com by Elsevier on October 23, 2019.
For personal use only. No other uses without permission. Copyright ©2019. Elsevier Inc. All rights reserved.
B. Pasculli et al. Seminars in Cancer Biology 51 (2018) 22–35

underpinning cancer traits, many efforts are being made to demonstrate specific DNA methylation patterns in the human breast, Proc. Natl. Acad. Sci. U. S.
the suitability of these factors in the clinical setting of BC and other A. 105 (37) (2008) 14076–14081.
[11] R. Maruyama, S. Choudhury, A. Kowalczyk, M. Bessarabova, B. Beresford-Smith,
tumour types. Due to space restrictions, this review does not deal with T. Conway, A. Kaspi, Z. Wu, T. Nikolskaya, V.F. Merino, P.K. Lo, X.S. Liu,
the hot-topic of ncRNAs as novel therapeutics, exhaustively reviewed Y. Nikolsky, S. Sukumar, I. Haviv, K. Polyak, Epigenetic regulation of cell type-
by these works very recently [181,182]. A global review of the current specific expression patterns in the human mammary epithelium, PLoS Genet. 7 (4)
(2011) e1001369.
literature about the use of epigenetic traits as biomarkers reveals the [12] B. Pal, T. Bouras, W. Shi, F. Vaillant, J.M. Sheridan, N. Fu, K. Breslin, K. Jiang,
existence of several discrepancies that could be related to different M.E. Ritchie, M. Young, G.J. Lindeman, G.K. Smyth, J.E. Visvader, Global changes
cohorts’ size, sample preparation, availability of profiling technologies, in the mammary epigenome are induced by hormonal cues and coordinated by
Ezh2, Cell Rep. 3 (2) (2013) 411–426.
data analysis. For instance, detection methods for circulating ncRNAs [13] R. El Helou, J. Wicinski, A. Guille, J. Adélaïde, P. Finetti, F. Bertucci, M. Chaffanet,
are the most eligible for clinical implementation as well as the most D. Birnbaum, E. Charafe-Jauffret, C. Ginestier, Brief reports: a distinct DNA me-
affected by issues concerning pre-analytical procedures such as the thylation signature defines breast cancer stem cells and predicts cancer outcome,
Stem Cells 32 (11) (2014) 3031–3036.
choice of the starting material (serum, plasma, whole blood, that are
[14] V.J. Peschansky, C. Wahlestedt, Non-coding RNAs as direct and indirect mod-
not inter-changeable) and processing, methods of quantification, ran- ulators of epigenetic regulation, Epigenetics 9 (1) (2014) 3–12.
ging from probe-based or LNA-based quantitative PCR array, micro- [15] J. Veeck, M. Esteller, Breast cancer epigenetics: from DNA methylation to
array to Nanostring nCounter technology and RNA seq, and the relative microRNAs, J. Mammary Gland Biol. Neoplasia 15 (1) (2010) 5–17.
[16] S. Karsli-Ceppioglu, A. Dagdemir, G. Judes, M. Ngollo, F. Penault-Llorca, A. Pajon,
data normalization. Y.J. Bignon, D. Bernard-Gallon, Epigenetic mechanisms of breast cancer: an up-
Despite these pending issues, we might expect that along with the date of the current knowledge, Epigenomics 6 (6) (2014) 651–664.
analysis of cancer-related genes mutations and chromosomal aberra- [17] A.M. Deaton, A. Bird, CpG islands and the regulation of transcription, Genes Dev.
25 (10) (2011) 1010–1022.
tions, the characterization of epigenetic modifications and detection of [18] J. Jovanovic, J.A. Rønneberg, J. Tost, V. Kristensen, The epigenetics of breast
ncRNAs will enter the routine clinical investigation very soon, towards cancer, Mol. Oncol. 4 (3) (2010) 242–254.
the development of novel screening tools for early cancer detection as [19] P. Novak, T. Jensen, M.M. Oshiro, G.S. Watts, C.J. Kim, B.W. Futscher,
Agglomerative epigenetic aberrations are a common event in human breast
well as diagnostic and prognostic devises, and companion tests for cancer, Cancer Res. 68 (20) (2008) 8616–8625.
addressing cancer patients to the most beneficial treatments with the [20] K.S. Son, H.S. Kang, S.J. Kim, S.Y. Jung, S.Y. Min, S.Y. Lee, S.W. Kim, Y. Kwon,
aim to ameliorate the overall management and monitoring of patients. K.S. Lee, K.H. Shin, J. Ro, Hypomethylation of the interleukin-10 gene in breast
cancer tissues, Breast 19 (6) (2010) 484–488.
[21] G. Sharma, S. Mirza, R. Parshad, A. Srivastava, S. Datta Gupta, P. Pandya,
Acknowledgments R. Ralhan, CpG hypomethylation of MDR1 gene in tumor and serum of invasive
ductal breast carcinoma patients, Clin. Biochem. 43 (4–5) (2010) 373–379.
[22] P. Singh, M. Yang, H. Dai, D. Yu, Q. Huang, W. Tan, K.H. Kernstine, D. Lin,
Work in P. Parrella lab is supported by the Italian Ministry of Health
B. Shen, Overexpression and hypomethylation of flap endonuclease 1 gene in
(MoH) co-funded by the European Regional Development Fund “A way breast and other cancers, Mol. Cancer Res. 6 (11) (2008) 1710–1717.
of making Europe” under the TRANSCAN ERA-NET on Translational [23] S.J. Kim, H.S. Kang, H.L. Chang, Y.C. Jung, H.B. Sim, K.S. Lee, J. Ro, E.S. Lee,
Cancer Research grant no. RRC-2014-2354565 and CANCER13-FP-011; Promoter hypomethylation of the N-acetyltransferase 1 gene in breast cancer,
Oncol. Rep. 19 (3) (2008) 663–668.
Italian Ministry of Health (MoH) “Ricerca Corrente 2016” and [24] J. Paredes, A. Albergaria, J.T. Oliveira, C. Jerónimo, F. Milanezi, F.C. Schmitt, P-
“5 × 1000” voluntary contributions”; and “Associazione Italiana cadherin overexpression is an indicator of clinical outcome in invasive breast
Ricerca sul Cancro” (AIRC) IG-1269/2006 and IG-16647/2015. carcinomas and is associated with CDH3 promoter hypomethylation, Clin. Cancer
Res. 11 (16) (2005) 5869–5877.
[25] P. Pakneshan, M. Szyf, R. Farias-Eisner, S.A. Rabbani, Reversal of the hypo-
References methylation status of urokinase (uPA) promoter blocks breast cancer growth and
metastasis, J. Biol. Chem. 279 (30) (2004) 31735–31744.
[26] A. Gupta, A.K. Godwin, L. Vanderveer, A. Lu, J. Liu, Hypomethylation of the sy-
[1] R.L. Siegel, K.D. Miller, A. Jemal, Cancer statistics, CA Cancer J. Clin. 67 (1)
nuclein gamma gene CpG island promotes its aberrant expression in breast car-
(2017) 7–30.
cinoma and ovarian carcinoma, Cancer Res. 63 (3) (2003) 664–673.
[2] C.K. Osborne, M.G. Yochmowitz, W.A. Knight, W.L. McGuire, The value of es-
[27] Y. Cao, Y. Li, N. Zhang, J. Hu, L. Yin, Z. Pan, X. Du, W. Zhang, F. Li, Quantitative
trogen and progesterone receptors in the treatment of breast cancer, Cancer 46 (12
DNA hypomethylation of ligand Jagged1 and receptor Notch1 signifies occurrence
(Suppl.)) (1980) 2884–2888.
and progression of breast carcinoma, Am. J. Cancer Res. 5 (6) (2015) 1897–1910.
[3] J.S. Ross, E.A. Slodkowska, W.F. Symmans, L. Pusztai, P.M. Ravdin,
[28] M. Hemberger, W. Dean, W. Reik, Epigenetic dynamics of stem cells and cell
G.N. Hortobagyi, The HER-2 receptor and breast cancer: ten years of targeted anti-
lineage commitment: digging Waddington's canal, Nat. Rev. Mol. Cell Biol. 10 (8)
HER-2 therapy and personalized medicine, Oncologist 14 (4) (2009) 320–368.
(2009) 526–537.
[4] T. Sorlie, R. Tibshirani, J. Parker, T. Hastie, J.S. Marron, A. Nobel, S. Deng,
[29] G. Severi, M.C. Southey, D.R. English, C.H. Jung, A. Lonie, C. McLean,
H. Johnsen, R. Pesich, S. Geisler, J. Demeter, C.M. Perou, P.E. Lønning,
H. Tsimiklis, J.L. Hopper, G.G. Giles, L. Baglietto, Epigenome-wide methylation in
P.O. Brown, A.L. Børresen-Dale, D. Botstein, Repeated observation of breast tumor
DNA from peripheral blood as a marker of risk for breast cancer, Breast Cancer
subtypes in independent gene expression data sets, Proc. Natl. Acad. Sci. U. S. A.
Res. Treat. 148 (3) (2014) 665–673.
100 (14) (2003) 8418–8423.
[30] P. Parrella, M. Poeta, A. Gallo, M. Prencipe, M. Scintu, A. Apicella, R. Rossiello,
[5] C.M. Perou, T. Sørlie, M.B. Eisen, M. van de Rijn, S.S. Jeffrey, C.A. Rees,
G. Liguoro, D. Seripa, C. Gravina, C. Rabitti, M. Rinaldi, T. Nicol, S. Tommasi,
J.R. Pollack, D.T. Ross, H. Johnsen, L.A. Akslen, O. Fluge, A. Pergamenschikov,
A. Paradiso, F. Schittulli, V. Altomare, V. Fazio, Nonrandom distribution of
C. Williams, S.X. Zhu, P.E. Lønning, A.L. Børresen-Dale, P.O. Brown, D. Botstein,
aberrant promoter methylation of cancer-related genes in sporadic breast tumors,
Molecular portraits of human breast tumours, Nature 406 (6797) (2000) 747–752.
Clin. Cancer Res. 10 (16) (2004) 5349–5354.
[6] T. Sørlie, C.M. Perou, R. Tibshirani, T. Aas, S. Geisler, H. Johnsen, T. Hastie,
[31] N.G. Bediaga, A. Acha-Sagredo, I. Guerra, A. Viguri, C. Albaina, I. Ruiz Diaz,
M.B. Eisen, M. van de Rijn, S.S. Jeffrey, T. Thorsen, H. Quist, J.C. Matese,
R. Rezola, M.J. Alberdi, J. Dopazo, D. Montaner, M. Renobales, A.F. Fernández,
P.O. Brown, D. Botstein, P.E. Lønning, A.L. Børresen-Dale, Gene expression pat-
J.K. Field, M.F. Fraga, T. Liloglou, M.M. de Pancorbo, DNA methylation epigen-
terns of breast carcinomas distinguish tumor subclasses with clinical implications,
otypes in breast cancer molecular subtypes, Breast Cancer Res. 12 (5) (2010) R77.
Proc. Natl. Acad. Sci. U. S. A. 98 (19) (2001) 10869–10874.
[32] T. Fleischer, A. Frigessi, K.C. Johnson, H. Edvardsen, N. Touleimat, J. Klajic,
[7] L.J. van ‘t Veer, H. Dai, M.J. van de Vijver, Y.D. He, A.A. Hart, M. Mao,
M.L. Riis, V.D. Haakensen, F. Wärnberg, B. Naume, A. Helland, A.L. Børresen-Dale,
H.L. Peterse, K. van der Kooy, M.J. Marton, A.T. Witteveen, G.J. Schreiber,
J. Tost, B.C. Christensen, V.N. Kristensen, Genome-wide DNA methylation profiles
R.M. Kerkhoven, C. Roberts, P.S. Linsley, R. Bernards, S.H. Friend, Gene expres-
in progression to in situ and invasive carcinoma of the breast with impact on gene
sion profiling predicts clinical outcome of breast cancer, Nature 415 (6871) (2002)
transcription and prognosis, Genome Biol. 15 (8) (2014) 435.
530–536.
[33] E. Martín-Sánchez, S. Mendaza, A. Ulazia-Garmendia, I. Monreal-Santesteban,
[8] A.E. Giuliano, J.L. Connolly, S.B. Edge, E.A. Mittendorf, H.S. Rugo, L.J. Solin,
I. Blanco-Luquin, A. Córdoba, F. Vicente-García, N. Pérez-Janices, D. Escors,
D.L. Weaver, D.J. Winchester, G.N. Hortobagyi, Breast cancer-major changes in
D. Megías, P. López-Serra, M. Esteller, J.J. Illarramendi, D. Guerrero-Setas, CHL1
the American Joint Committee on cancer eighth edition cancer staging manual, CA
hypermethylation as a potential biomarker of poor prognosis in breast cancer,
Cancer J. Clin. 67 (4) (2017) 290–303.
Oncotarget 8 (9) (2017) 15789–15801.
[9] R.A. Hinshelwood, S.J. Clark, Breast cancer epigenetics: normal human mammary
[34] M.O. Hoque, M. Prencipe, M.L. Poeta, R. Barbano, V.M. Valori, M. Copetti,
epithelial cells as a model system, J. Mol. Med. (Berl) 86 (12) (2008) 1315–1328.
A.P. Gallo, M. Brait, E. Maiello, A. Apicella, R. Rossiello, F. Zito, T. Stefania,
[10] N. Bloushtain-Qimron, J. Yao, E.L. Snyder, M. Shipitsin, L.L. Campbell, S.A. Mani,
A. Paradiso, M. Carella, B. Dallapiccola, R. Murgo, I. Carosi, M. Bisceglia,
M. Hu, H. Chen, V. Ustyansky, J.E. Antosiewicz, P. Argani, M.K. Halushka,
V.M. Fazio, D. Sidransky, P. Parrella, Changes in CpG islands promoter methyla-
J.A. Thomson, P. Pharoah, A. Porgador, S. Sukumar, R. Parsons, A.L. Richardson,
tion patterns during ductal breast carcinoma progression, Cancer Epidemiol.
M.R. Stampfer, R.S. Gelman, T. Nikolskaya, Y. Nikolsky, K. Polyak, Cell type-
Biomar. Prevention 18 (10) (2009) 2694–2700.

31

Downloaded for Sitti Ayu Suhartina yahya Yahya (sitti.ayu.yahya@mail.ugm.ac.id) at Gadjah Mada University from ClinicalKey.com by Elsevier on October 23, 2019.
For personal use only. No other uses without permission. Copyright ©2019. Elsevier Inc. All rights reserved.
B. Pasculli et al. Seminars in Cancer Biology 51 (2018) 22–35

[35] A.Q. van Hoesel, Y. Sato, D.A. Elashoff, R.R. Turner, A.E. Giuliano, J.M. Shamonki, Y. Nishioka, H. Kimura, K. Yoshidome, M. Tsujimoto, N. Matsuura, Loss of histone
P.J. Kuppen, C.J. van de Velde, D.S. Hoon, Assessment of DNA methylation status H4K20 trimethylation predicts poor prognosis in breast cancer and is associated
in early stages of breast cancer development, Br. J. Cancer 108 (10) (2013) with invasive activity, Breast Cancer Res. 16 (3) (2014) R66.
2033–2038. [61] C.A. Krusche, P. Wülfing, C. Kersting, A. Vloet, W. Böcker, L. Kiesel, H.M. Beier,
[36] A.Q. van Hoesel, C.J. van de Velde, P.J. Kuppen, G.J. Liefers, H. Putter, Y. Sato, J. Alfer, Histone deacetylase-1 and −3 protein expression in human breast cancer:
D.A. Elashoff, R.R. Turner, J.M. Shamonki, E.M. de Kruijf, J.G. van Nes, a tissue microarray analysis, Breast Cancer Res. Treat. 90 (1) (2005) 15–23.
A.E. Giuliano, D.S. Hoon, Hypomethylation of LINE-1 in primary tumor has poor [62] Z. Zhang, H. Yamashita, T. Toyama, H. Sugiura, Y. Ando, K. Mita, M. Hamaguchi,
prognosis in young breast cancer patients: a retrospective cohort study, Breast Y. Hara, S. Kobayashi, H. Iwase, Quantitation of HDAC1 mRNA expression in in-
Cancer Res. Treat. 134 (3) (2012) 1103–1114. vasive carcinoma of the breast, Breast Cancer Res. Treat. 94 (1) (2005) 11–16.
[37] A.E. Teschendorff, Y. Gao, A. Jones, M. Ruebner, M.W. Beckmann, D.L. Wachter, [63] B.M. Müller, L. Jana, A. Kasajima, A. Lehmann, J. Prinzler, J. Budczies,
P.A. Fasching, M. Widschwendter, DNA methylation outliers in normal breast K.J. Winzer, M. Dietel, W. Weichert, C. Denkert, Differential expression of histone
tissue identify field defects that are enriched in cancer, Nat. Commun. 7 (2016) deacetylases HDAC1, 2 and 3 in human breast cancer–overexpression of HDAC2
10478. and HDAC3 is associated with clinicopathological indicators of disease progres-
[38] R.D. Kornberg, Chromatin structure: a repeating unit of histones and DNA, Science sion, BMC Cancer 13 (2013) 215.
184 (4139) (1974) 868–871. [64] H. Ling, M. Fabbri, G.A. Calin, MicroRNAs and other non-coding RNAs as targets
[39] K. Luger, M.L. Dechassa, D.J. Tremethick, New insights into nucleosome and for anticancer drug development, Nat. Rev. Drug Discov. 12 (11) (2013) 847–865.
chromatin structure: an ordered state or a disordered affair? Nat. Rev. Mol. Cell [65] M. Esteller, Non-coding RNAs in human disease, Nat. Rev. Genet. 12 (12) (2011)
Biol. 13 (7) (2012) 436–447. 861–874.
[40] M. Tan, H. Luo, S. Lee, F. Jin, J.S. Yang, E. Montellier, T. Buchou, Z. Cheng, [66] E.P. Consortium, An integrated encyclopedia of DNA elements in the human
S. Rousseaux, N. Rajagopal, Z. Lu, Z. Ye, Q. Zhu, J. Wysocka, Y. Ye, S. Khochbin, genome, Nature 489 (7414) (2012) 57–74.
B. Ren, Y. Zhao, Identification of 67 histone marks and histone lysine crotonyla- [67] S. Djebali, C.A. Davis, A. Merkel, A. Dobin, T. Lassmann, A. Mortazavi, A. Tanzer,
tion as a new type of histone modification, Cell 146 (6) (2011) 1016–1028. J. Lagarde, W. Lin, F. Schlesinger, C. Xue, G.K. Marinov, J. Khatun, B.A. Williams,
[41] A. Barski, S. Cuddapah, K. Cui, T.Y. Roh, D.E. Schones, Z. Wang, G. Wei, C. Zaleski, J. Rozowsky, M. Röder, F. Kokocinski, R.F. Abdelhamid, T. Alioto,
I. Chepelev, K. Zhao, High-resolution profiling of histone methylations in the I. Antoshechkin, M.T. Baer, N.S. Bar, P. Batut, K. Bell, I. Bell, S. Chakrabortty,
human genome, Cell 129 (4) (2007) 823–837. X. Chen, J. Chrast, J. Curado, T. Derrien, J. Drenkow, E. Dumais, J. Dumais,
[42] N.D. Heintzman, R.K. Stuart, G. Hon, Y. Fu, C.W. Ching, R.D. Hawkins, R. Duttagupta, E. Falconnet, M. Fastuca, K. Fejes-Toth, P. Ferreira, S. Foissac,
L.O. Barrera, S. Van Calcar, C. Qu, K.A. Ching, W. Wang, Z. Weng, R.D. Green, M.J. Fullwood, H. Gao, D. Gonzalez, A. Gordon, H. Gunawardena, C. Howald,
G.E. Crawford, B. Ren, Distinct and predictive chromatin signatures of transcrip- S. Jha, R. Johnson, P. Kapranov, B. King, C. Kingswood, O.J. Luo, E. Park,
tional promoters and enhancers in the human genome, Nat. Genet. 39 (3) (2007) K. Persaud, J.B. Preall, P. Ribeca, B. Risk, D. Robyr, M. Sammeth, L. Schaffer,
311–318. L.H. See, A. Shahab, J. Skancke, A.M. Suzuki, H. Takahashi, H. Tilgner, D. Trout,
[43] N.D. Heintzman, G.C. Hon, R.D. Hawkins, P. Kheradpour, A. Stark, L.F. Harp, N. Walters, H. Wang, J. Wrobel, Y. Yu, X. Ruan, Y. Hayashizaki, J. Harrow,
Z. Ye, L.K. Lee, R.K. Stuart, C.W. Ching, K.A. Ching, J.E. Antosiewicz-Bourget, M. Gerstein, T. Hubbard, A. Reymond, S.E. Antonarakis, G. Hannon,
H. Liu, X. Zhang, R.D. Green, V.V. Lobanenkov, R. Stewart, J.A. Thomson, M.C. Giddings, Y. Ruan, B. Wold, P. Carninci, R. Guigó, T.R. Gingeras, Landscape
G.E. Crawford, M. Kellis, B. Ren, Histone modifications at human enhancers reflect of transcription in human cells, Nature 489 (7414) (2012) 101–108.
global cell-type-specific gene expression, Nature 459 (7243) (2009) 108–112. [68] G. Di Leva, M. Garofalo, C.M. Croce, MicroRNAs in cancer, Annu. Rev. Pathol. 9
[44] H. O’Geen, S.L. Squazzo, S. Iyengar, K. Blahnik, J.L. Rinn, H.Y. Chang, R. Green, (2014) 287–314.
P.J. Farnham, Genome-wide analysis of KAP1 binding suggests autoregulation of [69] S. Chandra Gupta, Y. Nandan Tripathi, Potential of long non-coding RNAs in
KRAB-ZNFs, PLoS Genet. 3 (6) (2007) e89. cancer patients: from biomarkers to therapeutic targets, Int. J. Cancer 140 (9)
[45] T. Kouzarides, Chromatin modifications and their function, Cell 128 (4) (2007) (2017) 1955–1967.
693–705. [70] Y. Li, Y. Zhang, S. Li, J. Lu, J. Chen, Y. Wang, J. Xu, X. Li, Genome-wide DNA
[46] T.S. Mikkelsen, M. Ku, D.B. Jaffe, B. Issac, E. Lieberman, G. Giannoukos, methylome analysis reveals epigenetically dysregulated non-coding RNAs in
P. Alvarez, W. Brockman, T.K. Kim, R.P. Koche, W. Lee, E. Mendenhall, human breast cancer, Sci. Rep. 5 (2015) 8790.
A. O'Donovan, A. Presser, C. Russ, X. Xie, A. Meissner, M. Wernig, R. Jaenisch, [71] M.V. Iorio, M. Ferracin, C.G. Liu, A. Veronese, R. Spizzo, S. Sabbioni, E. Magri,
C. Nusbaum, E.S. Lander, B.E. Bernstein, Genome-wide maps of chromatin state in M. Pedriali, M. Fabbri, M. Campiglio, S. Ménard, J.P. Palazzo, A. Rosenberg,
pluripotent and lineage-committed cells, Nature 448 (7153) (2007) 553–560. P. Musiani, S. Volinia, I. Nenci, G.A. Calin, P. Querzoli, M. Negrini, C.M. Croce,
[47] T. Jenuwein, C.D. Allis, Translating the histone code, Science 293 (5532) (2001) MicroRNA gene expression deregulation in human breast cancer, Cancer Res. 65
1074–1080. (16) (2005) 7065–7070.
[48] Q.Y. Zhao, P.J. Lei, X. Zhang, J.Y. Zheng, H.Y. Wang, J. Zhao, Y.M. Li, M. Ye, L. Li, [72] E. de Rinaldis, P. Gazinska, A. Mera, Z. Modrusan, G.M. Fedorowicz, B. Burford,
G. Wei, M. Wu, Global histone modification profiling reveals the epigenomic dy- C. Gillett, P. Marra, A. Grigoriadis, D. Dornan, L. Holmberg, S. Pinder, A. Tutt,
namics during malignant transformation in a four-stage breast cancer model, Clin. Integrated genomic analysis of triple-negative breast cancers reveals novel
Epigenetics 8 (2016) 34. microRNAs associated with clinical and molecular phenotypes and sheds light on
[49] T.L. Messier, J.A. Gordon, J.R. Boyd, C.E. Tye, G. Browne, J.L. Stein, J.B. Lian, the pathways they control, BMC Genom. 14 (2013) 643.
G.S. Stein, Histone H3 lysine 4 acetylation and methylation dynamics define breast [73] E. van Schooneveld, H. Wildiers, I. Vergote, P.B. Vermeulen, L.Y. Dirix, S.J. Van
cancer subtypes, Oncotarget 7 (5) (2016) 5094–5109. Laere, Dysregulation of microRNAs in breast cancer and their potential role as
[50] X. Chen, H. Hu, L. He, X. Yu, X. Liu, R. Zhong, M. Shu, A novel subtype classifi- prognostic and predictive biomarkers in patient management, Breast Cancer Res.
cation and risk of breast cancer by histone modification profiling, Breast Cancer 17 (2015) 21.
Res. Treat 157 (2) (2016) 267–279. [74] I. Fkih M’hamed, M. Privat, F. Ponelle, F. Penault-Llorca, A. Kenani, Y.J. Bignon,
[51] L. Millán-Ariño, A.B. Islam, A. Izquierdo-Bouldstridge, R. Mayor, J.M. Terme, Identification of miR-10b, miR-26a, miR-146a and miR-153 as potential triple-
N. Luque, M. Sancho, N. López-Bigas, A. Jordan, Mapping of six somatic linker negative breast cancer biomarkers, Cell Oncol. (Dordr) 38 (6) (2015) 433–442.
histone H1 variants in human breast cancer cells uncovers specific features of [75] M. Huarte, The emerging role of lncRNAs in cancer, Nat. Med. 21 (11) (2015)
H1.2, Nucleic Acids Res. 42 (7) (2014) 4474–4493. 1253–1261.
[52] S.W. Harshman, M.E. Hoover, C. Huang, O.E. Branson, S.B. Chaney, C.M. Cheney, [76] M. Guttman, J.L. Rinn, Modular regulatory principles of large non-coding RNAs,
T.J. Rosol, C.L. Shapiro, V.H. Wysocki, K. Huebner, M.A. Freitas, Histone H1 Nature 482 (7385) (2012) 339–346.
phosphorylation in breast cancer, J. Proteome Res. 13 (5) (2014) 2453–2467. [77] S.W. Cheetham, F. Gruhl, J.S. Mattick, M.E. Dinger, Long noncoding RNAs and the
[53] M. Haberland, R.L. Montgomery, E.N. Olson, The many roles of histone deacety- genetics of cancer, Br. J. Cancer 108 (12) (2013) 2419–2425.
lases in development and physiology: implications for disease and therapy, Nat. [78] W. Zhao, J. Luo, S. Jiao, Comprehensive characterization of cancer subtype as-
Rev. Genet. 10 (1) (2009) 32–42. sociated long non-coding RNAs and their clinical implications, Sci. Rep. 4 (2014)
[54] M. Yun, J. Wu, J.L. Workman, B. Li, Readers of histone modifications, Cell Res. 21 6591.
(4) (2011) 564–578. [79] R.A. Gupta, N. Shah, K.C. Wang, J. Kim, H.M. Horlings, D.J. Wong, M.C. Tsai,
[55] H.J. Kim, S.C. Bae, Histone deacetylase inhibitors: molecular mechanisms of ac- T. Hung, P. Argani, J.L. Rinn, Y. Wang, P. Brzoska, B. Kong, R. Li, R.B. West,
tion and clinical trials as anti-cancer drugs, Am. J. Transl. Res. 3 (2) (2011) M.J. van de Vijver, S. Sukumar, H.Y. Chang, Long non-coding RNA HOTAIR re-
166–179. programs chromatin state to promote cancer metastasis, Nature 464 (7291) (2010)
[56] A.J. de Ruijter, A.H. van Gennip, H.N. Caron, S. Kemp, A.B. van Kuilenburg, 1071–1076.
Histone deacetylases (HDACs): characterization of the classical HDAC family, [80] L. Ma, Role of miR-10b in breast cancer metastasis, Breast Cancer Res. 12 (5)
Biochem. J. 370 (Pt. 3) (2003) 737–749. (2010) 210.
[57] G. Blander, L. Guarente, The Sir2 family of protein deacetylases, Annu. Rev. [81] J.L. Rinn, M. Kertesz, J.K. Wang, S.L. Squazzo, X. Xu, S.A. Brugmann,
Biochem. 73 (2004) 417–435. L.H. Goodnough, J.A. Helms, P.J. Farnham, E. Segal, H.Y. Chang, Functional de-
[58] G. Arun, S. Diermeier, M. Akerman, K.C. Chang, J.E. Wilkinson, S. Hearn, Y. Kim, marcation of active and silent chromatin domains in human HOX loci by non-
A.R. MacLeod, A.R. Krainer, L. Norton, E. Brogi, M. Egeblad, D.L. Spector, coding RNAs, Cell 129 (7) (2007) 1311–1323.
Differentiation of mammary tumors and reduction in metastasis upon Malat1 [82] D. Meseure, S. Vacher, F. Lallemand, K.D. Alsibai, R. Hatem, W. Chemlali,
lncRNA loss, Genes Dev. 30 (1) (2016) 34–51. A. Nicolas, L. De Koning, E. Pasmant, C. Callens, R. Lidereau, A. Morillon,
[59] S. Lim, A. Janzer, A. Becker, A. Zimmer, R. Schüle, R. Buettner, J. Kirfel, Lysine- I. Bieche, Prognostic value of a newly identified MALAT1 alternatively spliced
specific demethylase 1 (LSD1) is highly expressed in ER-negative breast cancers transcript in breast cancer, Br. J. Cancer 114 (12) (2016) 1395–1404.
and a biomarker predicting aggressive biology, Carcinogenesis 31 (3) (2010) [83] R. Burnette, L.A. Simmons, R. Snyderman, Personalized health care as a pathway
512–520. for the adoption of genomic medicine, J. Pers. Med. 2 (4) (2012) 232–240.
[60] Y. Yokoyama, A. Matsumoto, M. Hieda, Y. Shinchi, E. Ogihara, M. Hamada, [84] R.T. Pu, L.E. Laitala, P.M. Alli, M.J. Fackler, S. Sukumar, D.P. Clark, Methylation

32

Downloaded for Sitti Ayu Suhartina yahya Yahya (sitti.ayu.yahya@mail.ugm.ac.id) at Gadjah Mada University from ClinicalKey.com by Elsevier on October 23, 2019.
For personal use only. No other uses without permission. Copyright ©2019. Elsevier Inc. All rights reserved.
B. Pasculli et al. Seminars in Cancer Biology 51 (2018) 22–35

profiling of benign and malignant breast lesions and its application to cyto- [110] A. Markou, G.M. Yousef, E. Stathopoulos, V. Georgoulias, E. Lianidou, Prognostic
pathology, Mod. Pathol. 16 (11) (2003) 1095–1101. significance of metastasis-related microRNAs in early breast cancer patients with a
[85] C. Jerónimo, I. Costa, M.C. Martins, P. Monteiro, S. Lisboa, C. Palmeira, long follow-up, Clin. Chem. 60 (1) (2014) 197–205.
R. Henrique, M.R. Teixeira, C. Lopes, Detection of gene promoter hypermethyla- [111] P. Parrella, R. Barbano, B. Pasculli, A. Fontana, M. Copetti, V.M. Valori,
tion in fine needle washings from breast lesions, Clin. Cancer Res. 9 (9) (2003) M.L. Poeta, G. Perrone, D. Righi, M. Castelvetere, M. Coco, T. Balsamo,
3413–3417. M. Morritti, F. Pellegrini, A. Onetti-Muda, E. Maiello, R. Murgo, V.M. Fazio,
[86] E. Evron, W.C. Dooley, C.B. Umbricht, D. Rosenthal, N. Sacchi, E. Gabrielson, Evaluation of microRNA-10b prognostic significance in a prospective cohort of
A.B. Soito, D.T. Hung, B. Ljung, N.E. Davidson, S. Sukumar, Detection of breast breast cancer patients, Mol. Cancer 13 (2014) 142.
cancer cells in ductal lavage fluid by methylation-specific PCR, Lancet 357 (9265) [112] S. Volinia, C.M. Croce, Prognostic microRNA/mRNA signature from the integrated
(2001) 1335–1336. analysis of patients with invasive breast cancer, Proc. Natl. Acad. Sci. U. S. A. 110
[87] R. Krassenstein, E. Sauter, E. Dulaimi, C. Battagli, H. Ehya, A. Klein-Szanto, (18) (2013) 7413–7417.
P. Cairns, Detection of breast cancer in nipple aspirate fluid by CpG island hy- [113] K.P. Sørensen, M. Thomassen, Q. Tan, M. Bak, S. Cold, M. Burton, M.J. Larsen,
permethylation, Clin. Cancer Res. 10 (1 (Pt. 1)) (2004) 28–32. T.A. Kruse, Long non-coding RNA HOTAIR is an independent prognostic marker of
[88] M.O. Hoque, Q. Feng, P. Toure, A. Dem, C.W. Critchlow, S.E. Hawes, T. Wood, metastasis in estrogen receptor-positive primary breast cancer, Breast Cancer Res.
C. Jeronimo, E. Rosenbaum, J. Stern, M. Yu, B. Trink, N.B. Kiviat, D. Sidransky, Treat. 142 (3) (2013) 529–536.
Detection of aberrant methylation of four genes in plasma DNA for the detection of [114] P. Magee, L. Shi, M. Garofalo, Role of microRNAs in chemoresistance, Ann. Transl.
breast cancer, J. Clin. Oncol. 24 (26) (2006) 4262–4269. Med. 3 (21) (2015) 332.
[89] H. Schwarzenbach, N. Nishida, G.A. Calin, K. Pantel, Clinical relevance of circu- [115] Y. Shen, D. Katsaros, L.W. Loo, B.Y. Hernandez, C. Chong, E.M. Canuto, N. Biglia,
lating cell-free microRNAs in cancer, Nat. Rev. Clin. Oncol. 11 (3) (2014) L. Lu, H. Risch, W.M. Chu, H. Yu, Prognostic and predictive values of long non-
145–156. coding RNA LINC00472 in breast cancer, Oncotarget 6 (11) (2015) 8579–8592.
[90] Y.K. Tong, Y.M. Lo, Diagnostic developments involving cell-free (circulating) nu- [116] L.L. Deng, Y.Y. Chi, L. Liu, N.S. Huang, L. Wang, J. Wu, LINC00978 predicts poor
cleic acids, Clin. Chim. Acta 363 (1–2) (2006) 187–196. prognosis in breast cancer patients, Sci. Rep. 6 (2016) 37936.
[91] P. Qi, X.Y. Zhou, X. Du, Circulating long non-coding RNAs in cancer: current status [117] H. Liu, J. Li, P. Koirala, X. Ding, B. Chen, Y. Wang, Z. Wang, C. Wang, X. Zhang,
and future perspectives, Mol. Cancer 15 (1) (2016) 39. Y.Y. Mo, Long non-coding RNAs as prognostic markers in human breast cancer,
[92] K. Schlosser, J. Hanson, P.J. Villeneuve, J. Dimitroulakos, L. McIntyre, L. Pilote, Oncotarget 7 (15) (2016) 20584–20596.
D.J. Stewart, Assessment of circulating LncRNAs under physiologic and pathologic [118] O. Van Grembergen, M. Bizet, E.J. de Bony, E. Calonne, P. Putmans, S. Brohée,
conditions in humans reveals potential limitations as biomarkers, Sci. Rep. 6 C. Olsen, M. Guo, G. Bontempi, C. Sotiriou, M. Defrance, F. Fuks, Portraying breast
(2016) 36596. cancers with long noncoding RNAs, Sci. Adv. 2 (9) (2016) e1600220.
[93] F. Wang, Z. Zheng, J. Guo, X. Ding, Correlation and quantitation of microRNA [119] N. Berteaux, N. Aptel, G. Cathala, C. Genton, J. Coll, A. Daccache, N. Spruyt,
aberrant expression in tissues and sera from patients with breast tumor, Gynecol. H. Hondermarck, T. Dugimont, J.J. Curgy, T. Forné, E. Adriaenssens, A novel H19
Oncol. 119 (3) (2010) 586–593. antisense RNA overexpressed in breast cancer contributes to paternal IGF2 ex-
[94] N. Xu, F. Chen, F. Wang, X. Lu, X. Wang, M. Lv, C. Lu, Clinical significance of high pression, Mol. Cell Biol. 28 (22) (2008) 6731–6745.
expression of circulating serum lncRNA RP11-445H22. 4 in breast cancer patients: [120] E. Raveh, I.J. Matouk, M. Gilon, A. Hochberg, The H19 Long non-coding RNA in
a Chinese population-based study, Tumour Biol. 36 (10) (2015) 7659–7665. cancer initiation, progression and metastasis − a proposed unifying theory, Mol.
[95] X. Wang, W. Zhang, J. Tang, R. Huang, J. Li, D. Xu, Y. Xie, R. Jiang, L. Deng, Cancer 14 (2015) 184.
X. Zhang, Y. Chai, X. Qin, B. Sun, LINC01225 promotes occurrence and metastasis [121] L.N. Harris, N. Ismaila, L.M. McShane, F. Andre, D.E. Collyar, A.M. Gonzalez-
of hepatocellular carcinoma in an epidermal growth factor receptor-dependent Angulo, E.H. Hammond, N.M. Kuderer, M.C. Liu, R.G. Mennel, C. Van Poznak,
pathway, Cell. Death. Dis. 7 (2016) e2130. R.C. Bast, D.F. Hayes, Use of biomarkers to guide decisions on adjuvant systemic
[96] H.M. Heneghan, N. Miller, A.J. Lowery, K.J. Sweeney, J. Newell, M.J. Kerin, therapy for women with early-stage invasive breast cancer: american society of
Circulating microRNAs as novel minimally invasive biomarkers for breast cancer, clinical oncology clinical practice guideline, J. Clin. Oncol. 34 (10) (2016)
Ann. Surg. 251 (3) (2010) 499–505. 1134–1150.
[97] W. Zhu, W. Qin, U. Atasoy, E.R. Sauter, Circulating microRNAs in breast cancer [122] W.J. Gradishar, B.O. Anderson, R. Balassanian, S.L. Blair, H.J. Burstein, A. Cyr,
and healthy subjects, BMC Res. Notes 2 (2009) 89. A.D. Elias, W.B. Farrar, A. Forero, S.H. Giordano, M.P. Goetz, L.J. Goldstein,
[98] C. Roth, B. Rack, V. Müller, W. Janni, K. Pantel, H. Schwarzenbach, Circulating S.J. Isakoff, J. Lyons, P.K. Marcom, I.A. Mayer, B. McCormick, M.S. Moran,
microRNAs as blood-based markers for patients with primary and metastatic R.M. O'Regan, S.A. Patel, L.J. Pierce, E.C. Reed, K.E. Salerno, L.S. Schwartzberg,
breast cancer, Breast Cancer Res. 12 (6) (2010) R90. A. Sitapati, K.L. Smith, M.L. Smith, H. Soliman, G. Somlo, M. Telli, J.H. Ward,
[99] A.M. McDermott, N. Miller, D. Wall, L.M. Martyn, G. Ball, K.J. Sweeney, D.A. Shead, R. Kumar, NCCN guidelines insights: breast cancer, version 1.2017, J.
M.J. Kerin, Identification and validation of oncologic miRNA biomarkers for lu- Natl. Compr. Canc. Netw. 15 (4) (2017) 433–451.
minal A-like breast cancer, PLoS One 9 (1) (2014) e87032. [123] A.S. Coates, E.P. Winer, A. Goldhirsch, R.D. Gelber, M. Gnant, M. Piccart-Gebhart,
[100] M. Chan, C.S. Liaw, S.M. Ji, H.H. Tan, C.Y. Wong, A.A. Thike, P.H. Tan, G.H. Ho, B. Thürlimann, H.J. Senn, P. Members, Tailoring therapies–improving the man-
A.S. Lee, Identification of circulating microRNA signatures for breast cancer de- agement of early breast cancer: St gallen international expert consensus on the
tection, Clin. Cancer Res. 19 (16) (2013) 4477–4487. primary therapy of early Breast cancer 2015, Ann. Oncol. 26 (8) (2015)
[101] A. de la Taille, Progensa PCA3 test for prostate cancer detection, Expert Rev. Mol. 1533–1546.
Diagn. 7 (5) (2007) 491–497. [124] E.B.C.T.C.G. (EBCTCG), Effects of chemotherapy and hormonal therapy for early
[102] K. Holm, C. Hegardt, J. Staaf, J. Vallon-Christersson, G. Jönsson, H. Olsson, breast cancer on recurrence and 15-year survival: an overview of the randomised
A. Borg, M. Ringnér, Molecular subtypes of breast cancer are associated with trials, Lancet 365 (9472) (2005) 1687–1717.
characteristic DNA methylation patterns, Breast Cancer Res. 12 (3) (2010) R36. [125] R. Peto, C. Davies, J. Godwin, R. Gray, H.C. Pan, M. Clarke, D. Cutter, S. Darby,
[103] C.G.A. Network, Comprehensive molecular portraits of human breast tumours, P. McGale, C. Taylor, Y.C. Wang, J. Bergh, A. Di Leo, K. Albain, S. Swain,
Nature 490 (7418) (2012) 61–70. M. Piccart, K. Pritchard, E. B. C. T. C. G (EBCTCG), Comparisons between different
[104] O.A. Stefansson, S. Moran, A. Gomez, S. Sayols, C. Arribas-Jorba, J. Sandoval, polychemotherapy regimens for early breast cancer: meta-analyses of long-term
H. Hilmarsdottir, E. Olafsdottir, L. Tryggvadottir, J.G. Jonasson, J. Eyfjord, outcome among 100,000 women in 123 randomised trials, Lancet 379 (9814)
M. Esteller, A DNA methylation-based definition of biologically distinct breast (2012) 432–444.
cancer subtypes, Mol. Oncol. 9 (3) (2015) 555–568. [126] B. Fisher, J. Dignam, N. Wolmark, A. DeCillis, B. Emir, D.L. Wickerham, J. Bryant,
[105] K. Holm, J. Staaf, M. Lauss, M. Aine, D. Lindgren, P.O. Bendahl, J. Vallon- N.V. Dimitrov, N. Abramson, J.N. Atkins, H. Shibata, L. Deschenes,
Christersson, R.B. Barkardottir, M. Höglund, Å. Borg, G. Jönsson, M. Ringnér, An R.G. Margolese, Tamoxifen and chemotherapy for lymph node-negative, estrogen
integrated genomics analysis of epigenetic subtypes in human breast tumors links receptor-positive breast cancer, J. Natl. Cancer Inst. 89 (22) (1997) 1673–1682.
DNA methylation patterns to chromatin states in normal mammary cells, Breast [127] J.J. Griggs, S.T. Hawley, J.J. Graff, A.S. Hamilton, R. Jagsi, N.K. Janz,
Cancer Res. 18 (1) (2016) 27. M.S. Mujahid, C.R. Friese, B. Salem, P.H. Abrahamse, S.J. Katz, Factors associated
[106] C. Stirzaker, E. Zotenko, J.Z. Song, W. Qu, S.S. Nair, W.J. Locke, A. Stone, with receipt of breast cancer adjuvant chemotherapy in a diverse population-based
N.J. Armstong, M.D. Robinson, A. Dobrovic, K.A. Avery-Kiejda, K.M. Peters, sample, J. Clin Oncol. 30 (25) (2012) 3058–3064.
J.D. French, S. Stein, D.J. Korbie, M. Trau, J.F. Forbes, R.J. Scott, M.A. Brown, [128] E. Senkus, S. Kyriakides, S. Ohno, F. Penault-Llorca, P. Poortmans, E. Rutgers,
G.D. Francis, S.J. Clark, Methylome sequencing in triple-negative breast cancer S. Zackrisson, F. Cardoso, E.G. Committee, Primary breast cancer: ESMO Clinical
reveals distinct methylation clusters with prognostic value, Nat. Commun. 6 Practice Guidelines for diagnosis, treatment and follow-up, Ann. Oncol. 26 (Suppl.
(2015) 5899. 5) (2015) v8–30.
[107] Y. Gao, A. Jones, P.A. Fasching, M. Ruebner, M.W. Beckmann, M. Widschwendter, [129] J.W. Martens, I. Nimmrich, T. Koenig, M.P. Look, N. Harbeck, F. Model, A. Kluth,
A.E. Teschendorff, The integrative epigenomic-transcriptomic landscape of ER J. Bolt-de Vries, A.M. Sieuwerts, H. Portengen, M.E. Meijer-Van Gelder,
positive breast cancer, Clin. Epigenetics 7 (2015) 126. C. Piepenbrock, A. Olek, H. Höfler, M. Kiechle, J.G. Klijn, M. Schmitt, S. Maier,
[108] S.E. Elsheikh, A.R. Green, E.A. Rakha, D.G. Powe, R.A. Ahmed, H.M. Collins, J.A. Foekens, Association of DNA methylation of phosphoserine aminotransferase
D. Soria, J.M. Garibaldi, C.E. Paish, A.A. Ammar, M.J. Grainge, G.R. Ball, with response to endocrine therapy in patients with recurrent breast cancer,
M.K. Abdelghany, L. Martinez-Pomares, D.M. Heery, I.O. Ellis, Global histone Cancer Res. 65 (10) (2005) 4101–4117.
modifications in breast cancer correlate with tumor phenotypes, prognostic fac- [130] N. Harbeck, I. Nimmrich, A. Hartmann, J.S. Ross, T. Cufer, R. Grützmann,
tors, and patient outcome, Cancer Res. 69 (9) (2009) 3802–3809. G. Kristiansen, A. Paradiso, O. Hartmann, A. Margossian, J. Martens, I. Schwope,
[109] L. Mulrane, S.F. Madden, D.J. Brennan, G. Gremel, S.F. McGee, S. McNally, A. Lukas, V. Müller, K. Milde-Langosch, J. Nährig, J. Foekens, S. Maier,
F. Martin, J.P. Crown, K. Jirström, D.G. Higgins, W.M. Gallagher, D.P. O’Connor, M. Schmitt, R. Lesche, Multicenter study using paraffin-embedded tumor tissue
miR-187 is an independent prognostic factor in breast cancer and confers in- testing PITX2 DNA methylation as a marker for outcome prediction in tamoxifen-
creased invasive potential in vitro, Clin. Cancer Res. 18 (24) (2012) 6702–6713. treated, node-negative breast cancer patients, J. Clin. Oncol. 26 (31) (2008)

33

Downloaded for Sitti Ayu Suhartina yahya Yahya (sitti.ayu.yahya@mail.ugm.ac.id) at Gadjah Mada University from ClinicalKey.com by Elsevier on October 23, 2019.
For personal use only. No other uses without permission. Copyright ©2019. Elsevier Inc. All rights reserved.
B. Pasculli et al. Seminars in Cancer Biology 51 (2018) 22–35

5036–5042. (1) (2016) 16.


[131] I. Nimmrich, A.M. Sieuwerts, M.E. Meijer-van Gelder, I. Schwope, J. Bolt-de Vries, [150] S.F. Tavazoie, C. Alarcón, T. Oskarsson, D. Padua, Q. Wang, P.D. Bos, W.L. Gerald,
N. Harbeck, T. Koenig, O. Hartmann, A. Kluth, D. Dietrich, V. Magdolen, J. Massagué, Endogenous human microRNAs that suppress breast cancer metas-
H. Portengen, M.P. Look, J.G. Klijn, R. Lesche, M. Schmitt, S. Maier, J.A. Foekens, tasis, Nature 451 (7175) (2008) 147–152.
J.W. Martens, DNA hypermethylation of PITX2 is a marker of poor prognosis in [151] R. Hoppe, J. Achinger-Kawecka, S. Winter, P. Fritz, W.Y. Lo, W. Schroth,
untreated lymph node-negative hormone receptor-positive breast cancer patients, H. Brauch, Increased expression of miR-126 and miR-10a predict prolonged re-
Breast Cancer Res. Treat. 111 (3) (2008) 429–437. lapse-free time of primary oestrogen receptor-positive breast cancer following
[132] M.H. Quentien, V. Vieira, M. Menasche, J.L. Dufier, J.P. Herman, A. Enjalbert, tamoxifen treatment, Eur. J. Cancer 49 (17) (2013) 3598–3608.
M. Abitbol, T. Brue, Truncation of PITX2 differentially affects its activity on [152] K.J. Png, N. Halberg, M. Yoshida, S.F. Tavazoie, A microRNA regulon that med-
physiological targets, J. Mol. Endocrinol. 46 (1) (2011) 9–19. iates endothelial recruitment and metastasis by cancer cells, Nature 481 (7380)
[133] R. Kavarthapu, C.H. Tsai Morris, M.L. Dufau, Prolactin induces up-regulation of its (2011) 190–194.
cognate receptor in breast cancer cells via transcriptional activation of its generic [153] Y. Khew-Goodall, G.J. Goodall, A microRNA that limits metastatic colonisation
promoter by cross-talk between ERα and STAT5, Oncotarget 5 (19) (2014) and endothelial recruitment, EMBO J. 31 (4) (2012) 786–787.
9079–9091. [154] M.P. Jansen, E.A. Reijm, A.M. Sieuwerts, K. Ruigrok-Ritstier, M.P. Look,
[134] S. Hua, C.B. Kallen, R. Dhar, M.T. Baquero, C.E. Mason, B.A. Russell, P.K. Shah, F.G. Rodríguez-González, A.A. Heine, J.W. Martens, S. Sleijfer, J.A. Foekens,
J. Liu, A. Khramtsov, M.S. Tretiakova, T.N. Krausz, O.I. Olopade, D.L. Rimm, E.M. Berns, High miR-26a and low CDC2 levels associate with decreased EZH2
K.P. White, Genomic analysis of estrogen cascade reveals histone variant H2A.Z expression and with favorable outcome on tamoxifen in metastatic breast cancer,
associated with breast cancer progression, Mol. Syst. Biol. 4 (2008) 188. Breast Cancer Res. Treat 133 (3) (2012) 937–947.
[135] A. Nagelkerke, S.J. van Kuijk, F.C. Sweep, I.D. Nagtegaal, N. Hoogerbrugge, [155] F.G. Rodríguez-González, A.M. Sieuwerts, M. Smid, M.P. Look, M.E. Meijer-van
J.W. Martens, M.A. Timmermans, H.W. van Laarhoven, J. Bussink, P.N. Span, Gelder, V. de Weerd, S. Sleijfer, J.W. Martens, J.A. Foekens, MicroRNA-30c ex-
Constitutive expression of γ-H2AX has prognostic relevance in triple negative pression level is an independent predictor of clinical benefit of endocrine therapy
breast cancer, Radiother. Oncol. 101 (1) (2011) 39–45. in advanced estrogen receptor positive breast cancer, Breast Cancer Res. Treat.
[136] H. Fiegl, S. Millinger, G. Goebel, E. Müller-Holzner, C. Marth, P.W. Laird, 127 (1) (2011) 43–51.
M. Widschwendter, Breast cancer DNA methylation profiles in cancer cells and [156] W. Shi, K. Gerster, N.M. Alajez, J. Tsang, L. Waldron, M. Pintilie, A.B. Hui,
tumor stroma: association with HER-2/neu status in primary breast cancer, Cancer J. Sykes, C. P'ng, N. Miller, D. McCready, A. Fyles, F.F. Liu, MicroRNA-301 med-
Res. 66 (1) (2006) 29–33. iates proliferation and invasion in human breast cancer, Cancer Res. 71 (8) (2011)
[137] S. Fujii, S. Yamashita, T. Yamaguchi, M. Takahashi, Y. Hozumi, T. Ushijima, 2926–2937.
H. Mukai, Pathological complete response of HER2-positive breast cancer to [157] Y. Zhu, F. Yu, Y. Jiao, J. Feng, W. Tang, H. Yao, C. Gong, J. Chen, F. Su, Y. Zhang,
trastuzumab and chemotherapy can be predicted by HSD17B4 methylation, E. Song, Reduced miR-128 in breast tumor-initiating cells induces chemother-
Oncotarget 8 (12) (2017) 19039–19048. apeutic resistance via Bmi-1 and ABCC5, Clin. Cancer Res. 17 (22) (2011)
[138] F.F. Cai, S. Chen, M.H. Wang, X.Y. Lin, L. Zhang, J.X. Zhang, L.X. Wang, J. Yang, 7105–7115.
J.H. Ding, X. Pan, Z.M. Shao, E. Biskup, Pyrosequencing quantified methylation [158] R. Zhao, J. Wu, W. Jia, C. Gong, F. Yu, Z. Ren, K. Chen, J. He, F. Su, Plasma miR-
level of BRCA1 promoter as prognostic factor for survival in breast cancer patient, 221 as a predictive biomarker for chemoresistance in breast cancer patients who
Oncotarget 7 (19) (2016) 27499–27510. previously received neoadjuvant chemotherapy, Onkologie 34 (12) (2011)
[139] M. Esteller, J.M. Silva, G. Dominguez, F. Bonilla, X. Matias-Guiu, E. Lerma, 675–680.
E. Bussaglia, J. Prat, I.C. Harkes, E.A. Repasky, E. Gabrielson, M. Schutte, [159] H. Wang, G. Tan, L. Dong, L. Cheng, K. Li, Z. Wang, H. Luo, Circulating miR-125b
S.B. Baylin, J.G. Herman, Promoter hypermethylation and BRCA1 inactivation in as a marker predicting chemoresistance in breast cancer, PLoS One 7 (4) (2012)
sporadic breast and ovarian tumors, J. Natl. Cancer Inst. 92 (7) (2000) 564–569. e34210.
[140] Y. Xu, L. Diao, Y. Chen, Y. Liu, C. Wang, T. Ouyang, J. Li, T. Wang, Z. Fan, T. Fan, [160] E.J. Jung, L. Santarpia, J. Kim, F.J. Esteva, E. Moretti, A.U. Buzdar, A. Di Leo,
B. Lin, D. Deng, S.A. Narod, Y. Xie, Promoter methylation of BRCA1 in triple- X.F. Le, R.C. Bast, S.T. Park, L. Pusztai, G.A. Calin, Plasma microRNA 210 levels
negative breast cancer predicts sensitivity to adjuvant chemotherapy, Ann. Oncol. correlate with sensitivity to trastuzumab and tumor presence in breast cancer
24 (6) (2013) 1498–1505. patients, Cancer 118 (10) (2012) 2603–2614.
[141] J.A. Ledermann, P. Harter, C. Gourley, M. Friedlander, I. Vergote, G. Rustin, [161] P. Gasparini, F. Lovat, M. Fassan, L. Casadei, L. Cascione, N.K. Jacob, S. Carasi,
C. Scott, W. Meier, R. Shapira-Frommer, T. Safra, D. Matei, A. Fielding, S. Spencer, D. Palmieri, S. Costinean, C.L. Shapiro, K. Huebner, C.M. Croce, Protective role of
P. Rowe, E. Lowe, D. Hodgson, M.A. Sovak, U. Matulonis, Overall survival in miR-155 in breast cancer through RAD51 targeting impairs homologous re-
patients with platinum-sensitive recurrent serous ovarian cancer receiving ola- combination after irradiation, Proc. Natl. Acad. Sci. U. S. A. 111 (12) (2014)
parib maintenance monotherapy: an updated analysis from a randomised, pla- 4536–4541.
cebo-controlled, double-blind, phase 2 trial, Lancet Oncol. 17 (11) (2016) [162] M.F. Godinho, A.M. Sieuwerts, M.P. Look, D. Meijer, J.A. Foekens, L.C. Dorssers,
1579–1589. T. van Agthoven, Relevance of BCAR4 in tamoxifen resistance and tumour ag-
[142] M. Robson, S.A. Im, E. Senkus, B. Xu, S.M. Domchek, N. Masuda, S. Delaloge, gressiveness of human breast cancer, Br. J. Cancer 103 (8) (2010) 1284–1291.
W. Li, N. Tung, A. Armstrong, W. Wu, C. Goessl, S. Runswick, P. Conte, Olaparib [163] P. Jonsson, C. Coarfa, F. Mesmar, T. Raz, K. Rajapakshe, J.F. Thompson,
for Metastatic Breast Cancer in Patients with a Germline BRCA Mutation, N. Engl. P.H. Gunaratne, C. Williams, Single-molecule sequencing reveals estrogen-regu-
J. Med. (2017). lated clinically relevant lncRNAs in breast cancer, Mol. Endocrinol. 29 (11) (2015)
[143] C.J. Lord, A. Ashworth, BRCAness revisited, Nat. Rev. Cancer 16 (2) (2016) 1634–1645.
110–120. [164] S.J. Shi, L.J. Wang, B. Yu, Y.H. Li, Y. Jin, X.Z. Bai, LncRNA-ATB promotes tras-
[144] J. Veeck, S. Ropero, F. Setien, E. Gonzalez-Suarez, A. Osorio, J. Benitez, tuzumab resistance and invasion-metastasis cascade in breast cancer, Oncotarget 6
J.G. Herman, M. Esteller, BRCA1 CpG island hypermethylation predicts sensitivity (13) (2015) 11652–11663.
to poly(adenosine diphosphate)-ribose polymerase inhibitors, J. Clin. Oncol. 28 [165] R.S. Redis, A.M. Sieuwerts, M.P. Look, O. Tudoran, C. Ivan, R. Spizzo, X. Zhang,
(29) (2010) e563–e564 author reply e565-6. V. de Weerd, M. Shimizu, H. Ling, R. Buiga, V. Pop, A. Irimie, R. Fodde,
[145] C. Van Poznak, M.R. Somerfield, R.C. Bast, M. Cristofanilli, M.P. Goetz, I. Bedrosian, J.W. Martens, J.A. Foekens, I. Berindan-Neagoe, G.A. Calin, CCAT2, a
A.M. Gonzalez-Angulo, D.G. Hicks, E.G. Hill, M.C. Liu, W. Lucas, I.A. Mayer, novel long non-coding RNA in breast cancer: expression study and clinical cor-
R.G. Mennel, W.F. Symmans, D.F. Hayes, L.N. Harris, Use of biomarkers to guide relations, Oncotarget 4 (10) (2013) 1748–1762.
decisions on systemic therapy for women with metastatic breast cancer: American [166] Y.M. Chen, Y. Liu, H.Y. Wei, K.Z. Lv, P. Fu, Linc-ROR induces epithelial-me-
society of clinical oncology clinical practice guideline, J. Clin. Oncol. 33 (24) senchymal transition and contributes to drug resistance and invasion of breast
(2015) 2695–2704. cancer cells, Tumour Biol. 37 (8) (2016) 10861–10870.
[146] F. Cardoso, A. Costa, E. Senkus, M. Aapro, F. André, C.H. Barrios, J. Bergh, [167] I.V. Pronina, V.I. Loginov, A.M. Burdennyy, M.V. Fridman, V.N. Senchenko,
G. Bhattacharyya, L. Biganzoli, M.J. Cardoso, L. Carey, D. Corneliussen-James, T.P. Kazubskaya, N.E. Kushlinskii, A.A. Dmitriev, E.A. Braga, DNA methylation
G. Curigliano, V. Dieras, N. El Saghir, A. Eniu, L. Fallowfield, D. Fenech, P. Francis, contributes to deregulation of 12 cancer-associated microRNAs and breast cancer
K. Gelmon, A. Gennari, N. Harbeck, C. Hudis, B. Kaufman, I. Krop, M. Mayer, progression, Gene 604 (2017) 1–8.
H. Meijer, S. Mertz, S. Ohno, O. Pagani, E. Papadopoulos, F. Peccatori, F. Pernault- [168] C. Nervi, E. De Marinis, G. Codacci-Pisanelli, Epigenetic treatment of solid tu-
Llorca, M.J. Piccart, J.Y. Pierga, H. Rugo, L. Shockney, G. Sledge, S. Swain, mours: a review of clinical trials, Clin. Epigenetics 7 (2015) 127.
C. Thomssen, A. Tutt, D. Vorobiof, B. Xu, L. Norton, E. Winer, 3rd ESO-ESMO [169] P. Chun, Histone deacetylase inhibitors in hematological malignancies and solid
International Consensus Guidelines for Advanced Breast Cancer (ABC 3), Ann. tumors, Arch. Pharm. Res. 38 (6) (2015) 933–949.
Oncol. 28 (1) (2017) 16–33. [170] M.B. Lustberg, B. Ramaswamy, Epigenetic therapy in Breast cancer, Curr. Breast
[147] R. Barbano, L.A. Muscarella, B. Pasculli, V.M. Valori, A. Fontana, M. Coco, A. la Cancer Rep. 3 (1) (2011) 34–43.
Torre, T. Balsamo, M.L. Poeta, G.F. Marangi, E. Maiello, M. Castelvetere, [171] C. Damaskos, N. Garmpis, S. Valsami, M. Kontos, E. Spartalis, T. Kalampokas,
F. Pellegrini, R. Murgo, V.M. Fazio, P. Parrella, Aberrant Keap1 methylation in E. Kalampokas, A. Athanasiou, D. Moris, A. Daskalopoulou, S. Davakis,
breast cancer and association with clinicopathological features, Epigenetics 8 (1) G. Tsourouflis, K. Kontzoglou, D. Perrea, N. Nikiteas, D. Dimitroulis, Histone
(2013) 105–112. deacetylase inhibitors: an attractive therapeutic strategy against breast cancer,
[148] Z. Zhang, H. Yamashita, T. Toyama, H. Sugiura, Y. Omoto, Y. Ando, K. Mita, Anticancer Res. 37 (1) (2017) 35–46.
M. Hamaguchi, S. Hayashi, H. Iwase, HDAC6 expression is correlated with better [172] C. Arce, C. Pérez-Plasencia, A. González-Fierro, E. de la Cruz-Hernández,
survival in breast cancer, Clin. Cancer Res. 10 (20) (2004) 6962–6968. A. Revilla-Vázquez, A. Chávez-Blanco, C. Trejo-Becerril, E. Pérez-Cárdenas,
[149] M. Braunstein, L. Liao, N. Lyttle, N. Lobo, K.J. Taylor, P.M. Krzyzanowski, L. Taja-Chayeb, E. Bargallo, P. Villarreal, T. Ramírez, T. Vela, M. Candelaria,
I. Kalatskaya, C.Q. Yao, L.D. Stein, P.C. Boutros, C.J. Twelves, R. Marcellus, M.F. Camargo, E. Robles, A. Dueñas-González, A proof-of-principle study of epi-
J.M. Bartlett, M. Spears, Downregulation of histone H2A and H2 B pathways is genetic therapy added to neoadjuvant doxorubicin cyclophosphamide for locally
associated with anthracycline sensitivity in breast cancer, Breast Cancer Res. 18 advanced breast cancer, PLoS One 1 (2006) e98.

34

Downloaded for Sitti Ayu Suhartina yahya Yahya (sitti.ayu.yahya@mail.ugm.ac.id) at Gadjah Mada University from ClinicalKey.com by Elsevier on October 23, 2019.
For personal use only. No other uses without permission. Copyright ©2019. Elsevier Inc. All rights reserved.
B. Pasculli et al. Seminars in Cancer Biology 51 (2018) 22–35

[173] T.H. Luu, R.J. Morgan, L. Leong, D. Lim, M. McNamara, J. Portnow, P. Frankel, regulating hexokinase 2 in breast cancer cells, EMBO J. 31 (8) (2012) 1985–1998.
D.D. Smith, J.H. Doroshow, C. Wong, A. Aparicio, D.R. Gandara, G. Somlo, A [187] J. Li, X. Li, X. Kong, Q. Luo, J. Zhang, L. Fang, MiRNA-26b inhibits cellular pro-
phase II trial of vorinostat (suberoylanilide hydroxamic acid) in metastatic breast liferation by targeting CDK8 in breast cancer, Int. J. Clin. Exp. Med. 7 (3) (2014)
cancer: a California Cancer Consortium study, Clin. Cancer Res. 14 (21) (2008) 558–565.
7138–7142. [188] X.Y. Li, Q.F. Luo, C.K. Wei, D.F. Li, J. Li, L. Fang, MiRNA-107 inhibits proliferation
[174] P.N. Munster, K.T. Thurn, S. Thomas, P. Raha, M. Lacevic, A. Miller, M. Melisko, and migration by targeting CDK8 in breast cancer, Int. J. Clin. Exp. Med. 7 (1)
R. Ismail-Khan, H. Rugo, M. Moasser, S.E. Minton, A phase II study of the histone (2014) 32–40.
deacetylase inhibitor vorinostat combined with tamoxifen for the treatment of [189] M.L. Si, S. Zhu, H. Wu, Z. Lu, F. Wu, Y.Y. Mo, MiR-21-mediated tumor growth,
patients with hormone therapy-resistant breast cancer, Br. J. Cancer 104 (12) Oncogene 26 (2007) 2799–2803.
(2011) 1828–1835. [190] L.B. Frankel, N.R. Christoffersen, A. Jacobsen, M. Lindow, A. Krogh, A.H. Lund,
[175] B. Ramaswamy, W. Fiskus, B. Cohen, C. Pellegrino, D.L. Hershman, E. Chuang, Programmed cell death 4 (PDCD4) is an important functional target of the
T. Luu, G. Somlo, M. Goetz, R. Swaby, C.L. Shapiro, V. Stearns, P. Christos, microRNA miR-21 in breast cancer cells, J. Biol. Chem. 283 (2) (2008)
I. Espinoza-Delgado, K. Bhalla, J.A. Sparano, Phase I-II study of vorinostat plus 1026–1033.
paclitaxel and bevacizumab in metastatic breast cancer: evidence for vorinostat- [191] S. Zhu, M.L. Si, H. Wu, Y.Y. Mo, MicroRNA-21 targets the tumor suppressor gene
induced tubulin acetylation and Hsp90 inhibition in vivo, Breast Cancer Res. tropomyosin 1 (TPM1), J. Biol. Chem. 282 (19) (2007) 14328–14336.
Treat. 132 (3) (2012) 1063–1072. [192] L. Ma, J. Teruya-Feldstein, R.A. Weinberg, TUMOR invasion and metastasis in-
[176] Y. Tu, D.L. Hershman, K. Bhalla, W. Fiskus, C.M. Pellegrino, E. Andreopoulou, itiated by microRNA-10b in breast cancer, Nature 449 (7163) (2007) 682–688.
D. Makower, K. Kalinsky, K. Fehn, S. Fineberg, A. Negassa, L.L. Montgomery, [193] L. Ma, Role of miR-10b in breast cancer metastasis, Breast Cancer Res. 12 (5)
L.S. Wiechmann, R.K. Alpaugh, M. Huang, J.A. Sparano, A phase I-II study of the (2010) 210.
histone deacetylase inhibitor vorinostat plus sequential weekly paclitaxel and [194] P.A. Gregory, A.G. Bert, E.L. Paterson, S.C. Barry, A. Tsykin, G. Farshid,
doxorubicin-cyclophosphamide in locally advanced breast cancer, Breast Cancer M.A. Vadas, Y. Khew-Goodall, G.J. Goodall, The miR-200 family and miR-205
Res. Treat. 146 (1) (2014) 145–152. regulate epithelial to mesenchymal transition by targeting ZEB1 and SIP1, Nat.
[177] W.W. Tan, J.B. Allred, A. Moreno-Aspitia, D.W. Northfelt, J.N. Ingle, M.P. Goetz, Cell Biol. 10 (5) (2008) 593–601.
E.A. Perez, Phase I study of panobinostat (LBH589) and letrozole in post- [195] S.M. Park, A.B. Gaur, E. Lengyel, M.E. Peter, The miR-200 family determines the
menopausal metastatic breast cancer patients, Clin. Breast Cancer 16 (2) (2016) epithelial phenotype of cancer cells by targeting the E-cadherin repressors ZEB1
82–86. and ZEB2, Genes Dev. 22 (7) (2008) 894–907.
[178] A. Schech, A. Kazi, S. Yu, P. Shah, G. Sabnis, Histone deacetylase inhibitor enti- [196] V.P. Tryndyak, F.A. Beland, I.P. Pogribny, E-cadherin transcriptional down-reg-
nostat inhibits tumor-initiating cells in triple-negative breast cancer cells, Mol. ulation by epigenetic and microRNA-200 family alterations is related to me-
Cancer Ther. 14 (8) (2015) 1848–1857. senchymal and drug-resistant phenotypes in human breast cancer cells, Int. J.
[179] D.A. Yardley, R.R. Ismail-Khan, B. Melichar, M. Lichinitser, P.N. Munster, Cancer 126 (11) (2010) 2575–2583.
P.M. Klein, S. Cruickshank, K.D. Miller, M.J. Lee, J.B. Trepel, Randomized phase II, [197] P.A. Gregory, C.P. Bracken, E. Smith, A.G. Bert, J.A. Wright, S. Roslan, M. Morris,
double-blind, placebo-controlled study of exemestane with or without entinostat L. Wyatt, G. Farshid, Y.Y. Lim, G.J. Lindeman, M.F. Shannon, P.A. Drew, Y. Khew-
in postmenopausal women with locally recurrent or metastatic estrogen receptor- Goodall, G.J. Goodall, An autocrine TGF-beta/ZEB/miR-200 signaling network
positive breast cancer progressing on treatment with a nonsteroidal aromatase regulates establishment and maintenance of epithelial-mesenchymal transition,
inhibitor, J. Clin. Oncol. 31 (17) (2013) 2128–2135. Mol. Biol. Cell 22 (10) (2011) 1686-98ù.
[180] R.M. Connolly, H. Li, R.C. Jankowitz, Z. Zhang, M.A. Rudek, S.C. Jeter, S.A. Slater, [198] M.V. Iorio, P. Casalini, C. Piovan, G. Di Leva, A. Merlo, T. Triulzi, S. Ménard,
P. Powers, A.C. Wolff, J.H. Fetting, A. Brufsky, R. Piekarz, N. Ahuja, P.W. Laird, C.M. Croce, E. Tagliabue, microRNA-205 regulates HER3 in human breast cancer,
H. Shen, D.J. Weisenberger, L. Cope, J.G. Herman, G. Somlo, A.A. Garcia, Cancer Res. 69 (6) (2009) 2195–2200.
P.A. Jones, S.B. Baylin, N.E. Davidson, C.A. Zahnow, V. Stearns, Combination [199] J. Chen, B.C. Wang, J.H. Tang, Clinical significance of microRNA-155 expression
epigenetic therapy in advanced breast cancer with 5-Azacitidine and entinostat: a in human breast cancer, J. Surg. Oncol. 106 (3) (2012) 260–266.
phase II national cancer institute/stand up to cancer study, Clin. Cancer Res. 23 [200] W. Kong, L. He, E.J. Richards, S. Challa, C.X. Xu, J. Permuth-Wey, J.M. Lancaster,
(11) (2017) 2691–2701. D. Coppola, T.A. Sellers, J.Y. Djeu, J.Q. Cheng, Upregulation of miRNA-155 pro-
[181] B. Pasculli, G.A. Calin, RNA interference in cancer therapy, Rev. Cell Biol. Mol. motes tumor angiogenesis by targeting VHL and is associated with poor prognosis
Med. (2015) 190–242. and triple-negative breast cancer, Oncogene 33 (6) (2014) 679–689.
[182] M. Matsui, D.R. Corey, Non-coding RNAs as drug targets, Nat. Rev. Drug Discov. [201] F.M. Buffa, C. Camps, L. Winchester, C.E. Snell, H.E. Gee, H. Sheldon, M. Taylor,
16 (3) (2017) 167–179. A.L. Harris, J. Ragoussis, microRNA-associated progression pathways and poten-
[183] S. Volinia, G.A. Calin, C.G. Liu, S. Ambs, A. Cimmino, F. Petrocca, R. Visone, tial therapeutic targets identified by integrated mRNA and microRNA expression
M. Iorio, C. Roldo, M. Ferracin, R.L. Prueitt, N. Yanaihara, G. Lanza, A. Scarpa, profiling in breast cancer, Cancer Res. 71 (17) (2011) 5635–5645.
A. Vecchione, M. Negrini, C.C. Harris, C.M. Croce, A microRNA expression sig- [202] S.T. Bailey, T. Westerling, M. Brown, Loss of estrogen-regulated microRNA ex-
nature of human solid tumors defines cancer gene targets, Proc. Natl. Acad. Sci. U. pression increases HER2 signaling and is prognostic of poor outcome in luminal
S. A. 103 (7) (2006) 2257–2261. breast cancer, Cancer Res. 75 (2) (2015) 436–445.
[184] P. Jin, T.H. Han, J. Ren, S. Saunders, E. Wang, F.M. Marincola, D.F. Stroncek, [203] P. Gasparini, L. Cascione, M. Fassan, F. Lovat, G. Guler, S. Balci, C. Irkkan,
Molecular signatures of maturing dendritic cells: implications for testing the C. Morrison, C.M. Croce, C.L. Shapiro, K. Huebner, microRNA expression profiling
quality of dendritic cell therapies, J. Transl. Med. 8 (2010) 4. identifies a four microRNA signature as a novel diagnostic and prognostic bio-
[185] S. Jiang, H.W. Zhang, M.H. Lu, X.H. He, Y. Li, H. Gu, M.F. Liu, E.D. Wang, marker in triple negative breast cancers, Oncotarget 5 (5) (2014) 1174–1184.
MicroRNA-155 functions as an OncomiR in breast cancer by targeting the sup- [204] M. Tuomarila, K. Luostari, Y. Soini, V.M. Kataja, A. Mannermaa, Overexpression of
pressor of cytokine signaling 1 gene, Cancer Res. 70 (8) (2010) 3119–3127. microRNA-200c predicts poor outcome in patients with PR-negative breast cancer,
[186] S. Jiang, L.F. Zhang, H.W. Zhang, S. Hu, M.H. Lu, S. Liang, B. Li, Y. Li, D. Li, PLoS One 9 (10) (2014) e109508.
E.D. Wang, M.F. Liu, A novel miR-155/miR-143 cascade controls glycolysis by

35

Downloaded for Sitti Ayu Suhartina yahya Yahya (sitti.ayu.yahya@mail.ugm.ac.id) at Gadjah Mada University from ClinicalKey.com by Elsevier on October 23, 2019.
For personal use only. No other uses without permission. Copyright ©2019. Elsevier Inc. All rights reserved.

Das könnte Ihnen auch gefallen