Skip to main content
Journal of Epidemiology logoLink to Journal of Epidemiology
. 2012 Sep 5;22(5):384–394. doi: 10.2188/jea.JE20120003

DNA Methylation in Peripheral Blood: A Potential Biomarker for Cancer Molecular Epidemiology

Lian Li 1,2,3, Ji-Yeob Choi 4, Kyoung-Mu Lee 5, Hyuna Sung 4, Sue K Park 1,2,4, Isao Oze 6, Kai-Feng Pan 7, Wei-Cheng You 7, Ying-Xuan Chen 8, Jing-Yuan Fang 8, Keitaro Matsuo 6, Woo Ho Kim 9, Yasuhito Yuasa 10, Daehee Kang 1,2,4
PMCID: PMC3798632  PMID: 22863985

Abstract

Aberrant DNA methylation is associated with cancer development and progression. There are several types of specimens from which DNA methylation pattern can be measured and evaluated as an indicator of disease status (from normal biological process to pathologic condition) and even of pharmacologic response to therapy. Blood-based specimens such as cell-free circulating nucleic acid and DNA extracted from leukocytes in peripheral blood may be a potential source of noninvasive cancer biomarkers. In this article, we describe the characteristics of blood-based DNA methylation from different biological sources, detection methods, and the factors affecting DNA methylation. We provide a comprehensive literature review of blood-based DNA methylation as a cancer biomarker and focus on the study of DNA methylation using peripheral blood leukocytes. Although DNA methylation patterns measured in peripheral blood have great potential to be useful and informative biomarkers of cancer risk and prognosis, large systematic and unbiased prospective studies that consider biological plausibility and data analysis issues will be needed in order to develop a clinically feasible blood-based assay.

Key words: DNA methylation, blood-based biomarker, serum, plasma, leukocyte, peripheral blood

1. INTRODUCTION

Biomarkers are biological molecules in body fluids or tissues that are quantitatively measured and evaluated as indicators of normal biological processes, pathogenesis, or pharmacologic response to a therapeutic intervention.1 Numerous types of biomarkers have been developed and used for early detection of cancer and prediction of prognosis and treatment response in cancer patients.

DNA methylation is a main component of the epigenetic mechanism that regulates embryonic development, transcription, chromatin structure, X-chromosome inactivation, genomic imprinting, and chromosome stability.2 DNA methylation occurs at the 5-carbon position of cytosine residues located in dinucleotide CpG sites. Although CpG within intergenic and transposable elements throughout the whole genome are mostly methylated, most CpG islands at the promoter region are unmethylated.3 In cancer, global loss of DNA methylation (global hypomethylation), as well as hypermethylation and hypomethylation of specific loci, has been observed. It has been suggested that altered DNA methylation initiates carcinogenesis and promotes cancer progression by activating oncogenes, suppressing tumor suppressor genes, and inducing chromosome instabilities.4

Because DNA is much more stable than other biological materials, such as RNA or protein, DNA methylation is easy to detect in small specimens and thus may be suitable for large-scale epidemiologic studies. Previous studies of the potential of DNA methylation as a cancer biomarker mainly used tumor tissue. However, an increasing number of studies are using body fluids such as urine, bronchial lavage fluid, breast milk, sputum, plasma and serum, and peripheral blood.5

In this review, we consider only studies that analyzed cell-free circulating DNA in plasma or serum or DNA from peripheral blood leukocytes. We describe the characteristics of blood-based DNA methylation from different biological sources, detection methods, and factors affecting DNA methylation. In addition, we comprehensively review the literature to investigate blood-based DNA methylation as a cancer biomarker, with a focus on studies using peripheral blood leukocytes.

2. ISSUES IN THE DEVELOPMENT OF METHYLATION-BASED BIOMARKERS USING PERIPHERAL BLOOD

2.1 Characteristics of different sources of blood-based DNA methylation

Blood-based DNA methylation is mainly derived from cell-free nucleic acid released from circulating cells in serum or plasma6 or DNA extracted from peripheral blood leukocytes or whole blood cells. Although cell-free DNA from circulating cells can serve as a surrogate for DNA from target tumor tissue, it is mixed with DNA from normal cells, which results in low specificity. In addition, the amount of DNA from serum or plasma is somewhat limited for use as a biomarker.7 However, a pooling method that uses DNA from groups of individuals has shown promise in identifying significant methylation markers.8

In contrast, the amount and quality of DNA extracted from peripheral blood leukocytes or whole blood are not usually a concern. Furthermore, it is common practice in many biospecimen repositories to bank DNA extracted from blood leukocytes. Although DNA from peripheral leukocytes is readily obtainable and easy to handle in laboratory processing and clinical use, the biological plausibility of DNA methylation in peripheral blood leukocytes and whole blood is uncertain.

Global methylation in peripheral blood leukocytes significantly differed between healthy controls and patients with pancreatic cancer, breast cancer, bilateral breast cancer, bladder cancer, and colorectal adenoma.913 Although methylation at specific loci in leukocytes was also observed in people with colorectal tumors, the correlation with target tissue showed little evidence of the origin of leukocyte methylation.14 Similarly ambiguous results were reported with regard to the correlation between methylation at specific loci in peripheral blood leukocytes/whole blood DNA and lung tissue DNA.15 Thus, it is controversial as to whether DNA methylation from peripheral blood leukocytes reflects methylation of target tissue.

It has been suggested that immunologic processes related to inflammation in cancer development lead to changes in leukocyte subpopulations, which could alter the epigenetic signatures in DNA from peripheral blood.16 Another possible explanation is that epigenetic change due to methylation is associated with the genetic variants of specific cancers.16,17 Increased knowledge of the origin and nature of DNA methylation in peripheral blood leukocytes is needed to determine whether DNA methylation in such cells can serve as an informative biomarker. In addition, future studies should investigate variation in DNA methylation in heterogeneous leukocyte subpopulations and differences in the processing of white blood cells.

2.2 Detection methods

To identify a sensitive and specific biomarker, it is important to select an appropriate method that is standardized, robust, sensitive, and cross-validated between laboratories and across different platforms. The details of such methods have been comprehensively reviewed in several articles.4,1821 We will describe the advantages and disadvantages of extant methods of DNA methylation and will focus on the methods frequently used to detect DNA methylation in peripheral blood. Table 1 summarizes the methods used to assess DNA methylation and several of the important factors to be considered in method selection, such as analytical sensitivity measured as limit of detection (LOD), quantitativeness, and time required.

Table 1. Comparison of selected characteristics related to laboratory validation of various DNA methylation assays.

Technology LODa Quantitativeness Time requirement Reference
Candidate geneb        
 MSP 0.1 No <2 hrs/96 74
 Bisulfite sequencing >2 Yes >4 hrs/96 75
 Pyrosequencing 2 Yes 4 hrs/96 76
 COBRA 3 No/Yes 5 hrs/80–160 77, 78
 MS-SnuPE 0.1 Yes 5 hrs/80–160 79, 80
 MethyLight 0.01 Yes <2 hrs/96 81
 MS-FLAG 0.01 Yes <2 hrs/96 82
Genome-wide profiling        
 RLGS No 5–14 d 83, 84
 MSRF No <5–14 d 85
 MeDIP/MIRA 0.1 Yes 2–3 d/12 86
 Beadchip (Infinium) 2.5 Yes 3 d/96 22
5-methylcytosine contents        
 HPLC >1 uM Yes 15–60 min 23
 HPCE 1 uM Yes 10 min 87
 LC-ESI-MS 0.2 fmol Yes 15 min 24

Abbreviations: LOD, limit of detection; MSP, methylation-specific PCR; COBRA, combined bisulfite restriction analysis; MS-SnuPE, methylation-sensitive single-nucleotide primer extension; MS-FLAG, methylation-specific fluorescent amplicon generation; RLGS, restriction landmark genomic scanning; MeDIP, methyl-DNA immunoprecipitation; MIRA, methylated-CpG island recovery assay; HPLC, high-performance liquid chromatography; HPCE, high-performance capillary electrophoresis; LC-ESI-MS, liquid chromatography–electrospray ionization-mass spectrometry.

aRatio of methylated cytosine to unmethylated cytosine (for the gene-specific methylation approach [%]) or the amount of DNA (for the global DNA methylation approach).

bPCR amplification of desired target was conducted after bisulfite conversion.

Methylation at specific loci can be examined in selected candidate genes or genome-wide. Most candidate gene analyses are based on bisulfite treatment and PCR/sequencing followed by quantitative measurement of DNA methylation level. The process of bisulfite treatment and PCR can be done using a relatively small amount of low-quality DNA and is thus suitable for DNA derived from serum or plasma, as well as that from peripheral blood leukocytes. There are 3 types of methods to measure methylation level at specific loci, ie, real-time PCR-based methods (methylation-specific melting curve analysis [MS-MCA], methylation-sensitive high-resolution melting [MS-HRM], HeavyMethyl, MethyLight, melting curve methylation-specific PCR [McMSP], sensitive melting analysis after real-time methylation-specific PCR [SMART-MSP], methylation-specific fluorescent amplicon generation [MS-FLAG], and quantitative analysis of methylated alleles [QAMA]); sequencing-based methods (direct bisulfite sequencing and pyrosequencing); and gel electrophoresis-based methods (combined bisulfite restriction analysis [COBRA] and methylation-sensitive single-nucleotide primer extension [MS-SnuPE]).

Methyl-sensitive enzyme digestion, affinity enrichment, and the bisulfite treatment-based array are used for genome-wide profiling. There are a variety of enzyme digestion methods such as restriction landmark genomic scanning (RLGS), methylation-sensitive restriction fingerprinting (MSRF), differential methylation hybridization (DMH), and methylated CpG island amplification/representational difference analysis (MCA-RDA). However, enzyme digestion methods generally require a large amount of DNA (approximately 10 µg) and have limited coverage for DNA as compared with affinity enrichment methods (methylated-CpG island recovery assay [MIRA], methyl-DNA immunoprecipitation [MeDIP], tiling array, CpG island microarray, and next-generation sequencing [NGS]) or a bisulfite treatment-based array.20 Neither enzyme digestion methods nor affinity enrichment methods can focus on specific CpG sites of interest, and thus the results are likely to be biased toward CpG-dense regions, due to the different efficiency of enzyme digestion and changes in antibody combination through different runs. In contrast, the bisulfite treatment-based array combined with bead array technology, such as the Infinium methylation array, requires only a small amount of DNA (250–500 ng) and is highly reproducible (r2 > 0.998).19 The correlation coefficients of the Infinium and GoldenGate assays, pyrosequencing, and bisulfite sequencing were reported to be greater than 0.8.22 Methods for genome-wide profiling can be classified as array-based analysis and deep sequencing, according to genotyping technology. Bock et al21 compared the different platforms of 4 types of genome-wide DNA methylation-mapping technologies, including methylated DNA immunoprecipitation sequencing (MeDIP-seq), methylated DNA capture by affinity purification sequencing (MethylCap-seq), reduced representation bisulfite sequencing (RRBS), and the Infinium methylation assay. They reported that the accuracy of the RRBS and Infinium assays was slightly higher than that of the other 2 methods. However, the genomic coverage of MeDIP-seq and MethylCap-seq was higher than that of the RRBS and Infinium assays.

Global DNA methylation can be measured by direct and indirect quantification assays. Direct methods, such as the [3H]-methyl incorporation assay, high-performance liquid chromatography (HPLC), high-performance capillary electrophoresis (HPCE), and liquid chromatography–electrospray ionization tandem mass spectrometry (LC-ESI-MS/MS), measure 5-methylcytosine content throughout the genome. LC-based methods are the most common and have good reproducibility. LC-ESI-MS/MS needs less DNA than the other methods (1 µg for LC-ESI-MS/MS vs 5–10 µg for HPLC) and requires less time per sample (15–60 min for separation using HPLC vs <15 min for separation using LC-ESI-MS/MS).23,24 Direct measurement of 5-methycytosine content in DNA requires a large amount of DNA and is labor intensive, which led to the development of an indirect method that measures methylation levels of repetitive elements (ALU, LINE1, and SAT). The repetitive elements represent over 45% of CpG dinucleotides in the human genome and are correlated with 5-methylcytosine levels throughout the genome.25 Using MethyLight, Weisenberger et al reported that the methylation level of repeated elements was correlated with global DNA methylation as measured by HPLC.25 However, it is uncertain whether methylation levels of repetitive elements perfectly represent 5-methylcytosine content.

2.3 Factors affecting DNA methylation

A valid biomarker should have greater interindividual than intraindividual variation and higher interclass than intraclass correlation coefficients. Several studies have shown that DNA methylation pattern changed over time according to various endogenous and exogenous factors, such as demographic and lifestyle factors (age, race, sex, smoking, and alcohol consumption), diet intake (folate, vitamin B, green tea, and phytoestrogen), environmental exposures (arsenic, cadmium, and benzene), and disease status (infection and cancer).26

The methylation levels of several genes were shown to be correlated with smoking, alcohol consumption, and high fat intake, although most studies using cell-free DNA in serum and/or plasma did not show any significant association.2729 Interestingly, several well-designed epidemiology studies found that obesity, dietary pattern, and physical activity were associated with global methylation in DNA extracted from peripheral blood leukocytes. Recently Teschendorff et al30 conducted a genome-wide scan of 27 000 CpG sites in 261 postmenopausal women and found that most CpGs were hypermethylated with age. Breitling et al31 used the same approach and found that specific methylation of F2RL3 was associated with tobacco smoking in 177 individuals and validated this result using mass spectrometry and the Sequenom EpiTYPER in 316 individuals. In addition, a very recent review32 showed that demographic factors (age, sex, race, family history of cancer, education and race), environmental factors (benzene, organic pollutants, lead, arsenic, air pollution), behavioral factors (smoking, alcohol drinking, physical activity, and folate intake), and even genetic variation in carbon-metabolizing enzymes were associated with global methylation level in lymphocyte DNA.9,3338 Thus, potential confounding factors affecting methylation status should be considered in the design of studies evaluating DNA methylation as a cancer biomarker. Furthermore, findings in the discovery stage should be cross-validated using independent samples.

3. PREVIOUS STUDIES OF DNA METHYLATION IN PERIPHERAL BLOOD AS A BIOMARKER OF CANCER RISK AND PROGNOSIS

We searched MEDLINE (PubMed) using the following keywords: DNA methylation, cell in serum and/or plasma, peripheral blood leukocytes, and cancer. We also searched the references of the retrieved articles. Among the identified studies, we included only those that had an epidemiologic design (cohort study, case-control study, or case-only study) and investigated DNA methylation as a biomarker of cancer risk and prognosis.

3.1 Circulating cell-free DNA methylation as a cancer biomarker

Table 2 shows the studies that used circulating cell-free DNA in serum and/or plasma to investigate DNA methylation as a diagnostic biomarker of cancer. In studies using circulating nucleic acid, the candidate gene approach was more frequent than genome-wide analysis, possibly due to the limited amount of available DNA. Previous studies have focused on genes and pathways related to carcinogenesis and tumor progression, namely, tumor oncogenes (TMEFF2, HPP1, and PGR), tumor suppressor genes (TIG1, APC, RASSF1A, and DAPK), cell cycle-related genes (P16INK4, 14-3-3δ, GSTP1, p15, p16, RAR-β, and SEPT9), cell adhesion molecules (CDH1 and CDH13), cell proliferation-related genes (ESR1, MYOD, and PTGS2), tissue invasion- and metastasis-related genes (TIMP3 and E-cadherin), and others (hMLH1, NGFR, AR, MGMT, HLTF, and TPEE). As compared with healthy individuals, DNA methylation of the tumor suppressor genes APC and RASSF1A was altered in circulating cell-free DNA of patients with breast or gastric cancer.3942 DNA methylation of cell cycle-related genes such as P16INK4, 14-3-3δ, GSTP1, p15, p16, RAR-β, and SEPT9 was reported in bladder, lung, prostate, and colorectal cancer and in head and neck squamous cell carcinoma.4348 However, almost all these studies failed to adjust for confounding factors, including well-known cancer risk factors. Only 1 study showing promoter hypermethylation in MGMT, P16INK4α, RASSF1A, DAPK, and RAR-β in lung cancer patients reported risk estimates adjusted for age, sex, smoking status, and protein tumor marker.47 Recently, Epigenomics AG49 conducted a multistage study to identify and validate methylation biomarkers for colorectal cancer. In the first stage, candidate markers were selected by restriction enzyme-based discovery methods using colorectal cancer tissue and normal tissue. In the second stage, candidate genes identified in the first stage (ie, TMEFF2, NGFR, and SEPT9) were confirmed by real-time assays using DNA from circulating plasma cells.48 Finally, SEPT9 methylation identified in the second stage was validated in a clinical trial.

Table 2. Associations between serum and/or plasma DNA methylation and cancer risk.

Genes Sample size
(cases/controls)
Assay Source Resultsa Reference

OR (95% CI), P
Breast cancer          
RASSF1A 33/29 MSP Plasma Case: 12%; Control: 0% 40
APC 79/19 QMSP Serum APC: P = 0.03 39
ESR1 ESR1: P = 0.33
RASSF1A RASSF1A: P = 0.002
APC 36/30 EpiTyper assay Plasma/Serum APC: P < 0.001 88
BIN1 BIN1: P < 0.001
BMP6 BMP6: P = 0.068
BRCA1 BRCA1: P < 0.001
CST6 CST6: P < 0.002
ESR-b ESR-b: P = 0.122
GSTP1 GSTP1: P = 0.003
P16 P16: P < 0.001
P21 P21: P < 0.0001
TIMP3 TIMP3: P < 0.0001
Bladder cancer          
P16INK4α 86/49 MSP Serum 13.6 (1.8–105.2), P = 0.0009 43
Gastric cancer          
RASSF1A 47/30 MSP Serum P < 0.01 41
APC 60/22 MethyLight Serum APC: P = 0.08 56
hMLH1 hMLH1: P = 0.03
TIMP3 TIMP3: P = 0.005
Head and neck squamous cell carcinoma      
p15 20/24 MethyLight Plasma p15: P = 0.0037 46
p16 p16: P = 0.016
Lung cancer          
DAPK 100/100 MSP Serum At least 1 gene positive:
5.3 (2.4–11.7)
At least 2 genes positive:
5.9 (1.5–22.7)
47
MGMT
P16INK4α
RASSF1A
RAR-β
Nasopharyngeal carcinoma        
CDH1 41/43 QMSP Plasma CDH1: P < 0.0001 89
DAPK DAPK: P = 0.002
p15 p15: P = 0.002
p16 p16: P < 0.0001
RASSF1A RASSF1A: P = 0.235
  At least 1 gene positive: P < 0.001
Prostate cancer          
GSTP1 168/11 QMSP Serum GSTP1: P < 0.0001 45
PTGS2 PTGS2: P = 0.05
TIG1 TIG1: P = 0.038
14-3-3δ 46/49 MSP Serum 14-3-3δ: P = 0.03 44
AR AR: P > 0.05
GSTPI GSTP1: P < 0.001

Abbreviations: MSP, methylation-specific PCR; QMSP, quantitative methylation-specific PCR; MSRE, methylation-sensitive restriction enzyme; HPCE, high-performance capillary electrophoresis; LC/MS, liquid chromatography/mass spectrometry; COBRA, combined bisulfite restriction analysis.

aHypermethylation of genes increased cancer risk.

Several studies have evaluated the potential of DNA methylation as a prognostic biomarker of cancer. Table 3 summarizes studies that investigated DNA methylation in circulating cell-free DNA as a prognostic biomarker. In a variety of cancers, including hepatocellular carcinoma, breast, bladder, cervical, and colorectal cancer,5054 both the methylation pattern in specific loci and global methylation level were observed with regard to disease-free survival and/or overall survival. Altered DNA methylation of APC was commonly associated with overall survival in breast, gastric, and esophageal cancer.28,5557 Promoter methylation of cell adhesion molecule genes CDH1 and CDH13 and cell proliferation-related gene MYOD was associated with relapse-free survival in cervical cancer.53,58 Promoter methylation of GSTP1 was associated with disease-free survival in prostate cancer, and hMLH1 was associated with overall survival in ovarian cancer.59,60 In hepatocellular carcinoma, global hypomethylation quantified with LINE1 was associated with overall survival.29 A few studies shown in Table 4 evaluated the predictive values, including sensitivity and specificity, of DNA methylation in predicting cancer outcomes. In colorectal cancer and prostate cancer, multimarker analysis had much higher sensitivity and specificity than did single-marker analysis.61,62 Moreover, in some cases, the sensitivity and specificity of methylation markers were reported to be moderately higher than those of present diagnostic markers in clinical use, such as PSA for prostate cancer, fecal occult blood testing for colorectal cancer, CA125 for ovarian cancer, and combined analysis of CA19-9 and CA125 for pancreatic cancer.

Table 3. Associations between serum and/or plasma DNA methylation and cancer prognosis.

Genes Sample size
(events/non-events)
Assay Resultsa Reference

Outcome HR (95% CI), P
Breast cancer        
RASSF1Ab 13/148 MethyLight Relapse-free survival 5.1 (1.3–19.8) 55
APC 17/85 MethyLight Overall survivalc APC/RASSF1A: 5.7 (1.9–16.9), P = 0.002 28
RASSF1A
PITX2 428 MethyLight Overall survival
Distant disease-free survival
PITX2: 3.4 (1.2–9.8), P = 0.021 51
RASSF1A RASSF1A: 5.6 (2.1–14.5), P < 0.001
  RASSF1A: 3.4(1.6–7.3), P = 0.002
Bladder cancer        
P14ARF 12/15 MSP Relapse-free survival P = 0.03 52
Cervical cancer        
MYOD1 53/40 MethyLight Relapse-free survival P = 0.04 58
CDH1 53/40 MethyLight Relapse-free survival CDH1/CDH13: 2.5 (1.3–4.6), P = 0.005 53
CDH13
Colorectal cancer        
HLTF 28/77 MethyLight Overall survival HLTF: 3.0 (1.4–6.4), P = 0.008 54
HPP1 HPP1: 5.1 (2.2–11.6), P = 0.001
hMLH1 hMLH1: 1.4 (0.6–3.1), P = 0.425
  HLTF/HPP1: 3.4 (1.4–8.1), P = 0.007
Esophageal cancer        
DAPK 59 QMSP Overall survival 0.2 (0.0–0.5), P = 0.0036 90
APC 52 QMSP Overall survivalc P = 0.016 57
Gastric cancer        
APC 32/26 MethyLight Overall survival APC: P = 0.006 56
CDH1 CDH1: P = 0.006
Hepatocellular carcinoma      
LINE1 85 COBRA Overall survival 1.7 (1.1–2.8), P = 0.021 29
Lung cancer        
DAPK 76 QMSP Overall survival DAPK: P = 0.587 90
MGMT MGMT: P = 0.202
14-3-3δ 75/40 MSP Overall survival 2.1 (1.2–3.5), P = 0.006 91
Ovarian cancer        
hMLH1b 78/53 MSP Overall survival 2.0 (1.2–3.3), P = 0.007 59
Prostate cancer        
GSTP1 55/55 REQP Disease-free survival 4.4 (2.2–8.8), P < 0.001 60

Abbreviations: MSP, methylation-specific PCR; QMSP, quantitative methylation-specific PCR; MSP, methylation-specific PCR; COBRA, combined bisulfite restriction analysis; REQP, restriction endonuclease quantitative PCR.

aHypermethylation of genes worsened prognosis.

bMeasurements were done at disease endpoint.

Table 4. Population validation of methylation-based biomarkers using plasma/serum DNA.

Genes Sample size
(cases/controls)
Assay Source Sensitivity
(%)
Specificity
(%)
Reference
Breast cancer            
APC, GSTP1, RASSF1A, RARβ2 93/76 QMSP Plasma 62 87 92
Colorectal cancer            
APC, MGMT, RASSF2A, Wif-1 243/276 MSP Plasma 87 92 62
SEPT9 97/172 Real-time qPCR Plasma 72 93 93
Hepatocellular carcinoma          
P15, P16, RASSF1A 50/50 MSP Serum 84 94 50
Ovarian cancer            
BRCA1, HIC1, PAX5, PGR, THBS1 33/33 MethDet test Plasma 85 61 94
Pancreatic cancer            
CCND2, PLAU, SOCS1, THBS, VHL 30/30 MethDet test Plasma 76 59 94
Prostate cancer            
GSTP1, RASSF1, RARB2 83/40 MSP Serum 89 61

Abbreviations: QMSP, quantitative methylation-specific PCR; MSP, methylation-specific PCR.

3.2 Leukocyte DNA methylation as a cancer biomarker

Table 5 shows the associations between methylation patterns of DNA extracted from leukocytes and cancer risk. Methylation at specific loci in DNA from peripheral blood leukocytes/whole blood was first reported in lung cancer.15 In a nested case-control study (n = 100), the researchers hypothesized that methylation status in DNA extracted from whole blood would reflect the status of lung tissue DNA. They identified a correlation between p53 gene hypomethylation in whole blood DNA and lung cancer. In breast cancer, methylation of specific loci in ERT (NUP155 and ZNF217), PCGT (TITF1, NEUROD1, and SFRP1), and DMHR (PTGS2) of DNA from extracted peripheral blood cells was associated with breast cancer risk.13 Flanagan et al10 compared the methylation pattern of peripheral blood DNA using a custom methylation microarray analysis covering 4 Mb with 51 candidate genes from 14 bilateral breast cancer cases and 14 normal controls and validated their initial findings regarding the tiled region around ATM in 190 pairs of cases and controls. The results proved their hypothesis that some systemic epigenetic changes would be detected in peripheral blood DNA in breast cancer. However, in a case-control study using lymphocyte DNA from 97 colon cancer cases and 190 age- and sex-matched controls, the mean fraction of CpG methylation was identical among cases and controls, and there was no relationship between colon cancer risk and quartile levels of CpG methylation.63

Table 5. Associations between DNA methylation in peripheral blood leukocytes and cancer risk.

Disease/Genes Sample size
(cases/controls)
Assay Results Reference

OR (95%CI)
Breast cancer        
NEUROD1 353/730 MethyLight 1.5 (1.1–2.0) 13
NUP155 1.4 (1.0–1.9)
SFRP1 1.4 (1.1–1.9)
TITF1 1.5 (1.1–2.2)
ZNF217 1.5 (1.1–2.0)
ATM 190 /190 MSRE-microarray,
Pyrosequencing
High vs Low: 3.2 (1.8–5.9) 10
 5-mdC 176/173 LC/MS Middle vs High: 1.5 (0.8–2.7)
Low vs High: 2.9 (1.7–4.9)
9
LINE1 40/40 MethyLight P > 0.05 95
ALU P > 0.05
SAT P = 0.01
Bladder cancer      
 mC contentsa 775/397 HPCE Q1 vs Q4: 2.7 (1.8–4.0)
Q2 vs Q4: 1.6 (1.1–2.4)
Q3 vs Q4: 2.1 (1.4–3.1)
11
LINE1 510/528 Pyrosequencing Middle vs High: 1.3 (0.8–2.3)
Low vs High: 1.9 (1.2–3.1)
64
 Gene panels 111/119 Illumina Infinium beadchip
array
AUC: 0.8 (0.7–0.8) 64
 (9 CpG sites)
LINE1 285/465 Pyrosequencing Low vs High: 1.8 (1.1–2.9) 96
Colon cancer        
IGFII 97/190 SOMA assay Q1 vs Q4: 1.1 (0.5–2.4)
Q2 vs Q4: 1.2 (0.5–2.6)
Q3 vs Q4: 1.4 (0.6–3.0)
63
Colorectal adenoma      
 mC contents 115/115 LC/MS Middle vs Low: 0.7 (0.3–1.5)
High vs Low: 0.2 (0.1–0.5)
12
Gastric cancer        
ALU
LINE1
302/421 Pyrosequencing Low vs High: 1.3 (0.9–1.9)
Low vs High: 1.4 (0.9–2.0)
67
Hereditary diffuse gastric cancer      
CDH1 22/21 Pyrosequencing 25% of cases displayed high CDH1
allelic expression imbalance
97
Head and neck squamous cell carcinoma      
LRE1 278/526 COBRA Middle vs High: 1.3 (0.9–2.0)
Low vs High: 1.6 (1.1–2.4)
34
Lung cancer        
P53 100/100 HpaII quantitative PCR assay 2.2 (1.0–4.7) 15
CSF3R 138/138 Illumina beadchip assay,
pyrosequencing
3.9 (2.0–6.1) 65
ERCC1 1.5 (1.1–2.0)
Ovarian cancer        
 Gene panels 255/148 Illumina Infinium AUC: 0.8 (0.7–0.9) 16
 (100 CpG sites) beadchip array    
Pancreatic cancer      
IL10 220/220 Illumina VeraCode array AUC: 0.8 17
LCN2
ZAP70
AIM2
TAL1

Abbreviations: MSRE, methylation-sensitive restriction enzyme; HPCE, high-performance capillary electrophoresis; 5-mdC, 5-methyldeoxycytosine; LC/MS, liquid chromatography/mass spectrometry; AUC, area under the curve; COBRA, combined bisulfite restriction analysis.

In genome-wide scanning using leukocyte DNA, potential methylation biomarkers were identified in several cancers, including ovarian, pancreatic, bladder, and non–small-cell lung cancers.16,17,64,65 Teschendorff et al16 evaluated the methylation signature of 27 000 CpG sites using DNA extracted from peripheral blood in 113 pretreatment ovarian cancer cases and 148 healthy controls and validated the results among an independent set of 122 post-treatment ovarian cancer cases. Marsit et al64 used an Infinium methylation chip and identified a panel of DNA methylation loci that might serve as a useful biomarker of bladder cancer. In the first phase, they identified a panel of 9 CpG loci in 112 cases and 118 controls and then validated the findings in 111 cases and 119 controls. In the discovery stage of another genome-wide scanning study, CSF3R and ERCC1 gene methylation was identified as a biomarker of small-cell lung cancer using a methylation array of 1505 CpG sites in 39 small-cell lung cancer cases and 44 matched controls, which was validated in an independent set of 138 matched case-control pairs using pyrosequencing.65 Pedersen et al17 conducted a 2-phase study using the GoldenGate methylation Beadchip for phase I and the Illumina custom VeraCode methylation assay for phase II in 220 pairs of cases and controls. They found that a panel of genes (IL10, LCN2, ZAP70, AIM2, and TAL1) might be a diagnostic biomarker of pancreatic cancer.

In addition to methylation in specific genes, associations between global hypomethylation in peripheral blood leukocytes and cancer risk were reported for several cancers, including breast, bladder, colorectal adenoma, head and neck squamous cell carcinoma, tongue and esophageal cancer, and gastric cancer, as shown in Table 5.9,11,12,34,64,6668 Some of these studies attempted to control for confounding factors in the association between methylation level and disease status by selecting subjects in certain cancer stages, as shown in a study of colorectal cancer,12 and by using stratified analysis of confounding factors, such as smoking status in a study of bladder cancer.11 Only 1 study investigated DNA methylation in peripheral blood leukocytes as a prognostic marker of cancer. Using pyrosequencing, Al-Moundhri et al found that global methylation and promoter methylation in p16 were associated with survival among 105 patients with gastric adenocarcinoma.68

4. CONCLUSION AND FUTURE DIRECTIONS

Because DNA methylation data are very complex and diverse, several points should be considered in study design and data analysis. For example, data could represent methylation content, methylation level, methylation pattern, methylation level profile, or methylation pattern profile.69 Second, the format of data might be discrete (qualitative measurement) or continuous (quantitative measurement), depending on the detection method. When continuous data are not normally distributed, they can be transformed or classified into groups for parametric analysis or, alternatively, tested by nonparametric analysis.70 However, methods of statistical analysis of methylation pattern and methylation profile have not been standardized, and the establishment of such techniques should be a topic of future studies.

A well-designed epidemiologic study is needed to evaluate the validity of a putative methylation-based biomarker. Strategies to validate biomarkers were well established in the community of the Early Detection Research Network (EDRN).71,72 The researchers developed a 5-phase strategy for identifying cancer biomarkers. These phases corresponded to 5 epidemiologic phases, as follows: (1) a preclinical exploratory phase in case-control studies with convenient samples, to identify promising directions; (2) a clinical assay and validation phase in population-based case-control studies, for validation in clinical settings; (3) a retrospective longitudinal phase in nested case-control studies, to determine whether the biomarker detects a disease before it becomes clinically significant; (4) a prospective screening phase in cross-sectional cohort studies, to identify the extent and characteristics of a disease and calculate the predictive value73; and (5) a cancer control phase in randomized trials, to quantify the impact of screening on reducing the disease burden in the population. The standard operating procedures of each phase should include details of assays, methods, and protocols for collection and processing of biological samples and other reference materials. In addition, with regard to the characteristics of methylation, the timing of environmental exposure may be critical for altering methylation in disease progression; thus, specimen collection should be done in a prospective longitudinal study and should be done repeatedly.

More data must be collected to determine if blood-based DNA methylation is biologically plausible. In addition, a large prospective study is necessary to determine whether DNA methylation measured in peripheral blood is a useful and informative cancer biomarker.

ACKNOWLEDGMENTS

This work was supported by the Basic Science Research Program through the National Research Foundation of Korea, funded by the Ministry of Education, Science and Technology [2011-0027212], and by the National Research Foundation of Korea (NRF) A3 Foresight program.

Conflicts of interest: None declared.

REFERENCES

  • 1.Wagner PD, Verma M, Srivastava S. Challenges for biomarkers in cancer detection. Ann N Y Acad Sci. 2004;1022:9–16 10.1196/annals.1318.003 [DOI] [PubMed] [Google Scholar]
  • 2.Robertson KD DNA methylation and human disease. Nat Rev Genet. 2005;6:597–610 10.1038/nrg1655 [DOI] [PubMed] [Google Scholar]
  • 3.Wilson AS, Power BE, Molloy PL. DNA hypomethylation and human diseases. Biochim Biophys Acta. 2007;1775:138–62 [DOI] [PubMed] [Google Scholar]
  • 4.Esteller M Cancer epigenomics: DNA methylomes and histone-modification maps. Nat Rev Genet. 2007;8:286–98 10.1038/nrg2005 [DOI] [PubMed] [Google Scholar]
  • 5.Shivapurkar N, Gazdar AF. DNA methylation based biomarkers in non-invasive cancer screening. Curr Mol Med. 2010;10:123–32 10.2174/156652410790963303 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Gormally E, Caboux E, Vineis P, Hainaut P. Circulating free DNA in plasma or serum as biomarker of carcinogenesis: practical aspects and biological significance. Mutat Res. 2007;635:105–17 10.1016/j.mrrev.2006.11.002 [DOI] [PubMed] [Google Scholar]
  • 7.Gormally E, Hainaut P, Caboux E, Airoldi L, Autrup H, Malaveille C, et al. . Amount of DNA in plasma and cancer risk: a prospective study. Int J Cancer. 2004;111:746–9 10.1002/ijc.20327 [DOI] [PubMed] [Google Scholar]
  • 8.Docherty SJ, Davis OS, Haworth CM, Plomin R, Mill J. DNA methylation profiling using bisulfite-based epityping of pooled genomic DNA. Methods. 2010;52:255–8 10.1016/j.ymeth.2010.06.017 [DOI] [PubMed] [Google Scholar]
  • 9.Choi JY, James SR, Link PA, McCann SE, Hong CC, Davis W, et al. . Association between global DNA hypomethylation in leukocytes and risk of breast cancer. Carcinogenesis. 2009;30:1889–97 10.1093/carcin/bgp143 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Flanagan JM, Munoz-Alegre M, Henderson S, Tang T, Sun P, Johnson N, et al. . Gene-body hypermethylation of ATM in peripheral blood DNA of bilateral breast cancer patients. Hum Mol Genet. 2009;18:1332–42 10.1093/hmg/ddp033 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Moore LE, Pfeiffer RM, Poscablo C, Real FX, Kogevinas M, Silverman D, et al. . Genomic DNA hypomethylation as a biomarker for bladder cancer susceptibility in the Spanish Bladder Cancer Study: a case-control study. Lancet Oncol. 2008;9:359–66 10.1016/S1470-2045(08)70038-X [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Lim U, Flood A, Choi SW, Albanes D, Cross AJ, Schatzkin A, et al. . Genomic methylation of leukocyte DNA in relation to colorectal adenoma among asymptomatic women. Gastroenterology. 2008;134:47–55 10.1053/j.gastro.2007.10.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Widschwendter M, Apostolidou S, Raum E, Rothenbacher D, Fiegl H, Menon U, et al. . Epigenotyping in peripheral blood cell DNA and breast cancer risk: a proof of principle study. PLoS ONE. 2008;3:e2656 10.1371/journal.pone.0002656 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Ally MS, Al-Ghnaniem R, Pufulete M. The relationship between gene-specific DNA methylation in leukocytes and normal colorectal mucosa in subjects with and without colorectal tumors. Cancer Epidemiol Biomarkers Prev. 2009;18:922–8 10.1158/1055-9965.EPI-08-0703 [DOI] [PubMed] [Google Scholar]
  • 15.Woodson K, Mason J, Choi SW, Hartman T, Tangrea J, Virtamo J, et al. . Hypomethylation of p53 in peripheral blood DNA is associated with the development of lung cancer. Cancer Epidemiol Biomarkers Prev. 2001;10:69–74 [PubMed] [Google Scholar]
  • 16.Teschendorff AE, Menon U, Gentry-Maharaj A, Ramus SJ, Gayther SA, Apostolidou S, et al. . An epigenetic signature in peripheral blood predicts active ovarian cancer. PLoS ONE. 2009;4:e8274 10.1371/journal.pone.0008274 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Pedersen KS, Bamlet WR, Oberg AL, de Andrade M, Matsumoto ME, Tang H, et al. . Leukocyte DNA methylation signature differentiates pancreatic cancer patients from healthy controls. PLoS ONE. 2011;6:e18223 10.1371/journal.pone.0018223 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Kristensen LS, Hansen LL. PCR-based methods for detecting single-locus DNA methylation biomarkers in cancer diagnostics, prognostics, and response to treatment. Clin Chem. 2009;55:1471–83 10.1373/clinchem.2008.121962 [DOI] [PubMed] [Google Scholar]
  • 19.Bibikova M, Fan JB. Genome-wide DNA methylation profiling. Wiley Interdiscip Rev Syst Biol Med. 2010;2:210–23 10.1002/wsbm.35 [DOI] [PubMed] [Google Scholar]
  • 20.Gupta R, Nagarajan A, Wajapeyee N. Advances in genome-wide DNA methylation analysis. Biotechniques. 2010;49:iii–xi 10.2144/000113493 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Bock C, Tomazou EM, Brinkman AB, Müller F, Simmer F, Gu H, et al. . Quantitative comparison of genome-wide DNA methylation mapping technologies. Nat Biotechnol. 2010;28:1106–14 10.1038/nbt.1681 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Bibikova M, Le J, Barnes B, Saedinia-Melnyk S, Zhou L, Shen R, et al. . Genome-wide DNA methylation profiling using Infinium assay. Epigenomics. 2009;1:177–200 10.2217/epi.09.14 [DOI] [PubMed] [Google Scholar]
  • 23.Catania J, Keenan BC, Margison GP, Fairweather DS. Determination of 5-methylcytosine by acid hydrolysis of DNA with hydrofluoric acid. Anal Biochem. 1987;167:347–51 10.1016/0003-2697(87)90175-8 [DOI] [PubMed] [Google Scholar]
  • 24.Song L, James SR, Kazim L, Karpf AR. Specific method for the determination of genomic DNA methylation by liquid chromatography-electrospray ionization tandem mass spectrometry. Anal Chem. 2005;77:504–10 10.1021/ac0489420 [DOI] [PubMed] [Google Scholar]
  • 25.Weisenberger DJ, Campan M, Long TI, Kim M, Woods C, Fiala E, et al. . Analysis of repetitive element DNA methylation by MethyLight. Nucleic Acids Res. 2005;33:6823–36 10.1093/nar/gki987 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Yuasa Y Epigenetics in molecular epidemiology of cancer a new scope. Adv Genet. 2010;71:211–35 10.1016/B978-0-12-380864-6.00007-9 [DOI] [PubMed] [Google Scholar]
  • 27.Brait M, Ford JG, Papaiahgari S, Garza MA, Lee JI, Loyo M, et al. . Association between lifestyle factors and CpG island methylation in a cancer-free population. Cancer Epidemiol Biomarkers Prev. 2009;18:2984–91 10.1158/1055-9965.EPI-08-1245 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Müller HM, Widschwendter A, Fiegl H, Ivarsson L, Goebel G, Perkmann E, et al. . DNA methylation in serum of breast cancer patients: an independent prognostic marker. Cancer Res. 2003;63:7641–5 [PubMed] [Google Scholar]
  • 29.Tangkijvanich P, Hourpai N, Rattanatanyong P, Wisedopas N, Mahachai V, Mutirangura A. Serum LINE-1 hypomethylation as a potential prognostic marker for hepatocellular carcinoma. Clin Chim Acta. 2007;379:127–33 10.1016/j.cca.2006.12.029 [DOI] [PubMed] [Google Scholar]
  • 30.Teschendorff AE, Menon U, Gentry-Maharaj A, Ramus SJ, Weisenberger DJ, Shen H, et al. . Age-dependent DNA methylation of genes that are suppressed in stem cells is a hallmark of cancer. Genome Res. 2010;20:440–6 10.1101/gr.103606.109 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Breitling LP, Yang R, Korn B, Burwinkel B, Brenner H. Tobacco-smoking-related differential DNA methylation: 27K discovery and replication. Am J Hum Genet. 2011;88:450–7 10.1016/j.ajhg.2011.03.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Terry MB, Delgado-Cruzata L, Vin-Raviv N, Wu HC, Santella RM. DNA methylation in white blood cells: association with risk factors in epidemiologic studies. Epigenetics. 2011;6:828–37 10.4161/epi.6.7.16500 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Pilsner JR, Liu X, Ahsan H, Ilievski V, Slavkovich V, Levy D, et al. . Genomic methylation of peripheral blood leukocyte DNA: influences of arsenic and folate in Bangladeshi adults. Am J Clin Nutr. 2007;86:1179–86 [DOI] [PubMed] [Google Scholar]
  • 34.Hsiung DT, Marsit CJ, Houseman EA, Eddy K, Furniss CS, McClean MD, et al. . Global DNA methylation level in whole blood as a biomarker in head and neck squamous cell carcinoma. Cancer Epidemiol Biomarkers Prev. 2007;16:108–14 10.1158/1055-9965.EPI-06-0636 [DOI] [PubMed] [Google Scholar]
  • 35.Terry MB, Ferris JS, Pilsner R, Flom JD, Tehranifar P, Santella RM, et al. . Genomic DNA methylation among women in a multiethnic New York City birth cohort. Cancer Epidemiol Biomarkers Prev. 2008;17:2306–10 10.1158/1055-9965.EPI-08-0312 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Zhang FF, Morabia A, Carroll J, Gonzalez K, Fulda K, Kaur M, et al. . Dietary patterns are associated with levels of global genomic DNA methylation in a cancer-free population. J Nut. 2011;141:1165–71 10.3945/jn.110.134536 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Wang X, Zhu H, Snieder H, Su S, Munn D, Harshfield G, et al. . Obesity related methylation changes in DNA of peripheral blood leukocytes. BMC Med. 2010;8:87 10.1186/1741-7015-8-87 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Zhang FF, Cardarelli R, Carroll J, Zhang S, Fulda KG, Gonzalez K, et al. . Physical activity and global genomic DNA methylation in a cancer-free population. Epigenetics. 2011;6:293–9 10.4161/epi.6.3.14378 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.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 10.1038/sj.bjc.6605013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.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 10.1158/1055-9965.EPI-08-1237 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Wang YC, Yu ZH, Liu C, Xu LZ, Yu W, Lu J, et al. . Detection of RASSF1A promoter hypermethylation in serum from gastric and colorectal adenocarcinoma patients. World J Gastroenterol. 2008;14:3074–80 10.3748/wjg.14.3074 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Müller HM, Fiegl H, Widschwendter A, Widschwendter M. Prognostic DNA methylation marker in serum of cancer patients. Ann N Y Acad Sci. 2004;1022:44–9 10.1196/annals.1318.008 [DOI] [PubMed] [Google Scholar]
  • 43.Valenzuela MT, Galisteo R, Zuluaga A, Villalobos M, Núñez MI, Oliver FJ, et al. . Assessing the use of p16INK4a promoter gene methylation in serum for detection of bladder cancer. Eur Urol. 2002;42:622–8; discussion 628–30 10.1016/S0302-2838(02)00468-2 [DOI] [PubMed] [Google Scholar]
  • 44.Reibenwein J, Pils D, Horak P, Tomicek B, Goldner G, Worel N, et al. . Promoter hypermethylation of GSTP1, AR, and 14-3-3sigma in serum of prostate cancer patients and its clinical relevance. Prostate. 2007;67:427–32 10.1002/pros.20533 [DOI] [PubMed] [Google Scholar]
  • 45.Ellinger J, Haan K, Heukamp LC, Kahl P, Büttner R, Müller SC, et al. . CpG island hypermethylation in cell-free serum DNA identifies patients with localized prostate cancer. Prostate. 2008;68:42–9 10.1002/pros.20651 [DOI] [PubMed] [Google Scholar]
  • 46.Wong TS, Man MW, Lam AK, Wei WI, Kwong YL, Yuen AP. The study of p16 and p15 gene methylation in head and neck squamous cell carcinoma and their quantitative evaluation in plasma by real-time PCR. Eur J Cancer. 2003;39:1881–7 10.1016/S0959-8049(03)00428-3 [DOI] [PubMed] [Google Scholar]
  • 47.Fujiwara K, Fujimoto N, Tabata M, Nishii K, Matsuo K, Hotta K, et al. . Identification of epigenetic aberrant promoter methylation in serum DNA is useful for early detection of lung cancer. Clin Cancer Res. 2005;11:1219–25 [PubMed] [Google Scholar]
  • 48.Lofton-Day C, Model F, Devos T, Tetzner R, Distler J, Schuster M, et al. . DNA methylation biomarkers for blood-based colorectal cancer screening. Clin Chem. 2008;54:414–23 10.1373/clinchem.2007.095992 [DOI] [PubMed] [Google Scholar]
  • 49.Epigenomics AG. Epigenomics. 2011. http://www.epigenomics.com
  • 50.Zhang YJ, Wu HC, Shen J, Ahsan H, Tsai WY, Yang HI, et al. . Predicting hepatocellular carcinoma by detection of aberrant promoter methylation in serum DNA. Clin Cancer Res. 2007;13:2378–84 10.1158/1078-0432.CCR-06-1900 [DOI] [PubMed] [Google Scholar]
  • 51.Göbel G, Auer D, Gaugg I, Schneitter A, Lesche R, Müller-Holzner E, et al. . Prognostic significance of methylated RASSF1A and PITX2 genes in blood- and bone marrow plasma of breast cancer patients. Breast Cancer Res Treat. 2011;130(1):109–17 10.1007/s10549-010-1335-8 [DOI] [PubMed] [Google Scholar]
  • 52.Domínguez G, Carballido J, Silva J, Silva JM, García JM, Menéndez J, et al. . p14ARF promoter hypermethylation in plasma DNA as an indicator of disease recurrence in bladder cancer patients. Clin Cancer Res. 2002;8:980–5 [PubMed] [Google Scholar]
  • 53.Widschwendter A, Ivarsson L, Blassnig A, Müller HM, Fiegl H, Wiedemair A, et al. . CDH1 and CDH13 methylation in serum is an independent prognostic marker in cervical cancer patients. Int J Cancer. 2004;109:163–6 10.1002/ijc.11706 [DOI] [PubMed] [Google Scholar]
  • 54.Wallner M, Herbst A, Behrens A, Crispin A, Stieber P, Göke B, et al. . Methylation of serum DNA is an independent prognostic marker in colorectal cancer. Clin Cancer Res. 2006;12:7347–52 10.1158/1078-0432.CCR-06-1264 [DOI] [PubMed] [Google Scholar]
  • 55.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 10.1158/0008-5472.CAN-04-2438 [DOI] [PubMed] [Google Scholar]
  • 56.Leung WK, To KF, Chu ES, Chan MW, Bai AH, Ng EK, et al. . Potential diagnostic and prognostic values of detecting promoter hypermethylation in the serum of patients with gastric cancer. Br J Cancer. 2005;92:2190–4 10.1038/sj.bjc.6602636 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Kawakami K, Brabender J, Lord RV, Groshen S, Greenwald BD, Krasna MJ, et al. . Hypermethylated APC DNA in plasma and prognosis of patients with esophageal adenocarcinoma. J Natl Cancer Inst. 2000;92:1805–11 10.1093/jnci/92.22.1805 [DOI] [PubMed] [Google Scholar]
  • 58.Widschwendter A, Müller HM, Fiegl H, Ivarsson L, Wiedemair A, Müller-Holzner E, et al. . DNA methylation in serum and tumors of cervical cancer patients. Clin Cancer Res. 2004;10:565–71 10.1158/1078-0432.CCR-0825-03 [DOI] [PubMed] [Google Scholar]
  • 59.Gifford G, Paul J, Vasey PA, Kaye SB, Brown R. The acquisition of hMLH1 methylation in plasma DNA after chemotherapy predicts poor survival for ovarian cancer patients. Clin Cancer Res. 2004;10:4420–6 10.1158/1078-0432.CCR-03-0732 [DOI] [PubMed] [Google Scholar]
  • 60.Bastian PJ, Palapattu GS, Lin X, Yegnasubramanian S, Mangold LA, Trock B, et al. . Preoperative serum DNA GSTP1 CpG island hypermethylation and the risk of early prostate-specific antigen recurrence following radical prostatectomy. Clin Cancer Res. 2005;11:4037–43 10.1158/1078-0432.CCR-04-2446 [DOI] [PubMed] [Google Scholar]
  • 61.Sunami E, Shinozaki M, Higano CS, Wollman R, Dorff TB, Tucker SJ, et al. . Multimarker circulating DNA assay for assessing blood of prostate cancer patients. Clin Chem. 2009;55:559–67 10.1373/clinchem.2008.108498 [DOI] [PubMed] [Google Scholar]
  • 62.Lee BB, Lee EJ, Jung EH, Chun HK, Chang DK, Song SY, et al. . Aberrant methylation of APC, MGMT, RASSF2A, and Wif-1 genes in plasma as a biomarker for early detection of colorectal cancer. Clin Cancer Res. 2009;15:6185–91 10.1158/1078-0432.CCR-09-0111 [DOI] [PubMed] [Google Scholar]
  • 63.Kaaks R, Stattin P, Villar S, Poetsch AR, Dossus L, Nieters A, et al. . Insulin-like growth factor-II methylation status in lymphocyte DNA and colon cancer risk in the Northern Sweden Health and Disease cohort. Cancer Res. 2009;69:5400–5 10.1158/0008-5472.CAN-08-3020 [DOI] [PubMed] [Google Scholar]
  • 64.Cash HL, Tao L, Yuan JM, Marsit CJ, Houseman EA, Xiang YB, et al. . LINE-1 hypomethylation is associated with bladder cancer risk among non-smoking Chinese. Int J Cancer. 2012;130(5):1151–9 10.1002/ijc.26098 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Wang L, Aakre JA, Jiang R, Marks RS, Wu Y, Chen J, et al. . Methylation markers for small cell lung cancer in peripheral blood leukocyte DNA. J Thorac Oncol. 2010;5:778–85 10.1097/JTO.0b013e3181d6e0b3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Baba S, Yamada Y, Hatano Y, Miyazaki Y, Mori H, Shibata T, et al. . Global DNA hypomethylation suppresses squamous carcinogenesis in the tongue and esophagus. Cancer Sci. 2009;100:1186–91 10.1111/j.1349-7006.2009.01171.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Hou L, Wang H, Sartori S, Gawron A, Lissowska J, Bollati V, et al. . Blood leukocyte DNA hypomethylation and gastric cancer risk in a high-risk Polish population. Int J Cancer. 2010;127:1866–74 10.1002/ijc.25190 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Al-Moundhri MS, Al-Nabhani M, Tarantini L, Baccarelli A, Rusiecki JA. The prognostic significance of whole blood global and specific DNA methylation levels in gastric adenocarcinoma. PLoS ONE. 2010;5:e15585 10.1371/journal.pone.0015585 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Laird PW The power and the promise of DNA methylation markers. Nat Rev Cancer. 2003;3:253–66 10.1038/nrc1045 [DOI] [PubMed] [Google Scholar]
  • 70.Siegmund KD, Laird PW. Analysis of complex methylation data. Methods. 2002;27:170–8 10.1016/S1046-2023(02)00071-3 [DOI] [PubMed] [Google Scholar]
  • 71.National Cancer Institute. Early Detection Research Network. http://edrn.nci.nih.gov/
  • 72.Kagan J, Srivastava S, Barker PE, Belinsky SA, Cairns P. Towards Clinical Application of Methylated DNA Sequences as Cancer Biomarkers: A Joint NCI's EDRN and NIST Workshop on Standards, Methods, Assays, Reagents and Tools. Cancer Res. 2007;67:4545–9 10.1158/0008-5472.CAN-06-2888 [DOI] [PubMed] [Google Scholar]
  • 73.Hudson JI, Pope HG Jr, Glynn RJ. The cross-sectional cohort study: an underutilized design. Epidemiology. 2005;16:355–9 10.1097/01.ede.0000158224.50593.e3 [DOI] [PubMed] [Google Scholar]
  • 74.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 10.1073/pnas.93.18.9821 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.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 10.1073/pnas.89.5.1827 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Dejeux E, Audard V, Cavard C, Gut IG, Terris B, Tost J. Rapid identification of promoter hypermethylation in hepatocellular carcinoma by pyrosequencing of etiologically homogeneous sample pools. J Mol Diagn. 2007;9:510–20 10.2353/jmoldx.2007.060209 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Xiong Z, Laird PW. COBRA: a sensitive and quantitative DNA methylation assay. Nucleic Acids Res. 1997;25:2532–4 10.1093/nar/25.12.2532 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Matsubayashi H, Sato N, Brune K, Blackford AL, Hruban RH, Canto M, et al. . Age- and disease-related methylation of multiple genes in nonneoplastic duodenum and in duodenal juice. Clin Cancer Res. 2005;11:573–83 [PubMed] [Google Scholar]
  • 79.Gonzalgo ML, Liang G. Methylation-sensitive single-nucleotide primer extension (Ms-SNuPE) for quantitative measurement of DNA methylation. Nat Protoc. 2007;2:1931–6 10.1038/nprot.2007.271 [DOI] [PubMed] [Google Scholar]
  • 80.Tierling S, Schuster M, Tetzner R, Walter J. A combined HM-PCR/SNuPE method for high sensitive detection of rare DNA methylation. Epigenetics Chromatin. 2010;3:12 10.1186/1756-8935-3-12 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Eads CA, Danenberg KD, Kawakami K, Saltz LB, Blake C, Shibata D, et al. . MethyLight: a high-throughput assay to measure DNA methylation. Nucleic Acids Res. 2000;28:E32 10.1093/nar/28.8.e32 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Bonanno C, Shehi E, Adlerstein D, Makrigiorgos GM. MS-FLAG, a novel real-time signal generation method for methylation-specific PCR. Clin Chem. 2007;53:2119–27 10.1373/clinchem.2007.094011 [DOI] [PubMed] [Google Scholar]
  • 83.Akama TO, Okazaki Y, Ito M, Okuizumi H, Konno H, Muramatsu M, et al. . Restriction landmark genomic scanning (RLGS-M)-based genome-wide scanning of mouse liver tumors for alterations in DNA methylation status. Cancer Res. 1997;57:3294–9 [PubMed] [Google Scholar]
  • 84.Ando Y, Hayashizaki Y. Restriction landmark genomic scanning. Nat Protoc. 2006;1(6):2774–83 10.1038/nprot.2006.350 [DOI] [PubMed] [Google Scholar]
  • 85.Huang TH, Laux DE, Hamlin BC, Tran P, Tran H, Lubahn DB. Identification of DNA methylation markers for human breast carcinomas using the methylation-sensitive restriction fingerprinting technique. Cancer Res. 1997;57:1030–4 [PubMed] [Google Scholar]
  • 86.Weber M, Davies JJ, Wittig D, Oakeley EJ, Haase M, Lam WL, et al. . Chromosome-wide and promoter-specific analyses identify sites of differential DNA methylation in normal and transformed human cells. Nat Genet. 2005;37:853–62 10.1038/ng1598 [DOI] [PubMed] [Google Scholar]
  • 87.Li M, Hu SL, Shen ZJ, He XD, Tao SN, Dong L, et al. . High-performance capillary electrophoretic method for the quantification of global DNA methylation: application to methotrexate-resistant cells. Anal Biochem. 2009;387:71–5 10.1016/j.ab.2008.12.033 [DOI] [PubMed] [Google Scholar]
  • 88.Radpour R, Barekati Z, Kohler C, Lv Q, Bürki N, Diesch C, et al. . Hypermethylation of tumor suppressor genes involved in critical regulatory pathways for developing a blood-based test in breast cancer. PLoS ONE. 2011;6:e16080 10.1371/journal.pone.0016080 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Wong TS, Kwong DL, Sham JS, Wei WI, Kwong YL, Yuen AP. Quantitative plasma hypermethylated DNA markers of undifferentiated nasopharyngeal carcinoma. Clin Cancer Res. 2004;10:2401–6 10.1158/1078-0432.CCR-03-0139 [DOI] [PubMed] [Google Scholar]
  • 90.Hoffmann AC, Kaifi JT, Vallböhmer D, Yekebas E, Grimminger P, Leers JM, et al. . Lack of prognostic significance of serum DNA methylation of DAPK, MGMT, and GSTPI in patients with non-small cell lung cancer. J Surg Oncol. 2009;100:414–7 10.1002/jso.21348 [DOI] [PubMed] [Google Scholar]
  • 91.Ramirez JL, Rosell R, Taron M, Sanchez-Ronco M, Alberola V, de Las Peñas R, et al. . 14-3-3{sigma} methylation in pretreatment serum circulating DNA of cisplatin-plus-gemcitabine-treated advanced non-small-cell lung cancer patients predicts survival: The Spanish Lung Cancer Group. J Clin Oncol. 2005;23:9105–12 10.1200/JCO.2005.02.2905 [DOI] [PubMed] [Google Scholar]
  • 92.Hoque MO, Feng Q, Toure P, Dem A, Critchlow CW, Hawes SE, et al. . Detection of aberrant methylation of four genes in plasma DNA for the detection of breast cancer. J Clin Oncol. 2006;24:4262–9 10.1200/JCO.2005.01.3516 [DOI] [PubMed] [Google Scholar]
  • 93.deVos T, Tetzner R, Model F, Weiss G, Schuster M, Distler J, et al. . Circulating methylated SEPT9 DNA in plasma is a biomarker for colorectal cancer. Clin Chem. 2009;55:1337–46 10.1373/clinchem.2008.115808 [DOI] [PubMed] [Google Scholar]
  • 94.Melnikov AA, Scholtens D, Talamonti MS, Bentrem DJ, Levenson VV. Methylation profile of circulating plasma DNA in patients with pancreatic cancer. J Surg Oncol. 2009;99:119–22 10.1002/jso.21208 [DOI] [PubMed] [Google Scholar]
  • 95.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;30:2489–96 [PMC free article] [PubMed] [Google Scholar]
  • 96.Wilhelm CS, Kelsey KT, Butler R, Plaza S, Gagne L, Zens MS, et al. . Implications of LINE1 methylation for bladder cancer risk in women. Clin Cancer Res. 2010;16:1682–9 10.1158/1078-0432.CCR-09-2983 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Pinheiro H, Bordeira-Carriço R, Seixas S, Carvalho J, Senz J, Oliveira P, et al. . Allele-specific CDH1 downregulation and hereditary diffuse gastric cancer. Hum Mol Genet. 2010;19:943–52 10.1093/hmg/ddp537 [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Journal of Epidemiology are provided here courtesy of Japan Epidemiological Association

RESOURCES