Skip to main content
Erschienen in: BMC Cancer 1/2017

Open Access 01.12.2017 | Research article

Hypermethylation of CDKN2A exon 2 in tumor, tumor-adjacent and tumor-distant tissues from breast cancer patients

verfasst von: Melanie Spitzwieser, Elisabeth Entfellner, Bettina Werner, Walter Pulverer, Georg Pfeiler, Stefan Hacker, Margit Cichna-Markl

Erschienen in: BMC Cancer | Ausgabe 1/2017

Abstract

Background

Breast carcinogenesis is a multistep process involving genetic and epigenetic changes. Tumor tissues are frequently characterized by gene-specific hypermethylation and global DNA hypomethylation. Aberrant DNA methylation levels have, however, not only been found in tumors, but also in tumor-surrounding tissue appearing histologically normal. This phenomenon is called field cancerization. Knowledge of the existence of a cancer field and its spread are of clinical relevance. If the tissue showing pre-neoplastic lesions is not removed by surgery, it may develop into invasive carcinoma.

Methods

We investigated the prevalence of gene-specific and global DNA methylation changes in tumor-adjacent and tumor-distant tissues in comparison to tumor tissues from the same breast cancer patients (n = 18) and normal breast tissues from healthy women (n = 4). Methylation-sensitive high resolution melting (MS-HRM) analysis was applied to determine methylation levels in the promoters of APC, BRCA1, CDKN2A (p16), ESR1, HER2/neu and PTEN, in CDKN2A exon 2 and in LINE-1, as indicator for the global DNA methylation extent. The methylation status of the ESR2 promoter was determined by pyrosequencing.

Results

Tumor-adjacent and tumor-distant tissues frequently showed pre-neoplastic gene-specific and global DNA methylation changes. The APC promoter (p = 0.003) and exon 2 of CDKN2A (p < 0.001) were significantly higher methylated in tumors than in normal breast tissues from healthy women. For both regions, significant differences were also found between tumor and tumor-adjacent tissues (p = 0.001 and p < 0.001, respectively) and tumor and tumor-distant tissues (p = 0.001 and p < 0.001, respectively) from breast cancer patients. In addition, tumor-adjacent (p = 0.002) and tumor-distant tissues (p = 0.005) showed significantly higher methylation levels of CDKN2A exon 2 than normal breast tissues serving as control. Significant correlations were found between the proliferative activity and the methylation status of CDKN2A exon 2 in tumor (r = −0.485, p = 0.041) and tumor-distant tissues (r = −0.498, p = 0.036).

Conclusions

From our results we can conclude that methylation changes in CDKN2A exon 2 are associated with breast carcinogenesis. Further investigations are, however, necessary to confirm that hypermethylation of CDKN2A exon 2 is associated with tumor proliferative activity.
Abkürzungen
ABCB1
ATP binding cassette subfamily B member 1
ABCC1
ATP binding cassette subfamily C member 1
ABCG2
ATP binding cassette subfamily G member 2
APC
Adenomatous polyposis coli
BRCA1
Breast cancer 1, early onset
CCND2
Cyclin D2
CDKN2A
Cyclin-dependent kinase inhibitor 2A
CpG
Cytosine-phosphatidyl-guanosine
DAPK1
Death-associated protein kinase 1
ER
Estrogen receptor
ESR1
Estrogen receptor 1, estrogen receptor α
ESR2
Estrogen receptor 2, estrogen receptor β
GSTP1
Glutathione S-transferase P1
HER2/neu
Human epidermal growth factor receptor 2
HIN-1
High in normal-1
IDC
Invasive ductal carcinoma
ILC
Invasive lobular carcinoma
LINE-1
Long interspersed element 1, retrotransposable element 1
LOD
Limit of detection
LOQ
Limit of quantification
MGMT
O6-methylguanine-DNA methyltransferase
MIB-1
Mindbomb E3 ubiquitin protein ligase 1
MS-HRM
Methylation-sensitive high resolution melting
PCR
Polymerase chain reaction
PR
Progesterone receptor
PSQ
Pyrosequencing
PTEN
Phosphatase and tensin homolog
RASSF1A
Ras association domain family member 1
S/N
Signal-to-noise ratio
Ta
Annealing temperature

Background

Breast cancer is the second most common cancer in the total world population and the most frequent cancer among women. In more developed countries, it is the second cause and in less developed regions, even the most frequent cause of cancer death in women [1]. Outcome and survival of breast cancer patients critically depend on the time of diagnosis of the malignant lesion.
Breast carcinogenesis is a multistep process involving not only genetic but also epigenetic changes, in particular aberrations in the extent of DNA methylation [24]. Tumors of many cancer types including breast cancer are frequently characterized by the co-occurrence of global DNA hypomethylation and gene-specific hypermethylation [5]. Hypermethylation of CpG dinucleotides (CpGs) in the promoter region of genes frequently results in transcriptional silencing [6] whereas global DNA hypomethylation is linked to chromosomal instability [7]. Several studies have already evaluated the applicability of gene-specific hypermethylation as diagnostic [8, 9], prognostic [1012] or predictive [13, 14] biomarker in breast cancer.
Tissues surrounding tumors frequently appear histologically normal but show pre-neoplastic lesions. This phenomenon is called “field cancerization” or “field effect” [15]. Aberrant DNA methylation levels in tumor-adjacent tissues have been reported for various cancer types including colorectal [16], prostate [17, 18] and breast cancer [1923]. Knowledge of the existence of a cancer field is of clinical relevance. If the tissue showing pre-neoplastic lesions is not removed by surgery, it may develop into invasive carcinoma.
In a previous study, we determined the promoter methylation status of six tumor suppressor genes (CCND2, DAPK1, GSTP1, HIN-1, MGMT and RASSF1A) in tumor, tumor-adjacent and tumor-distant tissues from breast cancer patients and normal breast tissues from healthy controls [24]. Promoter methylation levels of HIN-1, MGMT and RASSF1A were found to be potential biomarkers for detecting field cancerization in breast cancer patients.
In the present study, we also investigated the prevalence of pre-neoplastic DNA methylation changes in breast cancer patients. The set of breast tissue samples used previously [24] was analyzed for the methylation status of the following seven genes: adenomatous polyposis coli (APC); breast cancer 1, early onset (BRCA1); cyclin-dependent kinase inhibitor 2A (CDKN2A, p16); estrogen receptor α (ESR1); estrogen receptor β (ESR2); human epidermal growth factor receptor 2 (HER2/neu); and phosphatase and tensin homolog (PTEN). These genes were selected because they have previously been associated with breast cancer [2529]. We focused on determining the methylation status in the promoter region of these genes; in case of CDKN2A, we were, however, also interested in exon 2. Hypermethylation of CDKN2A exon 2 has previously been linked to late stage oesophageal cancer [30]. To the best of our knowledge, methylation levels for CDKN2A exon 2 in tissues from breast cancer patients have not been published so far. In addition to the gene-specific methylation status, we assessed the global DNA methylation status by using LINE-1 (long interspersed element 1; retrotransposable element 1) as indicator. Statistical analyses were carried out to test if there are significant differences in the gene-specific and/or the global DNA methylation status between tumor, tumor-adjacent and tumor-distant tissues from breast cancer patients and normal breast tissues from healthy women. In addition, we checked if the methylation status of any of the investigated regions is linked to clinicopathological parameters such as histologic type, histological grading, B classification, proliferative activity (MIB-1) and molecular subtype of the tumor.
In order to get a broader picture of the prevalence of pre-neoplastic DNA methylation changes in tumor-surrounding tissues in breast cancer patients, data that we published previously for the same set of breast tissue samples [24, 31] was included in part of the statistical tests.

Methods

Breast tissue samples

Biopsy samples were collected from 18 breast cancer patients at diagnosis of the disease (age at diagnosis: 39-76 years, mean age at diagnosis: 58 years). None of the patients had a family history of breast cancer. By ultrasound guided needle biopsy, three tissue samples were taken from each patient: the first sample directly from the tumor, the second one about 1 cm from the tumor center (“tumor-adjacent tissue”) and the third one at least 3 cm from the tumor center (“tumor-distant tissue”). Non-cancerous breast tissue samples were taken from four women (aged from 44 to 60 years; mean age: 53 years) during breast reductive surgery. From two of these women, samples of left and right breast were available. In case of CDKN2A exon 2, we additionally analyzed breast tissue samples (left and right breast) from further three healthy women. The study was approved by the Ethics Commission of the Medical University of Vienna (application number 1074/2011). All participants gave written informed consent.

Characteristics of breast cancer patients

Table 1 summarizes the characteristics of the breast cancer patients, including menopause status, histologic type, histological grading, B classification, proliferative activity (MIB-1), status of estrogen receptor (ER), progesterone receptor (PR) and human epidermal growth factor receptor 2 (HER2/neu) as well as the molecular subtype.
Table 1
Clinical and pathological characteristics of breast cancer patients
Patient
Menopause status
Histologic type
Histological grading
B classification
MIB-1 [%]
Receptor status
Molecular subtype
ER
PR
HER2/neu
1
Post
IDC
G2
B5b
10
+++
++
Luminal A
2
Post
IDC
G2
B5b
10
+++
++
Luminal A
3
Peri
IDC
G3
B5b
30
+++
Luminal A
4
Pre
IDC
G2
B5b
40
+++
++
+++
Luminal B
5
Post
IDC
G2
B5b
60
+++
++
Luminal A
6
Pre
IDC
G3
B5b
50
+++
+++
+++
Luminal B
7
Post
IDC
G3
B5b
20
pos.
pos.
Luminal A
8
Post
IDC
G2
B5b
20
+++
+++
Luminal A
9
Post
IDC
G3
B5
30
++
+++
Luminal A
10
Post
IDC
G3
B5b
20
+++
+++
+
Luminal B
11
Post
IDC
G1
B5c
20
+++
+++
+
Luminal B
12
Post
IDC
G3
B5b
70
Triple negative
13
Pre
ILC
n.s.
B5b
50
+++
++
Luminal A
14
Pre
IDC
G3
B5b
80
++
Luminal A
15
Post
IDC
G3
B5b
40
+++
++
Luminal A
16
Post
ILC
G2
B5b
30
+++
+++
Luminal A
17
Pre
Mucinous
G2
B5b
50
+++
++
+++
Luminal B
18
Post
IDC
G3
B5b
50
+++
HER2/neu
IDC invasive ductal carcinoma, ILC invasive lobular carcinoma, MIB-1 mindbomb E3 ubiquitin protein ligase 1 (proliferative activity), ER estrogen receptor, PR progesterone receptor, HER2/neu human epidermal growth factor receptor 2, n.s. not specified, + weakly positive, ++ moderately positive, +++ strongly positive, − negative

Breast cancer cell lines

Breast cancer cell lines MCF-7, MDA-MB-231 and ZR-75-1 were grown in Dulbecco’s Minimal Essential Medium (DMEM), Leibovitz’s L-15 and RPMI-1640, respectively. Culture media were supplemented with 10% fetal calf serum (PAA, Austria). Cell cultures were periodically checked for mycoplasma contamination.

DNA extraction

Genomic DNA was extracted using the QIAamp DNA Mini Kit (Qiagen, Germany) according to the manufacturer’s recommendations. The DNA concentration was determined with a Nanodrop 2000c spectrophotometer (Thermo Scientific, USA).

Bisulfite conversion

DNA extracted from biopsy samples and commercially available human control DNA (CpGenome Universal Methylated DNA, Millipore, USA and EpiTect Control DNA (human), unmethylated, Qiagen) were treated with sodium bisulfite using the EpiTect Fast Bisulfite Kit (Qiagen) following the manufacturer’s protocol.

Methylation-sensitive high resolution melting (MS-HRM)

Primer sequences for APC [32], BRCA1 [33], CDKN2A (promoter) [34] and PTEN [35] were taken from literature, those for CDKN2A (exon 2), ESR1, HER2/neu and LINE-1 were designed in-house. Nucleotide sequences were obtained from the National Center for Biotechnology Information (NCBI; [36]) database. The promoter region of the genes was identified using the Transcriptional Regulatory Element Database (TRED; [37]) or the Eukaryotic Promoter Database (EPD; [38]). Primers were designed with the Methyl Primer Express Software v1.0 (Applied Biosystems, USA). For each MS-HRM method, the annealing temperature (Ta) and the additional MgCl2 concentration were optimized in-house. Sequences of the MS-HRM primers and their optimized conditions are summarized in Table 2.
Table 2
Primer sequences and conditions for MS-HRM analysis
 
Primer sequence
Primer concentration [nM]
Additional MgCl2 concentration [mM]
Ta [°C]a
Amplicon length [bp]
No. of CpGsb
LOD/LOQ [%]
Reference
APC
f: 5′ AAGTAGTTGTGTAATTCGTTGGAT 3′
r: 5′ CACCTCCATTCTATCTCCAATA 3′
500
-
53
149
10
0.4/1.4
[32]
BRCA1
f: 5′ TTGTTGTTTAGCGGTAGTTTTTTGGTT 3′
r: 5′ AACCTATCCCCCGTCCAAAAA 3′
250
2
61
122
9
0.4/1.6
[33]
CDKN2A
f: 5′ CGGAGGAAGAAAGAGGAGGGGT 3′
r: 5′ CGCTACCTACTCTCCCCCTCT 3′
400
-
62
93
7
1.0/3.3
[34]
CDKN2A (exon 2)
f: 5′ GGCGGAGTTGTTGTTGTTTTATG 3′
r: 5′ ACAACACCACCAACGTATCCAA 3′
250
1.5
52
116
10
3.5/10.7
In-house
[NG_007485.1]
ESR1
f: 5′ TTAAAGTTGGAGGTTCGGGAGT 3′
r: 5′ CTACCCCGAAAACCTACGAATC 3′
250
-
52
112
8
0.1/0.5
In-house
[NG_008493]
HER2/neu
f: 5′ GGATTGGAGAAATTAGGGGAGT 3′
r: 5′ ACTCCGACTAAACCCGACTAAA 3′
250
2
52
137
15
0.4/1.4
In-house
[NG_007503.1]
LINE-1
f: 5′ TGTTAGATAGTGGGTGTAGGTT 3′
r: 5′ AAATACATCCGTCACCCCTTT 3′
250
-
57
139
9
11.7/22.3
In-house
[X58075.1]
PTEN
f: 5′ TCGGTTGGGTTTTTGGGTAGAGG 3′
r: 5′ CGCAAACTCTACTAAACATACCCAATAT 3′
250
-
60
156
9
0.1/0.5
[35]
f forward primer, r reverse primer, T a annealing temperature, bp base pairs, LOD limit of detection, LOQ limit of quantification
aTouchdown in 1 °C increments in cycles 1-7
bWithout CpGs in the primers
Polymerase chain reaction (PCR) and MS-HRM analysis were carried out using a Rotor-Gene Q instrument (Qiagen) and the EpiTect HRM PCR Kit (Qiagen). The reaction mixture per well had a total volume of 20 μl and included 10 μl 2× EpiTect HRM PCR Master Mix (Qiagen), varying amounts of MgCl2, forward and reverse primer, RNase-free water and 10 ng of bisulfite converted DNA. Amplification was performed under the following conditions: initial PCR activation step at 95 °C for 5 min followed by 50 cycles at 95 °C for 10 s, Ta of the respective primer set for 30 s (Table 2) and 72 °C for 10 s; denaturation step at 95 °C for 1 min followed by a hybridization step at 40 °C for 1 min. PCR was directly followed by a HRM step where the temperature was increased by 0.1 °C increments per 2 s.
MS-HRM data was evaluated with the Rotor-Gene Q Series Software 2.1.0 (Qiagen). The DNA methylation status was determined with the help of calibration curves established by analyzing DNA standards differing in their methylation status. These standards were prepared by mixing unmethylated and methylated human control DNA in different proportions. From the normalized HRM curves we calculated the average of the normalized fluorescence signal for each standard over the entire temperature. Calibration functions were established with SigmaPlot 11.0 (Systat Software Inc., USA). Limit of detection (LOD) and limit of quantification (LOQ) of the MS-HRM methods were determined by repeatedly analyzing unmethylated control DNA. We calculated the mean and the standard deviation. The LOD (signal-to-noise ratio 3) was determined by adding three times the standard deviation and the LOQ (signal-to-noise ratio 10) by adding ten times the standard deviation to the mean.

Pyrosequencing (PSQ)

Primer sequences for ESR2 were designed in-house using the PyroMark Assay Design Software 2.0.1.15 (Qiagen). The nucleotide sequence was taken from the NCBI database, the promoter region was identified using TRED. Primer sequences are listed in Table 3.
Table 3
Primer sequences and conditions for pyrosequencing analysis
Gene
Primer sequence
Primer concentration [nM]
Ta [°C]
Amplicon length [bp]
No. of CpGs analyzed
Chromosome, position of sequence to analyze, DNA strand
Reference
ESR2
f: 5′ GGAGGTTGAGAGAAATAATTGTTTTTTGA 3′
400
61
255
9
14,
[49,292-
49,369],
positive strand
in-house [NG_011535.1]
r: 5′ [Btn]ATAAACACACCCACCTTACCTTCTCTA 3′
s: 5′ GAAATAATTGTTTTTTGAAATTTG 3′
   
f forward primer, r reverse primer, s sequencing primer, [Btn] biotin, T a annealing temperature
PCR was performed in the Rotor-Gene Q instrument using the PyroMark PCR Kit (Qiagen). Each PCR reaction consisted of 12.5 μl PyroMark PCR Master Mix (2×), CoralLoad Concentrate (10×), forward and reverse primer, RNase-free water and 10 ng of bisulfite converted DNA. PCR conditions were as follows: initial activation step at 95 °C for 15 min, 50 cycles: 30 s at 94 °C, 30 s at 61 °C, 30 s at 72 °C and a final extension at 72 °C for 10 min. Pyrosequencing analyses were performed in the PyroMark Q24 Advanced instrument (Qiagen) by using PyroMark Q24 Advanced CpG Reagents (Qiagen). Sample preparation was carried out with the PyroMark Q24 Vacuum Workstation (Qiagen) as described previously [39].

Agarose gel electrophoresis

To check identity and purity, PCR products randomly selected were loaded onto a 2% agarose gel in 1× TAE buffer, stained with GelRed (Biotium, USA) and visualized with an UVT-20 M transilluminator (Herolab, Germany).

Statistical analysis

In MS-HRM analysis, bisulfite-treated DNA from breast tissue samples was analyzed in duplicates on at least two different days. In PSQ analysis, the bisulfite treated DNA was subjected to at least two PCR reactions. Each PCR product was subsequently sequenced once.
Methylation levels were treated either as categorical variables (MS-HRM methods: < LOD, < LOQ or ≥ LOQ; PSQ method: < LOQ or ≥ LOQ) or as continuous variables. If the methylation status was treated as continuous variable, a methylation status < LOD or < LOQ was substituted with a default value, namely half the LOD or half the LOQ, respectively, as proposed previously [40].
Statistical analyses were carried out with IBM SPSS Statistics Version 21.0. A p value < 0.05 (two-sided) was considered significant. Chi-squared test was used to evaluate if the methylation status is associated with any of the patients clinicopathological parameters. One-way ANOVA (analysis of variance), followed by post-hoc Tukey’s HSD (honest significant difference) test, was applied to test for significant differences in the DNA methylation status between tumor, tumor-adjacent and tumor-distant tissues as well as normal tissues from the healthy control group. Pearson’s correlation coefficient was used to assess the relationship between two continuous variables.
For principal component analysis (PCA), Qlucore’s Omics Explorer V3.2 (64bit) was used. The continuous methylation data was loaded into the software and each variable was normalized to mean 0 and variance 1. Ordering of the samples was based on the total variance captured by each component.

Results

MS-HRM and PSQ analysis

MS-HRM assays for determining the methylation status in the promoter regions of ESR1 and HER2/neu, exon 2 of CDKN2A and LINE-1 were developed and optimized in-house. The same holds for the PSQ method allowing the determination of the methylation status of the ESR2 promoter. MS-HRM assays for the promoters of APC, BRCA1, CDKN2A and PTEN were taken from literature. Primer sequences and experimental conditions are summarized in Tables 2 and 3. Figure 1 indicates the position of the CpGs targeted by the assays in relation to the respective transcription start site (TSS).
Figure 2 shows representative normalized melting curves of DNA standards obtained with the MS-HRM assays for ESR1 (a) and CDKN2A exon 2 (c). The corresponding calibration curves are shown in Fig. 2b and d, respectively. Each of the MS-HRM assays applied in the present study was validated with regard to limit of detection (LOD; S/N = 3), limit of quantification (LOQ; S/N = 10) and inter-day repeatability. LOD and LOQ were determined by repeatedly analyzing bisulfite-treated, unmethylated control DNA, the inter-day repeatability by analyzing mixtures of bisulfite-treated methylated and unmethylated control DNA on different days. With the exception of the assay for CDKN2A exon 2, the MS-HRM assays showed a PCR bias towards methylated alleles in PCR amplification, resulting in very low LODs and LOQs in the range from 0.1 to 1.0% and 0.5 to 3.3%, respectively (Table 2). LODs and LOQs of the MS-HRM assays for CDKN2A exon 2 and LINE-1 were 3.5% and 10.7% and 11.7% and 22.3%, respectively. The calibration curves (Fig. 2b and d) demonstrate the high inter-day repeatability of the assays. Figure 2e shows a representative pyrogram obtained with the PSQ assay for ESR2. By repeatedly amplifying unmethylated control DNA and subjecting the amplicons to pyrosequencing, the LOQ was determined to be 5% which is in line with the LOQ given by the manufacturer of the pyrosequencing instrument used.

DNA methylation status in breast tissues from healthy controls

DNA extracts from breast tissues of four healthy women were used to determine base levels of DNA methylation in non-cancerous breast tissues. In all breast tissue samples, BRCA1 and HER2/neu were unmethylated (methylation status < LOD), whereas ESR2 showed methylation levels between LOD and LOQ. PTEN was slightly methylated (methylation status < LOQ) in three breast tissue samples. Methylation levels slightly ≥ LOQ were only obtained for APC in one and for ESR1 in three samples. In case of CDKN2A exon 2, we analyzed breast tissue samples from seven healthy women. From five women, samples of left and right breast were available. In six healthy women, the methylation status of CDKN2A exon 2 was found to be < LOQ, in one woman it was even < LOD. The global methylation status was in the range from 88.4 to 90.7%. No differences were found between the tissue samples originating from the same women, neither in the gene-specific nor in the global methylation status.

DNA methylation status in tumor, tumor-adjacent and tumor-distant tissues from breast cancer patients

Figure 3 shows the frequency of DNA methylation in the eight gene-specific regions investigated (seven promoter regions plus exon 2 of CDKN2A) in tumor, tumor-adjacent and tumor-distant tissues from breast cancer patients. Methylation levels obtained by MS-HRM analyses (APC, BRCA1, CDKN2A, ESR1, HER2/neu and PTEN) were divided into three subclasses (< LOD, < LOQ and ≥ LOQ), those obtained by PSQ (ESR2) into two subclasses (< LOQ and ≥ LOQ). Exon 2 of CDKN2A was the only region found to be methylated (methylation status ≥ LOD) in each (18/18) of the tumors. In contrast, the promoter of CDKN2A was only methylated in 28% (5/18). In addition to CDKN2A exon 2, the promoters of APC (83%, 15/18), ESR1 (83%, 15/18) and ESR2 (71%, 12/17) were frequently methylated in tumors. Figure 3 indicates that gene-specific methylation (methylation status ≥ LOD) was almost as frequent in tumor-adjacent and tumor-distant tissues as in tumors. However, exon 2 of CDKN2A more frequently showed a methylation status ≥ LOQ in tumor (94%, 17/18) than in tumor-adjacent (50%, 9/18) and tumor-distant (56%, 10/18) tissues.
Figure 4 shows the distribution of gene-specific and global methylation levels in tumor, tumor-adjacent and tumor-distant tissues as well as in normal tissues from healthy women. With some exceptions, the promoter regions showed rather low methylation levels. The promoters of ESR1, HER2/neu and PTEN were almost unmethylated in each of the tissue samples investigated. Methylation levels ≥ 25% were only obtained for the promoters of APC in 44% (8/18), BRCA1 and CDKN2A in 11% (2/18) and ESR2 in 12% (2/17) of the tumor tissues and in addition for ESR2 in 6% (1/18) of the tumor-distant tissues. In contrast, exon 2 of CDKN2A showed a methylation status ≥ 25% in 78% (14/18) of the tumors and 11% (2/18) of the tumor-adjacent and tumor-distant tissues.
The methylation status of CDKN2A exon 2 in tumors was significantly higher than in tumor-adjacent (p < 0.001) and tumor-distant tissues (p < 0.001) from the same patients and in normal breast tissues from healthy women (p < 0.001) (Fig. 4). In addition, the methylation status in tumor-adjacent (p = 0.002) and tumor-distant tissues (p = 0.005) was significantly higher than that in normal breast tissues from healthy women. Significant differences were also found between the methylation status of APC in tumors and that in tumor-adjacent tissues (p = 0.001), tumor-distant tissues (p = 0.001) and breast tissues from healthy women (p = 0.003) (Fig. 4). In case of ESR2, tumor-adjacent (p < 0.001) and tumor-distant tissues (p < 0.001) but not the tumors showed significantly higher methylation levels than normal breast tissues from healthy women. Tumors showed a significantly lower global methylation extent than tumor-adjacent (p = 0.046) and tumor-distant tissues (p = 0.046) and breast tissues from healthy women (p < 0.001). Significant differences were also found between the global methylation extent in tumor-adjacent (p < 0.001) and tumor-distant tissues (p < 0.001) and that in normal breast tissues.
We did not find a significant difference between the methylation status in tumor-adjacent and tumor-distant tissues from breast cancer patients, neither for the promoter regions, nor for exon 2 of CDKN2A, nor for the global methylation extent.

Correlation between the methylation status in tumor, tumor-adjacent and tumor-distant tissues from breast cancer patients

For the following genes, a correlation was found between the methylation levels in tumors and those in tumor-adjacent tissues: CDKN2A (promoter) (r = 0.927, p < 0.001); BRCA1 (r = 0.878, p < 0.001); APC (r = 0.706, p = 0.001), ESR1 (r = 0.545, p = 0.019) and ESR2 (r = 0.543, p = 0.024). In addition, we found a correlation between the methylation levels in tumors and tumor-distant tissues for BRCA1 (r = 0.826, p < 0.001) and ESR1 (r = 0.555, p = 0.017). In case of BRCA1 (r = 0.994, p < 0.001), exon 2 of CDKN2A (r = 0.651, p = 0.003) and ESR2 (r = 0.580, p = 0.015), methylation levels in tumor-adjacent tissues correlated with those in tumor-distant tissues.
For the global methylation status, correlations were found between tumor and tumor-adjacent tissues (r = 0.737, p = 0.001), tumor and tumor-distant tissues (r = 0.679, p = 0.003) as well as tumor-adjacent and tumor-distant tissues (r = 0.778, p < 0.001).

Correlation between methylation levels of individual genes

Methylation levels of the BRCA1 promoter strongly correlated with those of the CDKN2A promoter in tumors (r = 0.990, p < 0.001), tumor-adjacent tissues (r = 0.920, p < 0.001) and tumor-distant tissues (r = 0.963, p < 0.001). In tumors, the methylation levels of ESR2 correlated with those of APC (r = 0.614, p = 0.009).
In addition, correlations were found with genes the methylation levels of which we had determined in previous studies [24, 31]. The methylation status of ESR2 in tumors correlated with that of ABCB1 (r = 0.614, p = 0.009) and ABCG2 (r = 0.543, p = 0.030). The methylation status of APC in tumors was found to correlate with that of MGMT (r = 0.679, p = 0.003). In tumor adjacent tissues, the promoter methylation status of ESR2 correlated with that of MGMT (r = 0.704, p = 0.002) and in tumor-distant tissues, the methylation status of APC correlated with that of ABCB1 (r = 0.756, p < 0.001).
The methylation status of CDKN2A exon2 was, however, not found to correlate with any of the other regions investigated.

Association between the DNA methylation status of individual genes with the age and clinicopathological parameters

For none of the genes investigated in the present study, we found a correlation between the age of the healthy women and the DNA methylation status in their breast tissues (based on breast tissues from four healthy women; in case of CDKN2A exon 2, breast tissues from seven women were taken into account). In tumor-distant tissues from breast cancer patients, the methylation status of ESR1 was negatively correlated with the age of the patients at diagnosis (r = −0.673, p = 0.002).
Methylation levels of CDKN2A exon 2 in tumor tissues and also in tumor-distant tissues showed a weak but significant negative correlation with the proliferative activity of the tumor (r = −0.485, p = 0.041 and r = −0.498, p = 0.036, respectively). All tumors of the luminal A and luminal B subtype showed significantly higher (p < 0.001) methylation levels than the breast tissues from healthy women. Tumor of patient 12, belonging to the triple negative subtype, showed the lowest methylation status (methylation status < LOQ) of CDKN2A exon 2 among all tumors investigated. Since our set of breast tissue samples contained only one tumor of a triple negative subtype, we determined the methylation status of CDKN2A exon 2 in MDA-MB-231, a triple negative breast cancer cell line and compared it with the methylation status of the ER positive cell lines MCF-7 (luminal A) and ZR-75-1 (luminal B). In line with the tumor tissue from patient 12, the triple negative tumor cell line MDA-MB-231 showed a methylation status < LOQ, whereas the methylation status in MCF-7 and ZR-75-1 cells was 22 and 66%, respectively. A rather low methylation status was also found for the tumor from patient 18, belonging to the HER2/neu positive subtype.

Gene-specific methylation patterns of the breast tissues analyzed

Figure 5 shows the gene-specific methylation patterns of the individual tumor, tumor-adjacent and tumor-distant tissues from breast cancer patients and normal breast tissues from healthy women. In order to get a broader picture of the prevalence of pre-neoplastic DNA methylation changes in tumor-surrounding tissues in breast cancer patients, we included methylation levels that we had obtained for the same set of breast tissue samples in previous studies (CCND2, DAPK1, GSTP1, HIN-1, MGMT and RASSF1A: [24]; ABCB1, ABCC1 and ABCG2: [31]). Gene-specific methylation patterns obtained for tumor tissues were obviously more heterogeneous with regard to both the frequency of methylation in the regions investigated and the extent of methylation than those obtained for tumor-adjacent and tumor-distant tissues from the same patients and normal breast tissues from healthy women. This observation was confirmed by principal component analysis (Fig. 6). Due to incomplete data sets, promoters of PTEN and ABCC1 and breast cancer patients 1 and 18 were excluded in this evaluation. Figure 6 also demonstrates that the gene-specific methylation patterns of tumor, tumor-adjacent and tumor-distant tissues of patient 12 and tumor of patient 14 deviated from the patterns of the respective tissue specimens of other patients. As mentioned above, tumor of patient 12 belonged to the triple negative subtype, the tumor of patient 14 was of the luminal A subtype (ER status: moderately positive, PR and HER2/neu status: negative). However, expression of the estrogen receptor was only moderate (50%) and in consequence of chemotherapeutic treatment, expression of the estrogen receptor was found to be decreased to 10%. These facts indicate that the tumor of patient 14 was not a typical representative of the luminal A subtype.
Since hypermethylation of the promoter region of genes frequently leads to transcriptional silencing, for each of the breast tissue samples we calculated the number of promoters showing a methylation status ≥ 5%. Due to incomplete data sets, promoters of PTEN and ABCC1 and breast cancer patients 1 and 18 were again excluded. Figure 7 shows the distribution of the frequency of promoters with a methylation status ≥ 5% in tumors, tumor-adjacent and tumor-distant tissues and normal breast tissues from healthy women. In tumor tissues, up to seven (out of 14) and in tumor-adjacent and tumor-distant tissues, up to six (out of 14) promoters showed a methylation status ≥ 5%, whereas in breast tissues from healthy women at most two promoters had a methylation status ≥ 5%. Figure 7 also indicates that in tumors most frequently six, in tumor-adjacent tissues two to four, in tumor-distant tissues most frequently two and in breast tissues from healthy women most frequently only one promoter had a methylation status ≥ 5%.

Discussion

The present study provides methylation levels for the promoters of APC, BRCA1, CDKN2A, ESR1, ESR2, HER2/neu and PTEN, CDKN2A exon 2 and LINE-1 in tumor, tumor-adjacent and tumor-distant tissues from 18 breast cancer patients and normal breast tissues from four healthy women. In general, methylation levels were obtained by MS-HRM analysis. In case of ESR2, however, a MS-HRM method developed in-house suffered from low amplification efficiency. However, the pyrosequencing method developed alternatively was found to be applicable to determine the methylation status of ESR2 in breast tissue samples. In contrast to pyrosequencing, which allows determining the methylation status of individual CpGs, MS-HRM assays give information about the average methylation status across all CpGs within the amplicon. However, from the shape of the melting curves, one can deduce if the region of interest is methylated homogenously or heterogeneously. In the present study, melting profiles indicating heterogeneous methylation were obtained e.g. for CDKN2A exon 2 in tumor tissues of patients 6, 14 and 18.
Among the eight gene-specific regions investigated (seven promoter regions plus exon 2 of CDKN2A), exon 2 of CDKN2A was the most frequently methylated one. In each of the tumors, it was methylated (methylation status ≥ LOD) and in 78% of the tumors, the methylation status was > 25%. In breast tumors, methylation levels were significantly higher than in tumor-adjacent and tumor-distant tissues. With levels between the LOD (3.5%) and LOQ (10.7%) of the MS-HRM assay, breast tissues from healthy women were significantly lower methylated than the tissue specimens from breast cancer patients. From these results we can conclude that methylation changes in CDKN2A exon 2 are associated with breast carcinogenesis.
Moreover, we found some interesting associations between the methylation status in CDKN2A exon 2 and clinicopathological parameters. Weak but significant negative correlations were found between the methylation status in both, tumors and tumor-distant tissues, and the proliferative activity of the tumor. In addition, all tumors of the luminal A and luminal B subtype were found to show substantially higher methylation levels than the breast tissues from healthy women, whereas in two patients the tumors of which belonged to the triple negative and the HER2/neu positive subtype, respectively, the methylation levels were similar to those in healthy women. In order to underpin our finding on the triple negative tumor, we determined the methylation status of CDKN2A exon 2 in the triple negative breast cancer cell line MDA-MB-231 and compared it with that obtained for the luminal A cell line MCF-7 and the luminal B cell line ZR-75-1. In line with our results for tumor tissues, the methylation status of CDKN2A exon 2 in the cell line MDA-MB-231 did not differ from that in normal tissues from healthy women, whereas the cell lines MCF-7 and ZR-75-1 showed substantially higher methylation levels. However, since most of the tumors analyzed in the present study belonged either to the luminal A or the luminal B subtype, our results only can give a hint that hypermethylation of CDKN2A exon 2 is associated with the molecular subtype of the tumor.
To the best of our knowledge, the methylation status of CDKN2A exon 2 has not been linked to breast cancer so far. However, in a previous study, CDKN2A exon 2 has been found to be methylated in eight out of 16 oesophageal tumors, but in none of 16 normal tissue samples [30]. The methylation frequency was higher in late stage (III and IV) than in early stage tumors. In contrast to CDKN2A exon 1, which was found to be methylated only in five tumors, methylation of CDKN2A exon 2 did not correlate with expression of CDKN2A [30].
CDKN2A has a crucial function in cell cycle control and it is known to be inactivated in various types of cancer [41]. The prevalence of aberrations in promoter methylation and their role in inactivation of CDKN2A have been investigated for various cancer types [42, 43]. Findings for breast cancer [4449] are, however, inconsistent. Some authors report higher frequency of CDKN2A promoter methylation in breast tumors [44, 45, 4749], whereas others have not found a difference between malignant and non-malignant breast tissues [46]. Surprisingly, we found the promoter of CDKN2A less frequently methylated than exon 2. Only in two (patients 12 and 14) out of 18 tumors, the promoter of CDKN2A was higher methylated than in normal breast tissues from healthy women.
Moreover, tumors of patients 12 and 14 were the only ones among our sample set in which the BRCA1 promoter was hypermethylated. BRCA1 is known to play an important role in the repair of DNA cross-links and double-strand breaks. Several papers have already described the association of BRCA1 promoter hypermethylation with sporadic breast cancer [5053]. Reported frequencies of BRCA1 promoter methylation are quite different, ranging from about 9-15% [50, 51] to 59% [53]. In addition, reports on the association of BRCA1 promoter methylation with the hormone receptor status of the tumor are inconsistent. In some studies, BRCA1 promoter methylation has been linked to the ER and PR status of the tumor [48, 54], whereas others have not found a linkage between these two features [53]. In the study of Stefansson et al. [55] and in a recent meta-analysis [56], hypermethylation of the BRCA1 promoter was significantly associated with triple negative breast cancer. By analyzing our set of tumor samples, we found an association between the BRCA1 promoter methylation status and the hormone receptor status. As mentioned above, tumor of patient 12 belonged to the triple negative subtype, whereas tumor of patient 14 was of the luminal A subtype (ER status: moderately positive, PR and HER2/neu status: negative). However, since ER expression was only moderate and significantly decreased following chemotherapeutic treatment, tumor of patient 14 cannot be considered a typical representative of the luminal A subtype.
In the present study, APC was found to be frequently methylated in tumors, tumor-adjacent and tumor-distant tissues from breast cancer patients, but not in normal breast tissues from healthy women. Tumors showed significantly higher promoter methylation levels than tumor-adjacent and tumor-distant tissues. Our results refer to promoter 1A, which has been found to be methylated in various types of cancer [57], whereas promoter 1B, located about 30 kb upstream of promoter 1A [58], has been reported to be unmethylated [57]. In the present study, the methylation frequency of the APC promoter in tumors was 83%, which is higher than the frequencies reported in literature which were in the range from 36 to 55% [35, 5963]. Most probably, this discrepancy is caused by the high sensitivity of our MS-HRM assay (LOD: 0.4%; LOQ: 1.4%). By determining the APC promoter methylation status in tumors and matched morphologically normal tissues from breast cancer patients, Van der Auwera et al. found significantly higher methylation levels in tumors which is in line with our results [63]. In contrast to other studies, in our set of samples, the APC methylation status was not associated with any of the clinicopathological parameters investigated. However, promoter methylation levels in tumors significantly correlated with those of ESR2 and MGMT.
In contrast to previous papers reporting that promoter methylation of PTEN is a frequent event in breast tumors [6467], in none of the tumors analyzed in the present study, the PTEN promoter was found to show a methylation status ≥ LOQ (although with 0.5%, the LOQ of the MS-HRM assay was very low). Results obtained by analyzing randomly selected tumor-adjacent and tumor-distant tissues of our sample set (data not shown) did not give a hint on PTEN promoter methylation in these tissue specimens.
Previous data on ESR1 methylation in breast cancer is inconsistent. In some studies, the ESR1 promoter was found to be frequently hypermethylated [54, 6870] whereas according to others, methylation of the ESR1 is a rather uncommon event in breast tumors [71]. In the tumor samples analyzed in the present study, the ESR1 promoter was frequently (83%) found to be methylated (methylation status ≥ LOD). However, the MS-HRM assay applied was very sensitive (LOD: 0.1%, LOQ: 0.5%). In tumors, the methylation levels were almost as low as in the tumor-adjacent and tumor-distant tissues from the same breast cancer patients and normal tissues from healthy controls. In contrast to the study of Wei et al. [54], we did not find a correlation between the methylation levels of ESR1 and BRCA1. However, in tumor-distant tissues, the promoter methylation status of ESR1 was negatively correlated with the age of the patients at diagnosis.
We found the ESR2 promoter frequently methylated in tumor, tumor-adjacent and tumor-distant tissues from breast cancer patients, whereas in normal breast tissues from healthy women, the methylation status was < LOQ (5%). However, methylation levels in tumor-adjacent and tumor-distant tissues, but not those in tumors were significantly higher than in normal breast tissues from healthy women. In tumor tissues, the promoter methylation levels of ESR2 correlated with those of APC, ABCB1 and ABCG2. In tumor-adjacent tissues, the methylation status correlated with that of MGMT. Rody et al. found the ESR2 promoter methylated in two-thirds of invasive breast cancers [72]. Increased methylation levels were also detected in pre-malignant lesions. In another study, ESR2 was methylated in 64% of breast tumors and 12% of adjacent normal tissues serving as control [28]. In contrast to our results, the methylation status of ESR2 was correlated with that of ESR1.
We also determined the promoter methylation status of HER2/neu, although in literature there was no hint at the involvement of DNA methylation in gene regulation. Overexpression of HER2/neu is known to stimulate cell proliferation and tumor progression [29]. In none of the breast tissues analyzed, the methylation status was ≥ 1.4%, the LOQ of the MS-HRM assay applied. According to literature, overexpression of the oncogene HER2/neu is mainly regulated by gene amplification [29, 73].
In order to assess the global methylation extent, we determined the methylation status of the repetitive element LINE-1. Global DNA hypomethylation is known to be a common feature in cancer tissues including breast cancer [74]. Among our set of tissue samples, normal breast tissues showed a significantly higher global methylation extent than the breast tissue specimens from breast cancer patients. In addition, in tumor-adjacent and tumor-distant tissues the global methylation extent was higher than in tumors. A significant difference between tumor and tumor-adjacent breast tissues was also found by Cho et al. [75]. In previous papers, the methylation status of LINE-1 in tumors was associated with the age of the patients at diagnosis [76] or clinicopathological parameters, e.g. the ER status of the tumor [74]. In the sample set analyzed in the present study, the methylation status of LINE-1 was not associated with any of the clinicopathological parameters investigated.
Our results presented here and those published recently [24, 31] demonstrate that in breast cancer patients, tumor-adjacent and tumor-distant tissues frequently show pre-neoplastic gene-specific and global DNA methylation changes. Knowledge of the presence of a cancer field is of clinical relevance, because if the tissue showing pre-neoplastic lesions is not removed by surgery, it may develop into invasive carcinoma.
Results obtained by PCA demonstrate that gene-specific methylation patterns of breast tumors are more heterogeneous than those of tumor-adjacent and tumor-distant tissues from the same breast cancer patients and normal breast tissues from healthy women. Methylation patterns of tumor, tumor-adjacent and tumor-distant tissue of a patient with triple negative breast tumor were found to deviate from those of other patients. Our finding is in line with previous studies reporting distinct methylation patterns in triple negative breast tumors [77, 78].
Hypermethylation of the gene promoter is frequently linked to transcriptional gene silencing. Among our set of breast tissue samples, in tumor tissues up to seven (out of 14), and in tumor-adjacent and tumor-distant tissues up to six (out of 14) promoters showed a methylation status ≥ 5%, whereas in normal breast tissues from healthy women at most two promoters had a methylation status ≥ 5%. Previous studies have already reported simultaneous hypermethylation of a number of tumor suppressor genes in breast tumors [12, 35, 7983], but also in matched normal tissues from breast cancer patients [12, 63].

Conclusions

Our results demonstrate that in breast cancer patients, tumor-adjacent and tumor-distant tissues frequently show pre-neoplastic gene-specific and global DNA methylation changes. Among the eight gene-specific regions investigated, exon 2 of CDKN2A was most frequently methylated in tumors, tumor-adjacent and tumor-distant tissues from breast cancer patients. In tumors, CDKN2A exon 2 was significantly higher methylated than in tumor-adjacent and tumor-distant tissues. Normal breast tissues from healthy women showed significantly lower methylation levels than the tissue specimens from the breast cancer patients. From our data we can conclude that methylation changes in exon 2 of CDKN2A are associated with breast carcinogenesis. In a previous paper, hypermethylation of CDKN2A exon 2 has been associated with oesophageal cancer. To the best of our knowledge, aberrant methylation of CDKN2A exon 2 has not been associated with breast cancer so far. Further investigations will show if hypermethylation of CDKN2A exon 2 is limited to breast and oesophageal cancer, or also occurs in other types of cancer. Our results suggest that in breast cancer, the methylation status of CDKN2A exon 2 is weakly associated with the proliferative activity and the molecular subtype of the tumor. These findings have, however, to be confirmed in further studies.

Acknowledgements

Not applicable.

Funding

This article was supported by the Open Access Publishing Fund of the University of Vienna.

Availability of data and materials

The datasets generated during and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors’ contributions

MS performed experiments, analyzed data, prepared figures and tables and performed statistical analyses. EE performed experiments and analyzed data. BW was involved in method development, WP in statistical analyses. GP and SH carried out tissue procurement and patient data collection. MCM supported conception and design and drafted the manuscript. All authors read and approved the final manuscript.

Competing interests

The authors declare that they have no competing interests.
Not applicable.
The study was approved by the Ethics Commission of the Medical University of Vienna (application number 1074/2011). All participants gave written informed consent.

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 2015;136(5):E359–86.CrossRefPubMed Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 2015;136(5):E359–86.CrossRefPubMed
2.
Zurück zum Zitat Widschwendter M, Jones PA. DNA methylation and breast carcinogenesis. Oncogene. 2002;21(35):5462–82.CrossRefPubMed Widschwendter M, Jones PA. DNA methylation and breast carcinogenesis. Oncogene. 2002;21(35):5462–82.CrossRefPubMed
3.
Zurück zum Zitat Jovanovic J, Rønneberg JA, Tost J, Kristensen V. The epigenetics of breast cancer. Mol Oncol. 2010;4(3):242–54.CrossRefPubMed Jovanovic J, Rønneberg JA, Tost J, Kristensen V. The epigenetics of breast cancer. Mol Oncol. 2010;4(3):242–54.CrossRefPubMed
4.
Zurück zum Zitat Cancer Genome Atlas Network. Comprehensive molecular portraits of human breast tumours. Nature. 2012;490(7418):61–70.CrossRef Cancer Genome Atlas Network. Comprehensive molecular portraits of human breast tumours. Nature. 2012;490(7418):61–70.CrossRef
5.
Zurück zum Zitat Ehrlich M. DNA methylation in cancer: too much, but also too little. Oncogene. 2002;21(35):5400–13.CrossRefPubMed Ehrlich M. DNA methylation in cancer: too much, but also too little. Oncogene. 2002;21(35):5400–13.CrossRefPubMed
6.
Zurück zum Zitat Esteller M. Epigenetic gene silencing in cancer: the DNA hypermethylome. Hum Mol Genet. 2007;16(1):R50–9. Esteller M. Epigenetic gene silencing in cancer: the DNA hypermethylome. Hum Mol Genet. 2007;16(1):R50–9.
7.
Zurück zum Zitat Eden A, Gaudet F, Waghmare A, Jaenisch R. Chromosomal instability and tumors promoted by DNA hypomethylation. Science. 2003;300(5618):455.CrossRefPubMed Eden A, Gaudet F, Waghmare A, Jaenisch R. Chromosomal instability and tumors promoted by DNA hypomethylation. Science. 2003;300(5618):455.CrossRefPubMed
8.
Zurück zum Zitat Brooks J, Cairns P, Zeleniuch-Jacquotte A. Promoter methylation and the detection of breast cancer. CCC. 2009;20(9):1539–50.PubMedPubMedCentral Brooks J, Cairns P, Zeleniuch-Jacquotte A. Promoter methylation and the detection of breast cancer. CCC. 2009;20(9):1539–50.PubMedPubMedCentral
10.
Zurück zum Zitat García-Giménez JL, Sanchis-Gomar F, Lippi G, Mena S, Ivars D, Gomez-Cabrera MC, et al. Epigenetic biomarkers: a new perspective in laboratory diagnostics. Clin Chim Acta. 2012;413(19-20):1576–82.CrossRefPubMed García-Giménez JL, Sanchis-Gomar F, Lippi G, Mena S, Ivars D, Gomez-Cabrera MC, et al. Epigenetic biomarkers: a new perspective in laboratory diagnostics. Clin Chim Acta. 2012;413(19-20):1576–82.CrossRefPubMed
12.
Zurück zum Zitat Li Y, Melnikov AA, Levenson V, Guerra E, Simeone P, Alberti S, et al. A seven-gene CpG-island methylation panel predicts breast cancer progression. BMC Cancer. 2015;15:417.CrossRefPubMedPubMedCentral Li Y, Melnikov AA, Levenson V, Guerra E, Simeone P, Alberti S, et al. A seven-gene CpG-island methylation panel predicts breast cancer progression. BMC Cancer. 2015;15:417.CrossRefPubMedPubMedCentral
13.
Zurück zum Zitat Duffy MJ, Napieralski R, Martens JW, Span PN, Spyratos F, Sweep FC, et al. Methylated genes as new cancer biomarkers. Eur J Cancer. 2009;45(3):335–46.CrossRefPubMed Duffy MJ, Napieralski R, Martens JW, Span PN, Spyratos F, Sweep FC, et al. Methylated genes as new cancer biomarkers. Eur J Cancer. 2009;45(3):335–46.CrossRefPubMed
14.
Zurück zum Zitat Duffy MJ. Tumor markers in clinical practice: a review focusing on common solid cancers. Med Princ Pract. 2013;22(1):4–11.CrossRefPubMed Duffy MJ. Tumor markers in clinical practice: a review focusing on common solid cancers. Med Princ Pract. 2013;22(1):4–11.CrossRefPubMed
15.
Zurück zum Zitat Chai H, Brown RE. Field effect in cancer-an update. Ann Clin Lab Sci. 2009;39(4):331–7.PubMed Chai H, Brown RE. Field effect in cancer-an update. Ann Clin Lab Sci. 2009;39(4):331–7.PubMed
16.
17.
Zurück zum Zitat Mehrotra J, Varde S, Wang H, Chiu H, Vargo J, Gray K, et al. Quantitative, spatial resolution of the epigenetic field effect in prostate cancer. Prostate. 2008;68(2):152–60.CrossRefPubMed Mehrotra J, Varde S, Wang H, Chiu H, Vargo J, Gray K, et al. Quantitative, spatial resolution of the epigenetic field effect in prostate cancer. Prostate. 2008;68(2):152–60.CrossRefPubMed
18.
Zurück zum Zitat Rybicki BA, Rundle A, Kryvenko ON, Mitrache N, Do KC, Jankowski M, et al. Methylation in benign prostate and risk of disease progression in men subsequently diagnosed with prostate cancer. Int J Cancer. 2016;138(12):2884–93.CrossRefPubMed Rybicki BA, Rundle A, Kryvenko ON, Mitrache N, Do KC, Jankowski M, et al. Methylation in benign prostate and risk of disease progression in men subsequently diagnosed with prostate cancer. Int J Cancer. 2016;138(12):2884–93.CrossRefPubMed
19.
Zurück zum Zitat Lewis CM, Cler LR, Bu DW, Zöchbauer-Müller S, Milchgrub S, Naftalis EZ, et al. Promoter hypermethylation in benign breast epithelium in relation to predicted breast cancer risk. Clin Cancer Res. 2005;11(1):166–72.PubMed Lewis CM, Cler LR, Bu DW, Zöchbauer-Müller S, Milchgrub S, Naftalis EZ, et al. Promoter hypermethylation in benign breast epithelium in relation to predicted breast cancer risk. Clin Cancer Res. 2005;11(1):166–72.PubMed
20.
Zurück zum Zitat Heaphy CM, Griffith JK, Bisoffi M. Mammary field cancerization: molecular evidence and clinical importance. Breast Cancer Res Tr. 2009;118(2):229–39.CrossRef Heaphy CM, Griffith JK, Bisoffi M. Mammary field cancerization: molecular evidence and clinical importance. Breast Cancer Res Tr. 2009;118(2):229–39.CrossRef
21.
Zurück zum Zitat Rivenbark AG, Coleman WB. Field cancerization in mammary carcinogenesis - implications for prevention and treatment of breast cancer. Exp Mol Pathol. 2012;93(3):391–8.CrossRefPubMed Rivenbark AG, Coleman WB. Field cancerization in mammary carcinogenesis - implications for prevention and treatment of breast cancer. Exp Mol Pathol. 2012;93(3):391–8.CrossRefPubMed
22.
Zurück zum Zitat Wang F, Yang Y, Fu Z, Xu N, Chen F, Yin H, et al. Differential DNA methylation status between breast carcinomatous and normal tissues. Biomed Pharmacother. 2014;68(6):699–707.CrossRefPubMed Wang F, Yang Y, Fu Z, Xu N, Chen F, Yin H, et al. Differential DNA methylation status between breast carcinomatous and normal tissues. Biomed Pharmacother. 2014;68(6):699–707.CrossRefPubMed
23.
Zurück zum Zitat Rauscher GH, Kresovich JK, Poulin M, Yan L, Macias V, Mahmoud AM, et al. Exploring DNA methylation changes in promoter, intragenic, and intergenic regions as early and late events in breast cancer formation. BMC Cancer. 2015;15:816.CrossRefPubMedPubMedCentral Rauscher GH, Kresovich JK, Poulin M, Yan L, Macias V, Mahmoud AM, et al. Exploring DNA methylation changes in promoter, intragenic, and intergenic regions as early and late events in breast cancer formation. BMC Cancer. 2015;15:816.CrossRefPubMedPubMedCentral
24.
Zurück zum Zitat Spitzwieser M, Holzweber E, Pfeiler G, Hacker S, Cichna-Markl M. Applicability of HIN-1, MGMT and RASSF1A promoter methylation as biomarkers for detecting field cancerization in breast cancer. Breast Cancer Res. 2015;17(1):125.CrossRefPubMedPubMedCentral Spitzwieser M, Holzweber E, Pfeiler G, Hacker S, Cichna-Markl M. Applicability of HIN-1, MGMT and RASSF1A promoter methylation as biomarkers for detecting field cancerization in breast cancer. Breast Cancer Res. 2015;17(1):125.CrossRefPubMedPubMedCentral
25.
Zurück zum Zitat Hoque MO, Prencipe M, Poeta ML, Barbano R, Valori VM, Copetti M, et al. Changes in CpG islands promoter methylation patterns during ductal breast carcinoma progression. Cancer Epidemiol Biomark Prev. 2009;18(10):2694–700.CrossRef Hoque MO, Prencipe M, Poeta ML, Barbano R, Valori VM, Copetti M, et al. Changes in CpG islands promoter methylation patterns during ductal breast carcinoma progression. Cancer Epidemiol Biomark Prev. 2009;18(10):2694–700.CrossRef
26.
Zurück zum Zitat Xu X, Gammon MD, Zhang Y, Cho YH, Wetmur JG, Bradshaw PT, et al. Gene promoter methylation is associated with increased mortality among women with breast cancer. Breast Cancer Res Tr. 2010;121(3):685–92.CrossRef Xu X, Gammon MD, Zhang Y, Cho YH, Wetmur JG, Bradshaw PT, et al. Gene promoter methylation is associated with increased mortality among women with breast cancer. Breast Cancer Res Tr. 2010;121(3):685–92.CrossRef
27.
Zurück zum Zitat Klajic J, Fleischer T, Dejeux E, Edvardsen H, Warnberg F, Bukholm I, et al. Quantitative DNA methylation analyses reveal stage dependent DNA methylation and association to clinico-pathological factors in breast tumors. BMC Cancer. 2013;13:456.CrossRefPubMedPubMedCentral Klajic J, Fleischer T, Dejeux E, Edvardsen H, Warnberg F, Bukholm I, et al. Quantitative DNA methylation analyses reveal stage dependent DNA methylation and association to clinico-pathological factors in breast tumors. BMC Cancer. 2013;13:456.CrossRefPubMedPubMedCentral
28.
Zurück zum Zitat Chattopadhyay S, Deo SV, Shukla NK, Husain SA. Association of promoter methylation of ERα and ERβ with sporadic breast cancer - a study from North India. Tumour Biol. 2014;35(8):7911–9.CrossRefPubMed Chattopadhyay S, Deo SV, Shukla NK, Husain SA. Association of promoter methylation of ERα and ERβ with sporadic breast cancer - a study from North India. Tumour Biol. 2014;35(8):7911–9.CrossRefPubMed
29.
Zurück zum Zitat Krishnamurti U, Silverman JF. HER2 in breast cancer: a review and update. Adv Anat Pathol. 2014;21(2):100–7.CrossRefPubMed Krishnamurti U, Silverman JF. HER2 in breast cancer: a review and update. Adv Anat Pathol. 2014;21(2):100–7.CrossRefPubMed
30.
Zurück zum Zitat Kempster S, Phillips WA, Baindur-Hudson S, Thomas RJ, Dow C, Rockman SP. Methylation of exon 2 of p16 is associated with late stage oesophageal cancer. Cancer Lett. 2000;150(1):57–62.CrossRefPubMed Kempster S, Phillips WA, Baindur-Hudson S, Thomas RJ, Dow C, Rockman SP. Methylation of exon 2 of p16 is associated with late stage oesophageal cancer. Cancer Lett. 2000;150(1):57–62.CrossRefPubMed
31.
Zurück zum Zitat Spitzwieser M, Pirker C, Koblmüller B, Pfeiler G, Hacker S, Berger W et al. Promoter methylation patterns of ABCB1, ABCC1 and ABCG2 in human cancer cell lines, multidrug-resistant cell models and tumor, tumor-adjacent and tumor-distant tissues from breast cancer patients. Oncotarget. 2016;7(45):73347–69. Spitzwieser M, Pirker C, Koblmüller B, Pfeiler G, Hacker S, Berger W et al. Promoter methylation patterns of ABCB1, ABCC1 and ABCG2 in human cancer cell lines, multidrug-resistant cell models and tumor, tumor-adjacent and tumor-distant tissues from breast cancer patients. Oncotarget. 2016;7(45):73347–69.
32.
Zurück zum Zitat Balic M, Pichler M, Strutz J, Heitzer E, Ausch C, Samonigg H, et al. High quality assessment of DNA methylation in archival tissues from colorectal cancer patients using quantitative high-resolution melting analysis. J Mol Diagn. 2009;11(2):102–8.CrossRefPubMedPubMedCentral Balic M, Pichler M, Strutz J, Heitzer E, Ausch C, Samonigg H, et al. High quality assessment of DNA methylation in archival tissues from colorectal cancer patients using quantitative high-resolution melting analysis. J Mol Diagn. 2009;11(2):102–8.CrossRefPubMedPubMedCentral
33.
Zurück zum Zitat Wong EM, Southey MC, Fox SB, Brown MA, Dowty JG, Jenkins MA, et al. Constitutional methylation of the BRCA1 promoter is specifically associated with BRCA1 mutation-associated pathology in early-onset breast cancer. Cancer Prev Res. 2011;4(1):23–33.CrossRef Wong EM, Southey MC, Fox SB, Brown MA, Dowty JG, Jenkins MA, et al. Constitutional methylation of the BRCA1 promoter is specifically associated with BRCA1 mutation-associated pathology in early-onset breast cancer. Cancer Prev Res. 2011;4(1):23–33.CrossRef
34.
Zurück zum Zitat Huang KT, Dobrovic A, Yan M, Karim RZ, Lee CS, Lakhani SR, et al. DNA methylation profiling of phyllodes and fibroadenoma tumours of the breast. Breast Cancer Res Tr. 2010;124(2):555–65.CrossRef Huang KT, Dobrovic A, Yan M, Karim RZ, Lee CS, Lakhani SR, et al. DNA methylation profiling of phyllodes and fibroadenoma tumours of the breast. Breast Cancer Res Tr. 2010;124(2):555–65.CrossRef
35.
Zurück zum Zitat Tserga A, Michalopoulos NV, Levidou G, Korkolopoulou P, Zografos G, Patsouris E, et al. Association of aberrant DNA methylation with clinicopathological features in breast cancer. Oncol Rep. 2012;27(5):1630–8.PubMed Tserga A, Michalopoulos NV, Levidou G, Korkolopoulou P, Zografos G, Patsouris E, et al. Association of aberrant DNA methylation with clinicopathological features in breast cancer. Oncol Rep. 2012;27(5):1630–8.PubMed
39.
Zurück zum Zitat Miklos W, Pelivan K, Kowol CR, Pirker C, Dornetshuber-Fleiss R, Spitzwieser M, et al. Triapine-mediated ABCB1 induction via PKC induces widespread therapy unresponsiveness but is not underlying acquired triapine resistance. Cancer Lett. 2015;361(1):112–20.CrossRefPubMed Miklos W, Pelivan K, Kowol CR, Pirker C, Dornetshuber-Fleiss R, Spitzwieser M, et al. Triapine-mediated ABCB1 induction via PKC induces widespread therapy unresponsiveness but is not underlying acquired triapine resistance. Cancer Lett. 2015;361(1):112–20.CrossRefPubMed
41.
Zurück zum Zitat Liggett Jr WH, Sidransky D. Role of the p16 tumor suppressor gene in cancer. J Clin Oncol. 1998;16(3):1197–206.CrossRefPubMed Liggett Jr WH, Sidransky D. Role of the p16 tumor suppressor gene in cancer. J Clin Oncol. 1998;16(3):1197–206.CrossRefPubMed
42.
Zurück zum Zitat Mitomi H, Fukui N, Tanaka N, Kanazawa H, Saito T, Matsuoka T, et al. Aberrant p16INK4a methylation is a frequent event in colorectal cancers: prognostic value and relation to mRNA expression and immunoreactivity. J Cancer Res Clin Oncol. 2010;136(2):323–31.CrossRefPubMed Mitomi H, Fukui N, Tanaka N, Kanazawa H, Saito T, Matsuoka T, et al. Aberrant p16INK4a methylation is a frequent event in colorectal cancers: prognostic value and relation to mRNA expression and immunoreactivity. J Cancer Res Clin Oncol. 2010;136(2):323–31.CrossRefPubMed
43.
Zurück zum Zitat Bhagat R, Kumar SS, Vaderhobli S, Premalata CS, Pallavi VR, Ramesh G, et al. Epigenetic alteration of p16 and retinoic acid receptor beta genes in the development of epithelial ovarian carcinoma. Tumour Biol. 2014;35(9):9069–78.CrossRefPubMed Bhagat R, Kumar SS, Vaderhobli S, Premalata CS, Pallavi VR, Ramesh G, et al. Epigenetic alteration of p16 and retinoic acid receptor beta genes in the development of epithelial ovarian carcinoma. Tumour Biol. 2014;35(9):9069–78.CrossRefPubMed
44.
Zurück zum Zitat Woodcock DM, Linsenmeyer ME, Doherty JP, Warren WD. DNA methylation in the promoter region of the p16 (CDKN2/MTS-1/INK4A) gene in human breast tumours. Brit J Can. 1999;79(2):251–6.CrossRef Woodcock DM, Linsenmeyer ME, Doherty JP, Warren WD. DNA methylation in the promoter region of the p16 (CDKN2/MTS-1/INK4A) gene in human breast tumours. Brit J Can. 1999;79(2):251–6.CrossRef
45.
Zurück zum Zitat Krassenstein R, Sauter E, Dulaimi E, Battagli C, Ehya H, Klein-Szanto A, et al. Detection of breast cancer in nipple aspirate fluid by CpG island hypermethylation. Clin Cancer Res. 2004;10(1):28–32.CrossRefPubMed Krassenstein R, Sauter E, Dulaimi E, Battagli C, Ehya H, Klein-Szanto A, et al. Detection of breast cancer in nipple aspirate fluid by CpG island hypermethylation. Clin Cancer Res. 2004;10(1):28–32.CrossRefPubMed
46.
Zurück zum Zitat Di Vinci A, Perdelli L, Banelli B, Salvi S, Casciano I, Gelvi I, et al. p16(INK4a) promoter methylation and protein expression in breast fibroadenoma and carcinoma. Int J Cancer. 2005;114(3):414–21.CrossRefPubMed Di Vinci A, Perdelli L, Banelli B, Salvi S, Casciano I, Gelvi I, et al. p16(INK4a) promoter methylation and protein expression in breast fibroadenoma and carcinoma. Int J Cancer. 2005;114(3):414–21.CrossRefPubMed
47.
Zurück zum Zitat Sinha S, Chunder N, Mukherjee N, Alam N, Roy A, Roychoudhury S, et al. Frequent deletion and methylation in SH3GL2 and CDKN2A loci are associated with early- and late-onset breast carcinoma. Ann Surg Oncol. 2008;15(4):1070–80.CrossRefPubMed Sinha S, Chunder N, Mukherjee N, Alam N, Roy A, Roychoudhury S, et al. Frequent deletion and methylation in SH3GL2 and CDKN2A loci are associated with early- and late-onset breast carcinoma. Ann Surg Oncol. 2008;15(4):1070–80.CrossRefPubMed
48.
Zurück zum Zitat Tao MH, Shields PG, Nie J, Millen A, Ambrosone CB, Edge SB, et al. DNA hypermethylation and clinicopathological features in breast cancer: the western New York exposures and breast cancer (WEB) study. Breast Cancer Res Tr. 2009;114(3):559–68.CrossRef Tao MH, Shields PG, Nie J, Millen A, Ambrosone CB, Edge SB, et al. DNA hypermethylation and clinicopathological features in breast cancer: the western New York exposures and breast cancer (WEB) study. Breast Cancer Res Tr. 2009;114(3):559–68.CrossRef
49.
Zurück zum Zitat Askari M, Sobti RC, Nikbakht M, Sharma SC. Promoter hypermethylation of tumour suppressor genes (p14/ARF and p16/INK4a): case-control study in north Indian population. Mol Biol Rep. 2013;40(8):4921–8.CrossRefPubMed Askari M, Sobti RC, Nikbakht M, Sharma SC. Promoter hypermethylation of tumour suppressor genes (p14/ARF and p16/INK4a): case-control study in north Indian population. Mol Biol Rep. 2013;40(8):4921–8.CrossRefPubMed
50.
Zurück zum Zitat Esteller M, Silva JM, Dominguez G, Bonilla F, Matias-Guiu X, Lerma E, et al. Promoter hypermethylation and BRCA1 inactivation in sporadic breast and ovarian tumors. J Natl Cancer Inst. 2000;92(7):564–9.CrossRefPubMed Esteller M, Silva JM, Dominguez G, Bonilla F, Matias-Guiu X, Lerma E, et al. Promoter hypermethylation and BRCA1 inactivation in sporadic breast and ovarian tumors. J Natl Cancer Inst. 2000;92(7):564–9.CrossRefPubMed
51.
Zurück zum Zitat Birgisdottir V, Stefansson OA, Bodvarsdottir SK, Hilmarsdottir H, Jonasson JG, Eyfjord JE. Epigenetic silencing and deletion of the BRCA1 gene in sporadic breast cancer. Breast Cancer Res. 2006;8(4):R38.CrossRefPubMedPubMedCentral Birgisdottir V, Stefansson OA, Bodvarsdottir SK, Hilmarsdottir H, Jonasson JG, Eyfjord JE. Epigenetic silencing and deletion of the BRCA1 gene in sporadic breast cancer. Breast Cancer Res. 2006;8(4):R38.CrossRefPubMedPubMedCentral
52.
Zurück zum Zitat Suijkerbuijk KP, Fackler MJ, Sukumar S, van Gils CH, van Laar T, van der Wall E, et al. Methylation is less abundant in BRCA1-associated compared with sporadic breast cancer. Ann Oncol. 2008;19(11):1870–4.CrossRefPubMedPubMedCentral Suijkerbuijk KP, Fackler MJ, Sukumar S, van Gils CH, van Laar T, van der Wall E, et al. Methylation is less abundant in BRCA1-associated compared with sporadic breast cancer. Ann Oncol. 2008;19(11):1870–4.CrossRefPubMedPubMedCentral
53.
Zurück zum Zitat Xu X, Gammon MD, Zhang Y, Bestor TH, Zeisel SH, Wetmur JG, et al. BRCA1 promoter methylation is associated with increased mortality among women with breast cancer. Breast Cancer Res Tr. 2009;115(2):397–404.CrossRef Xu X, Gammon MD, Zhang Y, Bestor TH, Zeisel SH, Wetmur JG, et al. BRCA1 promoter methylation is associated with increased mortality among women with breast cancer. Breast Cancer Res Tr. 2009;115(2):397–404.CrossRef
54.
Zurück zum Zitat Wei M, Xu J, Dignam J, Nanda R, Sveen L, Fackenthal J et al. Estrogen receptor alpha, BRCA1, and FANCF promoter methylation occur in distinct subsets of sporadic breast cancers. Breast Cancer Res Tr. 2008;111(1):113-120. Wei M, Xu J, Dignam J, Nanda R, Sveen L, Fackenthal J et al. Estrogen receptor alpha, BRCA1, and FANCF promoter methylation occur in distinct subsets of sporadic breast cancers. Breast Cancer Res Tr. 2008;111(1):113-120.
55.
Zurück zum Zitat Stefansson OA, Jonasson JG, Olafsdottir K, Hilmarsdottir H, Olafsdottir G, Esteller M, et al. CpG island hypermethylation of BRCA1 and loss of pRb as co-occurring events in basal/triple-negative breast cancer. Epigenetics. 2011;6(5):638–49.CrossRefPubMedPubMedCentral Stefansson OA, Jonasson JG, Olafsdottir K, Hilmarsdottir H, Olafsdottir G, Esteller M, et al. CpG island hypermethylation of BRCA1 and loss of pRb as co-occurring events in basal/triple-negative breast cancer. Epigenetics. 2011;6(5):638–49.CrossRefPubMedPubMedCentral
56.
Zurück zum Zitat Guo T, Ren Y, Wang B, Huang Y, Jia S, Tang W, et al. Promoter methylation of BRCA1 is associated with estrogen, progesterone and human epidermal growth factor receptor-negative tumors and the prognosis of breast cancer: a meta-analysis. Mol Clin Oncol. 2015;3(6):1353–60.PubMedPubMedCentral Guo T, Ren Y, Wang B, Huang Y, Jia S, Tang W, et al. Promoter methylation of BRCA1 is associated with estrogen, progesterone and human epidermal growth factor receptor-negative tumors and the prognosis of breast cancer: a meta-analysis. Mol Clin Oncol. 2015;3(6):1353–60.PubMedPubMedCentral
57.
Zurück zum Zitat Esteller M, Sparks A, Toyota M, Sanchez-Cespedes M, Capella G, Peinado MA, et al. Analysis of adenomatous polyposis coli promoter hypermethylation in human cancer. Cancer Res. 2000;60(16):4366–71.PubMed Esteller M, Sparks A, Toyota M, Sanchez-Cespedes M, Capella G, Peinado MA, et al. Analysis of adenomatous polyposis coli promoter hypermethylation in human cancer. Cancer Res. 2000;60(16):4366–71.PubMed
58.
Zurück zum Zitat Yamaguchi K, Nagayama S, Shimizu E, Komura M, Yamaguchi R, Shibuya T, et al. Reduced expression of APC-1B but not APC-1A by the deletion of promoter 1B is responsible for familial adenomatous polyposis. Sci Rep. 2016;6:26011.CrossRefPubMedPubMedCentral Yamaguchi K, Nagayama S, Shimizu E, Komura M, Yamaguchi R, Shibuya T, et al. Reduced expression of APC-1B but not APC-1A by the deletion of promoter 1B is responsible for familial adenomatous polyposis. Sci Rep. 2016;6:26011.CrossRefPubMedPubMedCentral
59.
Zurück zum Zitat Virmani AK, Rathi A, Sathyanarayana UG, Padar A, Huang CX, Cunnigham HT, et al. Aberrant methylation of the adenomatous polyposis coli (APC) gene promoter 1A in breast and lung carcinomas. Clin Cancer Res. 2001;7(7):1998–2004.PubMed Virmani AK, Rathi A, Sathyanarayana UG, Padar A, Huang CX, Cunnigham HT, et al. Aberrant methylation of the adenomatous polyposis coli (APC) gene promoter 1A in breast and lung carcinomas. Clin Cancer Res. 2001;7(7):1998–2004.PubMed
60.
Zurück zum Zitat Jin Z, Tamura G, Tsuchiya T, Sakata K, Kashiwaba M, Osakabe M, et al. Adenomatous polyposis coli (APC) gene promoter hypermethylation in primary breast cancers. Brit J Can. 2001;85(1):69–73.CrossRef Jin Z, Tamura G, Tsuchiya T, Sakata K, Kashiwaba M, Osakabe M, et al. Adenomatous polyposis coli (APC) gene promoter hypermethylation in primary breast cancers. Brit J Can. 2001;85(1):69–73.CrossRef
61.
Zurück zum Zitat Sarrío D, Moreno-Bueno G, Hardisson D, Sánchez-Estévez C, Guo M, Herman JG, et al. Epigenetic and genetic alterations of APC and CDH1 genes in lobular breast cancer: relationships with abnormal E-cadherin and catenin expression and microsatellite instability. Int J Cancer. 2003;106(2):208–15.CrossRefPubMed Sarrío D, Moreno-Bueno G, Hardisson D, Sánchez-Estévez C, Guo M, Herman JG, et al. Epigenetic and genetic alterations of APC and CDH1 genes in lobular breast cancer: relationships with abnormal E-cadherin and catenin expression and microsatellite instability. Int J Cancer. 2003;106(2):208–15.CrossRefPubMed
62.
Zurück zum Zitat Dulaimi E, Hillinck J. Ibanez de Caceres I, al-Saleem T, cairns P. Tumor suppressor gene promoter hypermethylation in serum of breast cancer patients. Clin Cancer Res. 2004;10(18):6189–93.CrossRefPubMed Dulaimi E, Hillinck J. Ibanez de Caceres I, al-Saleem T, cairns P. Tumor suppressor gene promoter hypermethylation in serum of breast cancer patients. Clin Cancer Res. 2004;10(18):6189–93.CrossRefPubMed
63.
Zurück zum Zitat Van der Auwera I, Bovie C, Svensson C, Trinh XB, Limame R, van Dam P, et al. Quantitative methylation profiling in tumor and matched morphologically normal tissues from breast cancer patients. BMC Cancer. 2010;10:97.CrossRefPubMedPubMedCentral Van der Auwera I, Bovie C, Svensson C, Trinh XB, Limame R, van Dam P, et al. Quantitative methylation profiling in tumor and matched morphologically normal tissues from breast cancer patients. BMC Cancer. 2010;10:97.CrossRefPubMedPubMedCentral
64.
Zurück zum Zitat García JM, Silva J, Peña C, García V, Rodríguez R, Cruz MA, et al. Promoter methylation of the PTEN gene is a common molecular change in breast cancer. Gene Chromosome Canc. 2004;41(2):117–24.CrossRef García JM, Silva J, Peña C, García V, Rodríguez R, Cruz MA, et al. Promoter methylation of the PTEN gene is a common molecular change in breast cancer. Gene Chromosome Canc. 2004;41(2):117–24.CrossRef
65.
Zurück zum Zitat Khan S, Kumagai T, Vora J, Bose N, Sehgal I, Koeffler PH, et al. PTEN promoter is methylated in a proportion of invasive breast cancers. Int J Cancer. 2004;112(3):407–10.CrossRefPubMed Khan S, Kumagai T, Vora J, Bose N, Sehgal I, Koeffler PH, et al. PTEN promoter is methylated in a proportion of invasive breast cancers. Int J Cancer. 2004;112(3):407–10.CrossRefPubMed
66.
Zurück zum Zitat Sadeq V, Isar N, Manoochehr T. Association of sporadic breast cancer with PTEN/MMAC1/TEP1 promoter hypermethylation. Med Oncol. 2011;28(2):420–3.CrossRefPubMed Sadeq V, Isar N, Manoochehr T. Association of sporadic breast cancer with PTEN/MMAC1/TEP1 promoter hypermethylation. Med Oncol. 2011;28(2):420–3.CrossRefPubMed
67.
Zurück zum Zitat Zhang HY, Liang F, Jia ZL, Song ST, Jiang ZF. PTEN mutation, methylation and expression in breast cancer patients. Oncol Lett. 2013;6(1):161–8.PubMedPubMedCentral Zhang HY, Liang F, Jia ZL, Song ST, Jiang ZF. PTEN mutation, methylation and expression in breast cancer patients. Oncol Lett. 2013;6(1):161–8.PubMedPubMedCentral
68.
Zurück zum Zitat Ramos EA, Camargo AA, Braun K, Slowik R, Cavalli IJ, Ribeiro EM, et al. Simultaneous CXCL12 and ESR1 CpG island hypermethylation correlates with poor prognosis in sporadic breast cancer. BMC Cancer. 2010;10:23.CrossRefPubMedPubMedCentral Ramos EA, Camargo AA, Braun K, Slowik R, Cavalli IJ, Ribeiro EM, et al. Simultaneous CXCL12 and ESR1 CpG island hypermethylation correlates with poor prognosis in sporadic breast cancer. BMC Cancer. 2010;10:23.CrossRefPubMedPubMedCentral
69.
Zurück zum Zitat Prabhu JS, Wahi K, Korlimarla A, Correa M, Manjunath S, Raman N, et al. The epigenetic silencing of the estrogen receptor (ER) by hypermethylation of the ESR1 promoter is seen predominantly in triple-negative breast cancers in Indian women. Tumour Biol. 2012;33(2):315–23.CrossRefPubMed Prabhu JS, Wahi K, Korlimarla A, Correa M, Manjunath S, Raman N, et al. The epigenetic silencing of the estrogen receptor (ER) by hypermethylation of the ESR1 promoter is seen predominantly in triple-negative breast cancers in Indian women. Tumour Biol. 2012;33(2):315–23.CrossRefPubMed
70.
Zurück zum Zitat Martínez-Galán J, Torres-Torres B, Núñez MI, López-Peñalver J, Del Moral R, Ruiz De Almodóvar JM, et al. ESR1 gene promoter region methylation in free circulating DNA and its correlation with estrogen receptor protein expression in tumor tissue in breast cancer patients. BMC Cancer. 2014;14:59.CrossRefPubMedPubMedCentral Martínez-Galán J, Torres-Torres B, Núñez MI, López-Peñalver J, Del Moral R, Ruiz De Almodóvar JM, et al. ESR1 gene promoter region methylation in free circulating DNA and its correlation with estrogen receptor protein expression in tumor tissue in breast cancer patients. BMC Cancer. 2014;14:59.CrossRefPubMedPubMedCentral
71.
Zurück zum Zitat Medina-Jaime AD, Reyes-Vargas F, Martinez-Gaytan V, Zambrano-Galvan G, Portillo-Delcampo E, Burciaga-Nava JA, et al. ESR1 and PGR gene promoter methylation and correlations with estrogen and progesterone receptors in ductal and lobular breast cancer. Asian Pac J Cancer Prev. 2014;15(7):3041–4.CrossRefPubMed Medina-Jaime AD, Reyes-Vargas F, Martinez-Gaytan V, Zambrano-Galvan G, Portillo-Delcampo E, Burciaga-Nava JA, et al. ESR1 and PGR gene promoter methylation and correlations with estrogen and progesterone receptors in ductal and lobular breast cancer. Asian Pac J Cancer Prev. 2014;15(7):3041–4.CrossRefPubMed
72.
Zurück zum Zitat Rody A, Holtrich U, Solbach C, Kourtis K, von Minckwitz G, Engels K, et al. Methylation of estrogen receptor beta promoter correlates with loss of ER-beta expression in mammary carcinoma and is an early indication marker in premalignant lesions. Endocr-Relat Cancer. 2005;12(4):903–16.CrossRefPubMed Rody A, Holtrich U, Solbach C, Kourtis K, von Minckwitz G, Engels K, et al. Methylation of estrogen receptor beta promoter correlates with loss of ER-beta expression in mammary carcinoma and is an early indication marker in premalignant lesions. Endocr-Relat Cancer. 2005;12(4):903–16.CrossRefPubMed
73.
Zurück zum Zitat Terada K, Okochi-Takada E, Akashi-Tanaka S, Miyamoto K, Taniyama K, Tsuda H, et al. Association between frequent CpG island methylation and HER2 amplification in human breast cancers. Carcinogenesis. 2009;30(3):466–71.CrossRefPubMed Terada K, Okochi-Takada E, Akashi-Tanaka S, Miyamoto K, Taniyama K, Tsuda H, et al. Association between frequent CpG island methylation and HER2 amplification in human breast cancers. Carcinogenesis. 2009;30(3):466–71.CrossRefPubMed
74.
Zurück zum Zitat Park SY, Seo AN, Jung HY, Gwak JM, Jung N, Cho NY, et al. Alu and LINE-1 hypomethylation is associated with HER2 enriched subtype of breast cancer. PLoS One. 2014;9(6):e100429.CrossRefPubMedPubMedCentral Park SY, Seo AN, Jung HY, Gwak JM, Jung N, Cho NY, et al. Alu and LINE-1 hypomethylation is associated with HER2 enriched subtype of breast cancer. PLoS One. 2014;9(6):e100429.CrossRefPubMedPubMedCentral
75.
Zurück zum Zitat Cho YH, Yazici H, Wu HC, Terry MB, Gonzalez K, Qu M, et al. Aberrant promoter hypermethylation and genomic hypomethylation in tumor, adjacent normal tissues and blood from breast cancer patients. Anticancer Res. 2010;30(7):2489–96.PubMedPubMedCentral Cho YH, Yazici H, Wu HC, Terry MB, Gonzalez K, Qu M, et al. Aberrant promoter hypermethylation and genomic hypomethylation in tumor, adjacent normal tissues and blood from breast cancer patients. Anticancer Res. 2010;30(7):2489–96.PubMedPubMedCentral
76.
Zurück zum Zitat van Hoesel AQ, van de Velde CJ, Kuppen PJ, Liefers GJ, Putter H, Sato Y, et al. Hypomethylation of LINE-1 in primary tumor has poor prognosis in young breast cancer patients: a retrospective cohort study. Breast Cancer Res Tr. 2012;134(3):1103–14.CrossRef van Hoesel AQ, van de Velde CJ, Kuppen PJ, Liefers GJ, Putter H, Sato Y, et al. Hypomethylation of LINE-1 in primary tumor has poor prognosis in young breast cancer patients: a retrospective cohort study. Breast Cancer Res Tr. 2012;134(3):1103–14.CrossRef
77.
Zurück zum Zitat Branham MT, Marzese DM, Laurito SR, Gago FE, Orozco JI, Tello OM, et al. Methylation profile of triple-negative breast carcinomas. Oncogene. 2012;1:e17.CrossRef Branham MT, Marzese DM, Laurito SR, Gago FE, Orozco JI, Tello OM, et al. Methylation profile of triple-negative breast carcinomas. Oncogene. 2012;1:e17.CrossRef
78.
Zurück zum Zitat Stirzaker C, Zotenko E, Song JZ, Qu W, Nair SS, Locke WJ, et al. Methylome sequencing in triple-negative breast cancer reveals distinct methylation clusters with prognostic value. Nat Commun. 2015;6:5899.CrossRefPubMed Stirzaker C, Zotenko E, Song JZ, Qu W, Nair SS, Locke WJ, et al. Methylome sequencing in triple-negative breast cancer reveals distinct methylation clusters with prognostic value. Nat Commun. 2015;6:5899.CrossRefPubMed
79.
Zurück zum Zitat Fackler MJ, McVeigh M, Evron E, Garrett E, Mehrotra J, Polyak K, et al. DNA methylation of RASSF1A, HIN-1, RAR-beta, Cyclin D2 and Twist in in situ and invasive lobular breast carcinoma. Int J Cancer. 2003;107(6):970–5.CrossRefPubMed Fackler MJ, McVeigh M, Evron E, Garrett E, Mehrotra J, Polyak K, et al. DNA methylation of RASSF1A, HIN-1, RAR-beta, Cyclin D2 and Twist in in situ and invasive lobular breast carcinoma. Int J Cancer. 2003;107(6):970–5.CrossRefPubMed
80.
Zurück zum Zitat Bae YK, Brown A, Garrett E, Bornman D, Fackler MJ, Sukumar S, et al. Hypermethylation in histologically distinct classes of breast cancer. Clin Cancer Res. 2004;10(18):5998–6005.CrossRefPubMed Bae YK, Brown A, Garrett E, Bornman D, Fackler MJ, Sukumar S, et al. Hypermethylation in histologically distinct classes of breast cancer. Clin Cancer Res. 2004;10(18):5998–6005.CrossRefPubMed
81.
Zurück zum Zitat Holm K, Hegardt C, Staaf J, Vallon-Christersson J, Jönsson G, Olsson H, et al. Molecular subtypes of breast cancer are associated with characteristic DNA methylation patterns. Breast Cancer Res. 2010;12(3):R36.CrossRefPubMedPubMedCentral Holm K, Hegardt C, Staaf J, Vallon-Christersson J, Jönsson G, Olsson H, et al. Molecular subtypes of breast cancer are associated with characteristic DNA methylation patterns. Breast Cancer Res. 2010;12(3):R36.CrossRefPubMedPubMedCentral
82.
Zurück zum Zitat Muggerud AA, Rønneberg JA, Wärnberg F, Botling J, Busato F, Jovanovic J, et al. Frequent aberrant DNA methylation of ABCB1, FOXC1, PPP2R2B and PTEN in ductal carcinoma in situ and early invasive breast cancer. Breast Cancer Res. 2010;12(1):R3.CrossRefPubMedPubMedCentral Muggerud AA, Rønneberg JA, Wärnberg F, Botling J, Busato F, Jovanovic J, et al. Frequent aberrant DNA methylation of ABCB1, FOXC1, PPP2R2B and PTEN in ductal carcinoma in situ and early invasive breast cancer. Breast Cancer Res. 2010;12(1):R3.CrossRefPubMedPubMedCentral
83.
Zurück zum Zitat Cho YH, Shen J, Gammon MD, Zhang YJ, Wang Q, Gonzalez K, et al. Prognostic significance of gene-specific promoter hypermethylation in breast cancer patients. Breast Cancer Res Tr. 2012;131(1):197–205.CrossRef Cho YH, Shen J, Gammon MD, Zhang YJ, Wang Q, Gonzalez K, et al. Prognostic significance of gene-specific promoter hypermethylation in breast cancer patients. Breast Cancer Res Tr. 2012;131(1):197–205.CrossRef
Metadaten
Titel
Hypermethylation of CDKN2A exon 2 in tumor, tumor-adjacent and tumor-distant tissues from breast cancer patients
verfasst von
Melanie Spitzwieser
Elisabeth Entfellner
Bettina Werner
Walter Pulverer
Georg Pfeiler
Stefan Hacker
Margit Cichna-Markl
Publikationsdatum
01.12.2017
Verlag
BioMed Central
Erschienen in
BMC Cancer / Ausgabe 1/2017
Elektronische ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-017-3244-2

Weitere Artikel der Ausgabe 1/2017

BMC Cancer 1/2017 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.