Submit Your Article CMED MEACR meeting
An official publication of the Middle-Eastern Association for Cancer Research
Clinical Cancer Investigation Journal
ISSN Print: 2278-1668, Online: 2278-0513
ARTICLE
Year: 2015   |   Volume: 4   |   Issue: 2   |   Page: 120-128     View issue
Serum/plasma DNA methylation pattern and early detection of breast cancer
Arootin Gharibiyan, Seyed Hashemi, Mahdie Hadi, Hamed Abdolghafoorian, Ali Ardekani

Breast cancer is the most common cancer among women. With its fatality rate reduced significantly if diagnosed early, developing cost-effective, noninvasive methods of early detection is highly investigated. Currently, mammography with magnetic resonance imaging is considered the optimal method of early detection in women who are at a significantly raised risk of developing breast cancer. Due to environmental effects and life-style changes in recent years, elevation of the risk of cancer incidents in lower risk populations is observed and therefore, the development of a relatively easy-performed and low-cost method for early detection of cancer in general and breast cancer in particular is needed. Serum-based analysis techniques have been quite popular subject of research recently as they can be performed with low technical knowledge, become automated and are cheap. In the present article, we have reviewed the literature related to the use of DNA methylation-detection based techniques for diagnosis of early-stage breast cancer using serum or plasma circulating tumor DNA and their power as a future biomarker. A reference to all genes that is reported to be differentially methylated in breast cancer accompanies the article.

Breast cancer, circulating tumor DNA markers, early detection, methylation pattern

INTRODUCTION

The most frequently diagnosed cancer among women and the second most common cancer overall (except for skin cancers) is breast cancer. According to estimates of the American Cancer Society in 2014, about 200,000 new cases of invasive breast cancer in women will be diagnosed, and about 40,000 of them will die. [1] In the Middle East, according to the estimate of the Iranian Ministry of Health and Medical Education in year 2011, breast cancer ranked first among female cancers, comprising 21.4% of all types of cancer. In developing countries, breast cancer affects women about one decade younger than it does in developed countries; the same situation is reported in Iran. [2],[3]

Identifying risk factors for breast cancer could help choosing screening decisions. It has been revealed that two main risk factors for breast cancer are: Extremely dense breast tissue and patient′s first-degree relatives with breast cancer, especially if diagnosed at younger age. Each category was associated with more than two-fold increased risk of breast cancer in women age 40-49. Many more risk factors such as race/ethnicity, physical activity, alcohol consumption, cigarette smoking and oral contraceptive pills is investigated, but none of them has been confirmed. [4],[5]

It is notable that only 5-12% of patients with breast cancer are in the high risk group carrying inherited factors; [1] thus other risk factors collectively constitute the cause for the great majority of breast cancer emergence. The effects of these factors are not detectable at very early stages by genetic screening.

Breast cancers are broadly categorized into in situ carcinoma and invasive (infiltrating) carcinoma. In situ breast carcinoma is sub-classified and distinguished on the basis of growth patterns and cytological features as either lobular or ductal carcinoma in situ (LCIS and DCIS respectively). DCIS is considerably more common than LCIS. [6] Breast cancer at the gene expression level is categorized into five molecular subtypes: Luminal A, luminal B, HER2+, basal-like, and normal-like each with distinct gene expression patterns. Different subtypes of breast cancer tumors have shown a significant difference in overall survival rates. [7],[8],[9] Immunohistochemistry and gene expression pattern (Mammaprint and Oncotype DX) are usually the methods of choice to detect these prognostic markers. Accurate classification of breast cancer subtypes coupled with early detection is critical to effective cancer treatments. However, the heterogeneity of different tumors and relatively high prices for these tests can be restrictive, making blood-based techniques appealing clinically.

BREAST CANCER DETECTION METHODS

Disease screening consists of actions taken to find a disease or condition at very early states while the patient has no symptoms or complaints. The recommended available screening methods for early detection of breast cancer are mammography, clinical breast examination (CBE) and breast self-examination (BSE). [2] Currently, mammography with magnetic resonance imaging (MRI) is recommended as the optimal and the most accurate method of early detection of breast cancer in women with significantly raised risk of breast cancer. [10],[11] Any woman of 30 years of age or older, with a suspicious breast symptom or sign should undergo further investigations including CBE and diagnostic mammogram. [12]

Mammography screening has a sensitivity (77-95%) and specificity (94-97%), respectively. [13] Sensitivity of mammography in women at increased risk of breast cancer is considerably lower (33-56%). MRI is a highly sensitive substitute screening method to detect breast tumors not seen in mammography. [10],[11],[14] Since mammography is thought to be more sensitive than MRI in detecting microcalcifications in early stages of breast cancer, they can be considered as complementary. Positive finding on either screening mammography or MRI mandates further diagnostic evaluations. [10],[11] There is little evidence to support for using BSE or CBE as useful screening tools. [5],[13]

In the past decade, a very active area of cancer research has been the development of diagnostic methods based on readily accessible body fluids (urine, nipple discharges, blood, and saliva). [15] Here, we discuss the most promising investigations, which are based on blood molecular markers. One method to detect cancer is an investigation of the patterns of proteins in serum and other bodily fluids. It seems that SELDI-TOF′s specificity and sensitivity in the serum proteome profiling are greater than some older methods, despite its technical difficulties, some studies showed that protein markers are valuable complimentary markers to diagnose cancer. [16],[17],[18],[19]

CIRCULATING TUMOR DNA

Scientists for more than 50 years have been able to detect small quantities of DNA in the plasma of normal individuals. In patients with conditions such as cancer and chronic autoimmune disorders, increased quantities of circulating DNA are detected. Cell-free nucleic acids in plasma were reported in 1948 by Mandel et al. for the 1 st time but recognized in 1977 and reported by Leon et al. In this study, the elevated circulating tumor DNA (ctDNA) concentration in serum of cancer patients was compared to normal controls. [20],[21],[22] Tumor DNA has been identified in the serum and plasma of different cancers patients [23] as reported in breast cancer patients and 3-4 times more ctDNA has been reported to be present in the serum of these patients when compared with that of healthy controls. [24]

The source of this cell-free DNA remains unclear. Different hypotheses are proposed for the origin of this circulating DNA. A tiny amount may originate from blood lymphocytes. Other possible sources of ctDNA include released DNA from necrotic or apoptotic tumor cells of primary and metastatic tumors or tumor DNA secretion and a partly contamination of serum by DNA released from leukocytes. [20],[25],[26],[27],[28] The most common hypothesis for ctDNA origin in the serum of cancer patients is the lyses of circulating tumor cells (CTCs). [26],[27] There is also some evidence that DNA is released from the tumor cells as a glyconucleoprotein complex, which may protect it from nucleases. [25]

Ashworth reported existence of CTC in the peripheral blood of patients with cancer for the 1 st time in 1869; [29] from then existence of CTC of many different tumors in the peripheral blood of patients has been revealed and been thought to be a source of ctDNA. Among all the cancers in women, presence of CTC in the blood has been most investigated in breast cancer. [30]

Different studies have searched for genetic and epigenetic alterations in plasma ctDNA in different types of cancers. ctDNA can be used for the development of noninvasive tests for cancer patients in screening, prognosis, prediction, monitoring of therapies and drug resistance. [24],[25],[31],[32],[33] In recent years, many studies have shown the usefulness of ctDNA as a surrogate marker for breast cancer detection. [26],[34],[35] Several studies have shown that circulating DNA of cancer patients contains most of the DNA alterations specific to cancer. [20],[21],[25] The sensitivity of polymerase chain reaction (PCR) techniques in the detection of these alterations in plasma DNA may be as high as 86%; and the specificity shown in these studies range from about 28% to 100%. [36] In cancer patients, the same mutations such as K-ras, N-ras, p53, have been found in tumor tissue and ctDNA. [20],[33] Divella et al. quantified the hTERT gene in the plasma of patients with primary breast cancer and tested its correlation with the clinical parameters of disease. They used reverse transcription-PCR and it was revealed that circulating hTERT DNA has a better diagnostic value than glycoprotein cancer antigen 15.3 biomarker in early breast cancer disease and hTERT could be a possible candidate as a tumor marker in patients with histology of infiltrating ductal carcinoma. [37] Chen et al. isolated DNA from the plasma or serum of patients with a suspected diagnosis of breast cancer to analyze this DNA for the presence of microsatellite instability and loss of heterozygosity and compared it with corresponding tumor DNA. [38]

Changes in the status of DNA methylation represent one of the most common molecular alterations in human neoplasia including breast cancer. These epigenetic alterations induce neoplastic process by transcriptional silencing of tumor suppressor gene expression and are responsible for initial steps of induction of tumor cell proliferation. Therefore, analysis of gene methylation patterns in tissues has the potential to play a profound significance in the early detection of cancer. [39] The most efficient method of early diagnosis would be the analysis of the methylation pattern in body fluids like serum, urine or milk in early cancer development. It has been shown that these methylation pattern changes originate from cancer cells. In [Table 1] and [Table 2] in this review, we have provided reported examples of many genes in hypermethylated or hypomethylated states in breast cancer using a variety of techniques for detection of methylation in DNA sequence.{Table 1}{Table 2}

Many studies have demonstrated alterations in methylation patterns of ctDNA in various malignancies. [23],[24],[26],[27],[31],[34],[40],[41],[42] Mirza found a significant positive correlation between promoter hypermethylation of estrogen receptor β (ERβ) and retinoic acid receptor β2 (RAR-β2) in matched tissue DNA and serum DNA. They identified no hypermethylation in serum DNA without presence in the primary tumor. [43] However, the frequency of detected methylated markers is two-fold lower in plasma samples compared with their frequency in the tissues of cancer patients. [39]

To find out whether changes observed in ctDNA is identical to those found in primary tumor, Silva et al. investigated one of the inactivation mechanisms of p16INK4a (i.e. hypermethylation) in their patients with breast carcinomas and searched for the possibility of finding this phenomenon in plasma DNA of these patients. They used a PCR-based methylation assay, based on the inability of some restriction enzymes to cut methylated DNA sequences, they found out aberrant DNA hypermethylation in exon 1 of p16INK4a in plasma DNA of these patients is identical to the alteration present in the corresponding carcinomas. [36] They also demonstrated the same microsatellite alterations in ctDNA tumor of 61% of breast cancer patients. [44] Also, Yazici et al. examined RASSF1A promoter methylation and found that RASSF1A promoter hypermethylation was more frequent in affected cancer patients, and their unaffected siblings compared with population-based controls. [45]

Based on the fact that extracellular nucleic acids circulate in blood and not only in plasma and these molecules are also bound to the surface of blood cells, Skvortsova et al., evaluated the cell surface-bound DNA as a source of material for methylation specific PCR (MSP) diagnostics along with ctDNA isolated from the plasma. Aberrant hypermethylation of RASSF1A, RAR-β2 and HIC-1 gene promoters was determined by MSP. The hypermethylation of all three genes was detected considerably more frequently in the cell bound ctDNA than in the plasma ctDNA of the tumor-bearing patients. [39]

The potential to diagnose different cancers including breast cancer [23] based on abnormal hypermethylation or hypomethylation in ctDNA has been demonstrated. [46] Yamamoto et al. analyzed methylation status in serum DNA before and after surgery and their results demonstrated that the origin of the aberrantly methylated genes was the tumor tissue. [47] Also, benign, inflammatory and malignant diseases could be differentially identified through methylation analysis of ctDNA [46] Fiegl et al. demonstrated that RASSF1A DNA methylation disappears in serum throughout treatment with tamoxifen. [32]

Tumor suppressor genes such as APC, RASSF1A, TMS-1, DKK3 and DAPK, cell cycle-related genes (14-3-3-sigma, GSTP1, p16 and RAR-β), metastasis-related genes (ITIH5 and E-cadherin), and others (ESR1, SLC19A3 and HIN-1) have also been studied as potent biomarkers. [28] Skvortsova et al. demonstrated the prevalence of RAR-β2 and RASSF1A promoter hypermethylation in serum of 95% of the cancer patients and 60% of the patients with fibroadenoma and was not found in healthy women. [39] Another study showed significant promoter hypermethylation in HIN-1, RAR-β, RASSF1A and Twist genes in serum DNA of patients with DCIS or invasive ductal carcinoma (IDC). In this study detection of hypermethylation of at least one of the RAR-β and RASSF1A genes from ctDNA provided a sensitivity and specificity of 94.1% and 88.8%, respectively for the detection of DCIS/IDC. [23]

Significantly higher methylation frequencies of two genes (APC and RASSF1A) have been shown in the serum DNA of breast cancer patients. Also, in this study, it was shown that detection of a methylated gene in serum was significantly associated with the detection of CTC in peripheral blood. CTCs were detected in 45% of patients with at least one gene methylated. [26] Chimonidou et al. demonstrated CST6 promoter hypermethylation in serum DNA of 39.8% of the patients, while it was not found in healthy individuals. [31] Detection of methylated SLC19A3 gene promoter in serum DNA showed that hypermethylation of this gene differentiated between breast cancer patients and healthy controls with a sensitivity of 87% and a specificity of 85%. [35]

Recently, an analysis published by Chimonidou et al., [48] evaluated connection between the presence of CTCs and ctDNA in patients with breast cancer, after surgical removal of the primary tumor. They demonstrated concordance between serum level of SOX17 hypermethylation and CTC for 70.9% of the patients. Hypermethylation of ITIH5, DKK3 and RASSF1A in ctDNA of breast cancer patients achieved 67% sensitivity with a specificity of 69%. [40] Silva et al. investigated microsatellite alterations (in D17S855, D17S654, D16S421, TH2, D10S197, and D9S161 polymorphic markers) as well as mutations in the p53 gene and aberrant hypermethylation at the first exon of p16INK4a in tumor and plasma DNA. They identified 56 cases (90%) in which there were at least one alteration in tumor DNA and 41 cases (66%) with a similar alteration in ctDNA. [44] One-step MSP assay of methylated GSTP1, RASSF1A, and RAR-β2 gene promoters showed that hypermethylation of these three genes differentiated between breast cancer patients and healthy controls with a sensitivity of 78%. In this study in which samples exhibited hypermethylation of GSTP1, RASSF1A, and RAR-β2 genes in serum DNA, hypermethylation of the same genes in tumor tissues were reported. [47] In another study Bae et al. demonstrated almost all of the primary breast tumors (97%) were abnormally methylated in at least one of cyclin D2, RAR-β, twist and HIN-1 genes. Hypermethylation in the ctDNA was detected in 67% of the patients with confirmed hypermethylation of the genes in the tumor. [41] RAR-β2 and ERβ promoters were reported to be hypermethylated in 26% and 61% of tumors and in 20% and 50% of ctDNA of breast cancer patients, respectively. Thus, concordance between tumor and serum hypermethylation status was observed. [43] Papadopoulou et al. analyzed ctDNA and methylation patterns of GSTP1, RASSF1A and ATM genes in plasma of breast and prostate cancer patients. They demonstrated that the combination of DNA increase and promoter hypermethylation of GSTP1, RASSF1A, and ATM genes can identify 54% of breast cancer and 88% of prostate cancer patients. [49]

Branham et al. reported hypermethylation for 110 CpG islan (CPGI) within more than 60 cancer-related genes in tumors with triple-negative (TN) features. Breast cancers identified as TN are defined as tumors that lack the expressions of estrogen and progesterone receptors and epidermal growth factor receptor 2. Their results revealed that 18 of CpGI (located within 16 different cancer-involved genes) were associated with TN tumors, indicative of a specific methylation profile. This panel includes: Five hypermethylated genes (CDKN2B, CD44, MGMT, RB, p73) and 11 hypomethylated genes (GSTP1, PMS2, MSH2, MLH1, MSH3, MSH6, DLC1, CACNA1A, CACNA1G, TWIST1, ID4). [50]

Martínez-Galαn et al. investigated ESR1, APC, RAR-β, 14-3-3-ó and E-cadherin promoter hypermethylation in serum of 106 women with breast cancer, 34 with benign breast disease and 74 with no evidence of breast disease. Results indicated that hypermethylation of ESR1 and 14-3-3-ó genes differentiated between breast cancer patients and healthy individuals with a sensitivity of 81% and a specificity of 88%. [51] In 2003, Ramirez et al. reported hypermethylation of MGMT, p16, DAPK and RASSF1A genes in serum DNA among glioblastoma patients. The methylation profile of ctDNA has also been used to identify other cancers. [24] Forshew et al. identified mutations throughout the tumor suppressor gene TP53 in ctDNA from 46 to 69 plasma samples (67%) of advanced ovarian cancer patients. [52] Methylation pattern changes in cancer genes in blood could be a prime target for early detection of cancers in the future. Currently, technologies are being developed to detect minor changes in the blood DNA of cancer patients.

METHODS USED IN DNA METHYLATION PATTERN ANALYSIS

A wide range of methods is used to discriminate methylated and umethylated cytosine in DNA sequence. These methods are based on the chemical and physical differences of cytosine and 5 m-cytosine that can combine into various techniques. Three of the methods are briefly described below, and the remaining are listed in [Table 3].{Table 3}

Bisulfite modification

This powerful chemical reaction can change the indistinguishable cytosine to uracil and leaves 5-methylcytosine residue intact. This technique allows specific changes in the DNA sequence depending on the DNA methylation status. The use of this technique allows downstream processes like PCR and sequencing of DNA and the use of sequence specific restriction enzymes for identification of methylation patterns. [53]

Allele-specific bisulfite sequencing

It is very much like regular sequencing of single alleles. Primers not overlapping CpG sites are used to amplify the desired sequence, the design of such primers have been much easier after the initiation of ENCODE and CANCER methylome projects, these databases allow the rapid and accurate detection of differentially methylated regions in cancer and normal cells and they help locating optimal sequence. After bisulfite conversion and PCR, products are ligated to a cloning vector to amplify individually. Each vector is then sequenced by any sequencing method suitable for the study. If a sufficient number of clones are sequenced, this method can be quantitative, as each clone represents a single allele. [54],[55]

Methylation specific quantum dot fluorescence resonance energy transfer

Another technique to detect methylation of different genes is methylation specific quantum dot fluorescence resonance energy transfer (MS-qFRET). It is a nanotechnology assay that enables the detection of methylation and its changes in an ultrasensitive manner. MS-qFRET is a combination of the high specificity of MSP and the high sensitivity of quantum dot fluorescence resonance energy transfer technology. The procedure of this method composed of treating DNA template with sodium bisulfite that converts unmethylated cytosines to uracil. Amplification of converted template is done using biotinylated methylation-specific primers (biotinylated forward primer and the reverse primer is labeled with an organic fluorophore). Quantum dots, conjugated with streptavidin, serve as a scaffold to capture amplicons and as a donor for transferring energy to the Cy5 acceptor. Cy5 acceptor has been incorporated into the amplicons during PCR. Thus, the status of DNA methylation can be determined according to the level of fluorescence resonance energy transfer. This technique causes reduction in a number of PCR cycles, and allows for multiplexed analyses. [56],[57]

These techniques offer different costs of the utilization, specificity, output range and speed and ease. In [Table 3], we have listed the most popular methods used for detection of DNA methylation patterns.

PERSPECTIVE

The cancer phenotype is a combination of genetic and environmental effects like most other traits in living organisms. The promise of DNA methylation analysis as a cancer biomarker partially comes from the fact that sequencing methods utilized in the process of methylation pattern analysis can investigate the effects of genetic and environmentally induced epigenetic changes at the same time.

Many studies have shown that ctDNA methylation analysis can be a highly selective method for screening early breast cancer. Regarding the variety of methods used to evaluate methylation patterns, it is not unimaginable for robust methods to be developed for assessing cancer progression by methylation pattern analysis specifically, the lower costs of DNA sequencing in recent years make sequence-based methods more widely accessible by many laboratories and eventually patients. This method is particularly applicable in cases where the bodily fluids are easily obtained and may be routinely used in diagnostic laboratories in the near future.[75]

References

Available from: http://seer.cancer.gov/statfacts/html/breast.html [Last accessed on 2015 Jan 12].

Babu GR, Samari G, Cohen SP, Mahapatra T, Wahbe RM, Mermash S, et al. Breast cancer screening among females in Iran and recommendations for improved practice: A review. Asian Pac J Cancer Prev 2011;12:1647-55.

Sadjadi A, Nouraie M, Ghorbani A, Alimohammadian M, Malekzadeh R. Epidemiology of breast cancer in the Islamic Republic of Iran: First results from a population-based cancer registry. East Mediterr Health J 2009;15:1426-31.

Nelson HD, Zakher B, Cantor A, Fu R, Griffin J, O′Meara ES, et al. Risk factors for breast cancer for women aged 40 to 49 years: A systematic review and meta-analysis. Ann Intern Med 2012;156:635-48.

Gilbert FJ. Breast cancer screening in high risk women. Cancer Imaging 2008;8 Spec No A: S6-9.

Zhang F, Chen JY. Breast cancer subtyping from plasma proteins. BMC Med Genomics 2013;6 Suppl 1:S6.

Sørlie T, Perou CM, Tibshirani R, Aas T, Geisler S, Johnsen H, et al. Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad Sci U S A 2001;98:10869-74.

Perou CM, Sørlie T, Eisen MB, van de Rijn M, Jeffrey SS, Rees CA, et al. Molecular portraits of human breast tumours. Nature 2000;406:747-52.

Cancer Genome Atlas Network. Comprehensive molecular portraits of human breast tumours. Nature 2012;490:61-70.

Lee JM, Halpern EF, Rafferty EA, Gazelle GS. Evaluating the correlation between film mammography and MRI for screening women with increased breast cancer risk. Acad Radiol 2009;16:1323-8.

Michell M. Breast screening review - A radiologist′s perspective. Breast 2012;85:845-847.

Warner E, Heisey R, Carroll JC. Applying the 2011 Canadian guidelines for breast cancer screening in practice. CMAJ 2012;184:1803-7.

Nelson HD, Tyne K, Naik A, Bougatsos C, Chan BK, Humphrey L, et al. Screening for breast cancer: An update for the U.S. Preventive Services Task Force. Ann Intern Med 2009;151:727-37, W237.

Malhotra GK, Zhao X, Band H, Band V. Histological, molecular and functional subtypes of breast cancers. Cancer Biol Ther 2010;10:955-60.

Michener CM, Ardekani AM, Petricoin EF 3 rd , Liotta LA, Kohn EC. Genomics and proteomics: Application of novel technology to early detection and prevention of cancer. Cancer Detect Prev 2002;26:249-55.

Ardekani AM, Akhondi MM, Sadeghi MR. Application of genomic and proteomic technologies to early detection of cancer. Arch Iran Med 2008;11:427-34.

Petricoin EF, Liotta LA. SELDI-TOF-based serum proteomic pattern diagnostics for early detection of cancer. Curr Opin Biotechnol 2004;15:24-30.

Fan Y, Wang J, Yang Y, Liu Q, Fan Y, Yu J, et al. Detection and identification of potential biomarkers of breast cancer. J Cancer Res Clin Oncol 2010;136:1243-54.

Braunschweig T, Krieg RC, Bar-Or R, Smeets D, Schwamborn K, Fogt F, et al. Protein profiling of non-malignant and malignant ascites by SELDI-TOF MS: Proof of principle. Int J Mol Med 2009;23:3-8.

Schwarzenbach H, Hoon DS, Pantel K. Cell-free nucleic acids as biomarkers in cancer patients. Nat Rev Cancer 2011;11:426-37.

Sharma VK, Vouros P, Glick J. Mass spectrometric based analysis, characterization and applications of circulating cell free DNA isolated from human body fluids. Int J Mass Spectrom 2011;304:172-83.

Leon SA, Shapiro B, Sklaroff DM, Yaros MJ. Free DNA in the serum of cancer patients and the effect of therapy. Cancer Res 1977;37:646-50.

Kim JH, Shin MH, Kweon SS, Park MH, Yoon JH, Lee JS, et al. Evaluation of promoter hypermethylation detection in serum as a diagnostic tool for breast carcinoma in Korean women. Gynecol Oncol 2010;118:176-81.

Ramirez JL, Taron M, Balaña C, Sarries C, Mendez P, de Aguirre I, et al. Serum DNA as a tool for cancer patient management. Rocz Akad Med Bialymst 2003;48:34-41.

Van De Voorde L, Speeckaert R, Van Gestel D, Bracke M, De Neve W, Delanghe J, et al. DNA methylation-based biomarkers in serum of patients with breast cancer. Mutat Res 2012;751:304-25.

Van der Auwera I, Elst HJ, Van Laere SJ, Maes H, Huget P, van Dam P, et al. The presence of circulating total DNA and methylated genes is associated with circulating tumour cells in blood from breast cancer patients. Br J Cancer 2009;100:1277-86.

Jahr S, Hentze H, Englisch S, Hardt D, Fackelmayer FO, Hesch RD, et al. DNA fragments in the blood plasma of cancer patients: Quantitations and evidence for their origin from apoptotic and necrotic cells. Cancer Res 2001;61:1659-65.

Li L, Choi JY, Lee KM, Sung H, Park SK, Oze I, et al. DNA methylation in peripheral blood: A potential biomarker for cancer molecular epidemiology. J Epidemiol 2012;22:384-94.

Ashworth TR. A case of cancer in which cells similar to those in the tumors were seen in the blood after death. Aust Med J 1869;14:146-9.

Obermayr E, Sanchez-Cabo F, Tea MK, Singer CF, Krainer M, Fischer MB, et al. Assessment of a six gene panel for the molecular detection of circulating tumor cells in the blood of female cancer patients. BMC Cancer 2010;10:666.

Chimonidou M, Tzitzira A, Strati A, Sotiropoulou G, Sfikas C, Malamos N, et al. CST6 promoter methylation in circulating cell-free DNA of breast cancer patients. Clin Biochem 2013;46:235-40.

Fiegl H, Millinger S, Mueller-Holzner E, Marth C, Ensinger C, Berger A, et al. Circulating tumor-specific DNA: A marker for monitoring efficacy of adjuvant therapy in cancer patients. Cancer Res 2005;65:1141-5.

Liggett TE, Melnikov AA, Marks JR, Levenson VV. Methylation patterns in cell-free plasma DNA reflect removal of the primary tumor and drug treatment of breast cancer patients. Int J Cancer 2011;128:492-9.

Murtaza M, Dawson SJ, Tsui DW, Gale D, Forshew T, Piskorz AM, et al. Non-invasive analysis of acquired resistance to cancer therapy by sequencing of plasma DNA. Nature 2013;497:108-12.

Ng EK, Leung CP, Shin VY, Wong CL, Ma ES, Jin HC, et al. Quantitative analysis and diagnostic significance of methylated SLC19A3 DNA in the plasma of breast and gastric cancer patients. PLoS One 2011;6:e22233.

Silva JM, Dominguez G, Villanueva MJ, Gonzalez R, Garcia JM, Corbacho C, et al. Aberrant DNA methylation of the p16INK4a gene in plasma DNA of breast cancer patients. Br J Cancer 1999;80:1262-4.

Divella R, Tommasi S, Lacalamita R, Daniele A, Abbate I, Garrisi VM, et al. Circulating hTERT DNA in early breast cancer. Anticancer Res 2009;29:2845-9.

Chen X, Bonnefoi H, Diebold-Berger S, Lyautey J, Lederrey C, Faltin-Traub E, et al. Detecting tumor-related alterations in plasma or serum DNA of patients diagnosed with breast cancer. Clin Cancer Res 1999;5:2297-303.

Skvortsova TE, Rykova EY, Tamkovich SN, Bryzgunova OE, Starikov AV, Kuznetsova NP, et al. Cell-free and cell-bound circulating DNA in breast tumours: DNA quantification and analysis of tumour-related gene methylation. Br J Cancer 2006;94:1492-5.

Kloten V, Becker B, Winner K, Schrauder MG, Fasching PA, Anzeneder T, et al. Promoter hypermethylation of the tumor-suppressor genes ITIH5, DKK3, and RASSF1A as novel biomarkers for blood-based breast cancer screening. Breast Cancer Res 2013;15:R4.

Bae YK, Shim YR, Choi JH, Kim MJ, Gabrielson E, Lee SJ, et al. Gene promoter hypermethylation in tumors and plasma of breast cancer patients. Cancer Res Treat 2005;37:233-40.

Suijkerbuijk KP, van Diest PJ, van der Wall E. Improving early breast cancer detection: Focus on methylation. Ann Oncol 2011;22:24-9.

Mirza Sharma G. Clinical significance of promoter hypermethylation of ERb and RARb2 in tumor and serum DNA in Indian breast cancer patients. Ann Surg Oncol 2012;19:3107-3115.

Silva JM, Dominguez G, Garcia JM, Gonzalez R, Villanueva MJ, Navarro F, et al. Presence of tumor DNA in plasma of breast cancer patients: Clinicopathological correlations. Cancer Res 1999;59:3251-6.

Yazici H, Terry MB, Cho YH, Senie RT, Liao Y, Andrulis I, et al. Aberrant methylation of RASSF1A in plasma DNA before breast cancer diagnosis in the Breast Cancer Family Registry. Cancer Epidemiol Biomarkers Prev 2009;18:2723-5.

Levenson VV. DNA methylation as a universal biomarker. Expert Rev Mol Diagn 2010;10:481-8.

Yamamoto N, Nakayama T, Kajita M, Miyake T, Iwamoto T, Kim SJ, et al. Detection of aberrant promoter methylation of GSTP1, RASSF1A, and RARß2 in serum DNA of patients with breast cancer by a newly established one-step methylation-specific PCR assay. Breast Cancer Res Treat 2012;132:165-73.

Chimonidou M, Strati A, Malamos N, Georgoulias V, Lianidou ES. SOX17 promoter methylation in circulating tumor cells and matched cell-free DNA isolated from plasma of patients with breast cancer. Clin Chem 2013;59:270-9.

Papadopoulou E, Davilas E, Sotiriou V, Georgakopoulos E, Georgakopoulou S, Koliopanos A, et al. Cell-free DNA and RNA in plasma as a new molecular marker for prostate and breast cancer. Ann N Y Acad Sci 2006;1075:235-43.

Branham MT, Marzese DM, Laurito SR, Gago FE, Orozco JI, Tello OM, et al. Methylation profile of triple-negative breast carcinomas. Oncogenesis 2012;1:e17.

Martínez-Galán J, Torres B, Del Moral R, Muñoz-Gámez JA, Martín-Oliva D, Villalobos M, et al. Quantitative detection of methylated ESR1 and 14-3-3-sigma gene promoters in serum as candidate biomarkers for diagnosis of breast cancer and evaluation of treatment efficacy. Cancer Biol Ther 2008;7:958-65.

Forshew T, Murtaza M, Parkinson C, Gale D, Tsui DW, Kaper F, et al. Noninvasive identification and monitoring of cancer mutations by targeted deep sequencing of plasma DNA. Sci Transl Med 2012;4:136ra68.

Wang RY, Gehrke CW, Ehrlich M. Comparison of bisulfite modification of 5-methyldeoxycytidine and deoxycytidine residues. Nucleic Acids Res 1980;8:4777-90.

Available from: http://cbbiweb.uthscsa.edu/KMethylomes/ [Last accessed on 2015 Jan 12].

Frommer M, McDonald LE, Millar DS, Collis CM, Watt F, Grigg GW, et al. A genomic sequencing protocol that yields a positive display of 5-methylcytosine residues in individual DNA strands. Proc Natl Acad Sci U S A 1992;89:1827-31.

Bailey VJ, Easwaran H, Zhang Y, Griffiths E, Belinsky SA, Herman JG, et al. MS-qFRET: A quantum dot-based method for analysis of DNA methylation. Genome Res 2009;19:1455-61.

Bailey VJ, Keeley BP, Razavi CR, Griffiths E, Carraway HE, Wang TH. DNA methylation detection using MS-qFRET, a quantum dot-based nanoassay. Methods 2010;52:237-41.

Herman JG, Graff JR, Myöhänen S, Nelkin BD, Baylin SB. Methylation-specific PCR: A novel PCR assay for methylation status of CpG islands. Proc Natl Acad Sci U S A 1996;93:9821-6.

Cottrell SE, Distler J, Goodman NS, Mooney SH, Kluth A, Olek A, et al. A real-time PCR assay for DNA-methylation using methylation-specific blockers. Nucleic Acids Res 2004;32:e10.

Shen L, Waterland RA. Methods of DNA methylation analysis. Curr Opin Clin Nutr Metab Care 2007;10:576-81.

Xiong Z, Laird PW. COBRA: A sensitive and quantitative DNA methylation assay. Nucleic Acids Res 1997;25:2532-4.

Radpour R, Haghighi MM, Fan AX, Torbati PM, Hahn S, Holzgreve W, et al. High-throughput hacking of the methylation patterns in breast cancer by in vitro transcription and thymidine-specific cleavage mass array on MALDI-TOF silico-chip. Mol Cancer Res 2008;6:1702-9.

Smiraglia DJ, Kazhiyur-Mannar R, Oakes CC, Wu YZ, Liang P, Ansari T, et al. Restriction landmark genomic scanning (RLGS) spot identification by second generation virtual RLGS in multiple genomes with multiple enzyme combinations. BMC Genomics 2007;8:446.

Yamagata K. DNA methylation profiling using live-cell imaging. Methods 2010;52:259-66.

Boon K, Tomfohr JK, Bailey NW, Garantziotis S, Li Z, Brass DM, et al. Evaluating genome-wide DNA methylation changes in mice by Methylation Specific Digital Karyotyping. BMC Genomics 2008;9:598.

Yang X, Yan L, Davidson NE. DNA methylation in breast cancer. Endocr Relat Cancer 2001;8:115-27.

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:R3.

Hill VK, Ricketts C, Bieche I, Vacher S, Gentle D, Lewis C, et al. Genome-wide DNA methylation profiling of CpG islands in breast cancer identifies novel genes associated with tumorigenicity. Cancer Res 2011;71:2988-99.

Szyf M, Pakneshan P, Rabbani SA. DNA methylation and breast cancer. Biochem Pharmacol 2004;68:1187-97.

Skryabin NA, Tolmacheva EN, Lebedev IN, Zav′ialova MV, Slonimskaia EM, Cherdyntseva NV. Dynamics of methylation changes within functional groups of genes during breast cancer progression. Mol Biol (Mosk) 2013;47:302-10.

Shen X, Li S, Zhang L, Li H, Hong G, Zhou X, et al. An integrated approach to uncover driver genes in breast cancer methylation genomes. PLoS One 2013;8:e61214.

Heyn H, Carmona FJ, Gomez A, Ferreira HJ, Bell JT, Sayols S, et al. DNA methylation profiling in breast cancer discordant identical twins identifies DOK7 as novel epigenetic biomarker. Carcinogenesis 2013;34:102-8.

Swift-Scanlan T, Vang R, Blackford A, Fackler MJ, Sukumar S. Methylated genes in breast cancer: Associations with clinical and histopathological features in a familial breast cancer cohort. Cancer Biol Ther 2011;11:853-65.

Chen ST, Choo KB, Hou MF, Yeh KT, Kuo SJ, Chang JG. Deregulated expression of the PER1, PER2 and PER3 genes in breast cancers. Carcinogenesis 2005;26:1241-6.

Mehrotra J, Vali M, McVeigh M, Kominsky SL, Fackler MJ, Lahti-Domenici J, et al. Very high frequency of hypermethylated genes in breast cancer metastasis to the bone, brain, and lung. Clin Cancer Res 2004;10:3104-9.

Copyright © 2024 Clinical Cancer Investigation Journal
Creative Commons License 
ISSN Print: 2278-1668, Online: 2278-0513