Abstract
Epigenome-wide studies of DNA methylation using blood-derived DNA from cancer patients are complicated by the heterogeneity of cell types within blood and the associated cell lineage specification of DNA methylation signatures. Here, we applied a novel set of analytic approaches to assess the association between cancer case-status and DNA methylation adjusted for leukocyte variation using blood specimens from three case-control cancer studies (bladder: 223 cases, 205 controls; head and neck: 92 cases, 92 controls; and ovarian: 131 cases, 274 controls). Using previously published data on leukocyte-specific CpG loci and a recently described approach to deconvolute subject-specific blood composition, we performed an epigenome-wide analysis to examine the association between blood-based DNA methylation patterns and each of the three aforementioned solid tumor types adjusted for cellular heterogeneity in blood. After adjusting for leukocyte profile in our epigenome-wide analysis, the omnibus association between case-status and methylation was significant for all three studies (bladder cancer: P = 0.047; HNSCC: P = 0.013; ovarian cancer: P = 0.0002). Subsequent analyses revealed that CpG sites associated with cancer were enriched for transcription factor binding motifs involved with cancer-associated pathways. These results support the existence of cancer-associated DNA methylation profiles in the blood of solid tumor patients that are independent of alterations in normal leukocyte distributions. Adoption of the methods developed here will make it feasible to rigorously assess the influence of variability of normal leukocyte profiles when investigating cancer related changes in blood-based epigenome-wide association studies.
Keywords: EWAS, epigenomics, bladder cancer, HNSCC, ovarian cancer
Introduction
Genome-wide association studies (GWAS) have impacted our comprehension of genetic susceptibilities to a broad variety of complex diseases. However, DNA sequence variations identified through GWAS are estimated to account for less than 30% of phenotypic variability in humans,1 and explain only a small portion of familial cancer risk.2 Epigenetic mechanisms likely account for some of this variability and this possibility has been a catalyst for the development of epigenome-wide association studies (EWAS).3 In particular, the involvement of epigenetics in carcinogenesis is widely appreciated.4 There is intense interest in DNA methylation as cancer biomarkers, owing to the chemical stability of these marks, their involvement in crucial genomic functions, as well as the potential for modification by clinically relevant environmental exposures.
Despite the superficial similarities between GWAS and EWAS, there are fundamental differences between genetic and epigenetic variation that must be considered.3 When using blood derived DNA, different cell lineages show striking differences regarding epigenetic marks,5-11 which is in sharp contrast to the uniform allelic variation in SNPs among different blood cell types. This means that global epigenetic patterns are complex functions of normal blood cell development.12,13 However, there is also evidence that exposures to endogenous and exogenous agents can alter DNA methylation14-16 and may differentially affect tissues depending on the degree of contact with the tissue, cellular proliferation rate, and other characteristics impacting susceptibility to epigenetic insult. Since personal characteristics (e.g., age, gender, and race), behaviors (e.g.. smoking and alcohol consumption), exogenous exposures, and pathologic states can modulate the circulating immune profile,17-23 there is a high potential for confounding by inter-subject immune variations. Cell lineage specification of DNA methylation has not been formally and rigorously interrogated until now in the EWAS literature. Given that EWAS is inherently more confounded by the mixtures of cells in samples compared with GWAS, new analytical strategies must be developed to simultaneously assess cell lineage epigenetic patterns when searching for alterations that are disease and/or exposure related.
In order to evaluate the influence of leukocyte distribution on DNA methylation it is necessary to access the relevant epigenetic subtypes within blood. There are three general approaches in evaluating proportions of circulating leukocytes24: manual hematological cytology, automated cell counting with differential, and antibody-based flow cytometry (FACS). The first two approaches are widely used clinically but do not delineate epigenetically distinct lymphocyte subsets, such as T cells, B cells or natural killer (NK) cells, or T cell subsets, including CD4+, CD8+, and regulatory T cells (Treg). FACS has the potential to delineate these and other subsets but generally requires fresh blood samples and is too costly and laborious for routine comprehensive blood profiling.25-28 Moreover, as all of these approaches require intact cells, they cannot be applied to studies in which intact cells are not accessible or only extracted DNA is available. In response to these limitations, and because so few population data sets for EWAS include detailed cell type information, we have developed a relevant immune cell epigenetic reference library and statistical approach6 that can be readily applied to peripheral blood methylomic data (which is particularly well suited for this purpose due to its relative stability and strong role in differentiation) for evaluating leukocyte subtypes. This approach, which has been independently validated,29 renders the aforementioned hematological methods unnecessary for direct ascertainment of subtype data from populations under study, and allows for application to peripheral blood DNA (whereas the latter approaches all require intact cells). We judge that these issues urgently need attention as demonstrated by numerous recent reports of cancer-related methylation profiles from blood in patients with various solid tumor types, including breast,30 ovarian,31,32 pancreatic,33 bladder,34 head and neck,35 colorectal,36 and lung cancers.37 A very recent paper showed genetic risk for the immune disorder rheumatoid arthritis was mediated by DNA methylation, but this association was only observed after adjusting for normal leukocyte distributions.38 As observed cancer-associated methylation in these studies may be substantially driven by cancer-associated shifts in the proportions of circulating leukocytes,6,8 it remains unclear if additional variation in DNA methylation can be detected in blood that is independent of interpersonal leukocytic variability.
Here we present a novel approach to blood-based EWAS in three independent case-control studies of solid tumor patients designed to assess cancer-associated methylation states that arise independently of shifts in relative leukocyte proportions in the blood. Our data suggest that using lineage-specific DNA methylation information to account for leukocyte composition can overcome confounding by cell mixtures, and promises to greatly improve blood-based epigenome-wide association studies.
Results
EWAS were performed on three separate previously described population-based case-control studies of bladder cancer (223 cases, 205 controls),39,40 head and neck squamous cell carcinoma (HNSCC; 92 cases, 92 controls),35,41 and a publicly available data set for ovarian cancer (131 cases and 274 controls).32 A description of each of the three study populations is provided in Table 1.
Table 1. Description of each of the three study populations by case-control status.
| Bladder Cancer (New Hampshire) | HNSCC (greater Boston) | Ovarian Cancer (United Kingdom) | |||||||||
|---|---|---|---|---|---|---|---|---|---|---|---|
| Cases (n = 223) |
Controls (n = 205) |
P value | Cases (n = 92) |
Controls (n = 92) |
P value | Cases (n = 131) |
Controls (n = 274) |
P value | |||
| Age, median years (range) | 66.0 (25–74) | 64.0 (28–74) | 0.05a | 58.0 (31–84) | 59.0 (32–86) | 0.54a | 67.0 (49–91) | 64.0 (52–78) | 0.08a | ||
| Gender | |||||||||||
| Female | 52 (23.3%) | 65 (31.7%) | 0.06b | 28 (30.4%) | 28 (30.4%) | >0.99b | 266 (100%) | 274 (100%) | >0.99b | ||
| Male | 171 (76.7%) | 140 (68.3%) | 64 (69.6%) | 64 (69.6%) | 0 (0.0%) | 0 (0.0%) | |||||
| Race | |||||||||||
| White | 223 (100%) | 205 (100%) | >0.99b | 84 (91.3%) | 85 (92.4%) | >0.99b | — | — | — | ||
| Non-White | 0 (0.0%) | 0 (0.0%) | 8 (8.7%) | 7 (7.6%) | — | — | |||||
| Cigarette smoking | |||||||||||
| Never-smoker | 40 (17.9%) | 58 (28.3%) | <0.001b | 17 (18.5%) | 32 (34.8%) | 0.04b | — | — | — | ||
| Former smoker | 111 (49.8%) | 112 (54.6%) | 59 (64.1%) | 47 (51.1%) | — | — | |||||
| Current smoker | 72 (32.3%) | 35 (17.1%) | 16 (17.4%) | 13 (14.1%) | |||||||
| HPV serology (E6, E7 or L1) | |||||||||||
| Negative | — | — | — | 66 (71.7%) | 83 (90.2%) | 0.002b | — | — | — | ||
| Positive | — | — | 26 (28.3%) | 9 (9.8%) | — | — | |||||
Abbreviations: HNSCC = head and neck squamous cell carcinoma; aWilcoxon rank-sum test for a difference between cases and controls; bFisher's exact test for a difference between cases and controls.
Cancer-associated differential methylation
For each study, differential methylation between cases and controls was examined among all autosomal loci (26 486 CpG sites) and among CpGs that did not significantly vary between leukocyte types (leukocyte-specific differentially methylated positions [DMPs]). After exclusion of 9849 putative leukocyte DMPs and adjustment for false-discovery rate,42 we observed 3,987 differentially methylated loci (Q ≤ 0.05) for bladder cancer cases relative to controls (Table S1), representing 24% of the 16 637 non-leukocyte-specific loci. Of the 3987 differentially methylated loci, 2105 loci were observed to be hypomethylated among bladder cancer cases, with a median methylation β-value that was 0.017 lower (roughly corresponding to a 2% decrease in methylation), and 1882 were hypermethylated, with a median β-value that was 0.032 higher among these cases compared with controls. A much lower proportion of non-leukocyte-specific loci were found to be differentially methylated in peripheral blood between the HNSCC cases and controls: after adjusting for multiple comparisons, there were only 4 significantly differentially methylated CpGs (Q ≤ 0.05), all of which were hypomethylated in cases, with a median difference in β-value of 0.015, constituting less than 0.1% of total non-leukocyte specific CpGs. Importantly, the sample-size for the HNSCC study (n = 184) was much smaller than for the bladder cancer study (n = 428) impacting our power to detect significant differences. Likewise, a smaller number of differentially methylated loci were detected between ovarian cancer cases and controls, with 17 differentially methylated loci (Q ≤ 0.05). Of these, 9 were hypomethylated in cases, with a median difference in methylation β-value that was 0.009 lower, and 17 were hypermethylated with a median difference that was 0.003 higher.
By contrast, the frequency of differentially methylated loci changed when differential methylation was assessed for all 26,486 autosomal CpG loci (Table S2). This was particularly evident for the HNSCC and ovarian cancer studies.
Leukocyte-adjusted epigenome-wide association studies
Since potentially important information may be lost through simple exclusion of putative DMPs, we applied an approach to blood-based EWAS that takes into account all autosomal CpG loci, while adjusting for leukocyte composition using subject-specific estimates inferred through epigenetic deconvolution of blood (Fig. 1).6 The omnibus cancer-specific associations with DNA methylation in blood (Fig. 2A) were significant for all three studies, both prior to adjusting for leukocyte composition (β) and after adjusting for leukocyte composition (α). However, the volcano plots contrasting effect size and significance level for individual loci for each respective study (Figs. 2B–D) clearly illustrate the difference in results that were obtained before and after adjusting for leukocyte composition (if there were no difference, the points would perfectly overlap). This was not unexpected, given that a significant cancer-associated difference in circulating immune profile (Г) was observed in all three studies. Cancer-associated differences in the proportions of the specific leukocyte types are presented in Figure 3. Further, we conducted sensitivity analyses to rule out the possibility that treatment or racial/ethnic differences were driving the observed case-associated blood methylation for bladder cancer and HNSCC, respectively. We excluded all non-White subjects in the HNSCC study (7 cases, 8 controls; all bladder cancer subjects were White, and race was not available for the publicly available UK ovarian cancer cohort) and all bladder cancer cases that received treatment other than surgery (55 cases; all ovarian and HNSCC blood draws occurred prior to treatment), which yielded similar results (Figs. S1 and S2).

Figure 1. Schematic of epigenome-wide association study (EWAS) approaches using blood. (A) Typical EWAS approach using blood, where the marginal effect of an exposure on blood DNA methylation is examined without accounting for the effect leukocyte distribution on the methylation profile. (B) Leukocyte-adjusted EWAS approach to examine the direct effect of an exposure or phenotype (e.g., case-status) on DNA methylation in blood.
Figure 2. Epigenome-wide analysis of cancer-associated DNA methylation and shifts in leukocyte composition in peripheral blood for bladder, head and neck (HNSCC) and ovarian cancers. (A) Omnibus P values for cancer-associated DNA methylation relative to control subjects, unadjusted (β) and adjusted (α) for leukocyte composition, and cancer-associated shifts in relative leukocyte proportions in the blood (Γ). Leukocyte-adjusted (α; depicted as black diamonds) and –unadjusted (β; blue open circles) associations for individual CpG loci for each respective solid tumor type are presented in the volcano plots (B–D), which depict –log(P values) vs. regression coefficients for case-status adjusted for age. The models for bladder cancer and HNSCC were additionally adjusted for sex and smoking habit. The horizontal red line on each plot represents the –log value where P = 0.05. Note: β = Association between case-status and methylation without adjustment for leukocyte composition; α = Association between case-status and methylation adjusted for leukocyte composition; Γ = Association between case-status and leukocyte composition
Figure 3. (A) Relative percent differences for each leukocyte subtype are for cases compared with controls in studies of bladder cancer, HNSCC, and ovarian cancer. (B) Average leukocyte proportions in the control subjects from each respective case-control study. Relative leukocyte proportions were determined by applying the deconvolution methods of Houseman et al. (2012) to the peripheral blood methylation profile of each study subject. Note: *P ≤ 0.05; **P ≤ 0.01; ***P ≤ 0.001; ****P ≤ 0.0001; Black error bars represent the standard error for each estimate. Bladder cancer and HNSCC estimates are adjusted for age, gender, and smoking status (current/former/never); ovarian cancer estimates are adjusted for age.
The top ten CpG loci (based on ranked absolute α coefficient for each respective leukocyte-adjusted EWAS) are presented in Table 2, along with CpG island status, distance from transcription start site and information on the associated gene. There were no individual CpGs common to all three cancers among their respective top 50 loci (Table S3), although bladder and ovarian cancer each had one of their top hits located within the promoter region of DPPA3 (albeit at separate loci); this gene encodes developmental pluripotency-associated protein 3, which is an enzyme associated with embryonic development and stem cell differentiation.43
Table 2. Top 10 cancer-associated CpG loci in blood for bladder, head and neck (HNSCC) and ovarian cancer after adjustment for leukocyte composition, based on ranked test statistic.
| Locus ID | Regression Coefficient (α) | Chromosome | Located in a CpG Island |
Distance from TSSa (bases) | Associated Gene |
Transcription Factor |
Associated Gene Descriptionb | CpG-me Correlationc with Gene Expression in Tumor (P value) |
|
|---|---|---|---|---|---|---|---|---|---|
| Bladder Cancer | |||||||||
| cg07510052 | +0.020 | 5 | Yes | 41 | HIGD2A | No | Proposed subunit of cytochrome c oxidase | n/a | |
| cg06572974 | -0.057 | 15 | No | 1200 | CHD2 | Yes | Alters expression through chromatin structure modifications. Alternatively spliced transcript variants encoding distinct isoforms have been found | ρ = 0.18 (0.02) | |
| cg00429618 | +0.014 | 9 | Yes | 440 | ZNF322B | No | Zinc Finger Protein 322 Pseudogene | ρ = 0.04 (0.62) | |
| cg25307081 | +0.031 | 16 | Yes | 140 | BRD7 | Yes | Component of the SWI/SNF chromatin-remodeling complex. Alternative splicing results in multiple transcript variants | n/a | |
| cg07081888 | +0.040 | 19 | Yes | 470 | NALP4 | No | Involved in the activation of proinflammatory caspases | n/a | |
| cg08950375 | +0.031 | 2 | No | 580 | COL6A3 | No | Encodes the α-3 chain of type VI collagen | ρ = -0.03 (0.68) | |
| cg09425215 | -0.027 | 15 | Yes | 428 | CHD2 | Yes | See above | n/a | |
| cg20938689 | +0.031 | 5 | Yes | 131 | COX7C | No | Catalyzes the electron transfer from reduced cytochrome C to oxygen | n/a | |
| cg00461841 | -0.038 | 16 | No | 760 | ATF7IP2 | No | Recruiter that couples transcriptional factors to general transcriptional machinery apparatus, modulating transcription regulation and chromatin formation |
ρ = 0.18 (0.02) | |
| cg20150565 | -0.026 | 20 | Yes | 743 | ZNFX1 | Yes | NFX1-Type Zinc Finger-Containing Protein 1 | ρ = 0.06 (0.47) | |
| HNSCC | |||||||||
| cg03835292 | -0.034 | 22 | Yes | 607 | LGALS2 | No | Binds β-galactoside. Function is presently unknown. | ρ = 0.08 (0.37) | |
| cg05473677 | -0.034 | 3 | Yes | 330 | OSTalpha | No | heterodimerizes with OSTbeta for bile acid export from enterocytes into blood | ρ = -0.05 (0.60) | |
| cg08241785 | 0.022 | 5 | No | 41 | F2RL2 | No | Plays an essential role in hemostasis and thrombosis | ρ = -0.02 (0.84) | |
| cg00725635 | 0.022 | 1 | No | 82 | B3GALT2 | No | Membrane-bound glycoprotein with diverse enzymatic functions | ρ = -0.03 (0.76) | |
| cg17628717 | -0.020 | 7 | Yes | 154 | HECW1 | No | Mediates ubiquitylation and subsequent degradation of DVL1 | ρ = -0.08 (0.38) | |
| cg11435943 | -0.022 | 18 | No | 706 | SERPINB12 | No | Inhibitor of trypsin and plasmin. Reported to be downregulated in oral squamous cell carcinoma (Shiiba M et al. [2010]). | ρ = 0.12 (0.17) | |
| cg02449608 | -0.014 | 19 | No | 696 | C19orf18 | No | Uncharacterized | ρ = 0.04 (0.64) | |
| cg25943702 | -0.026 | 22 | No | 99 | BRD1 | No | Component of the MOZ/MORF complex which has a histone H3 acetyltransferase activity |
ρ = -0.03 (0.75) | |
| cg22346765 | -0.019 | 6 | No | 594 | UNC5CL | No | Inhibits NF-kappa-B-dependent transcription | ρ = -0.02 (0.82) | |
| cg03266453 | 0.020 | 2 | Yes | 296 | EN1 | Yes | Homeobox protein involved in developmental control | ρ = 0.12 (0.19) | |
| Ovarian Cancer | |||||||||
| cg01980222 | -0.032 | 6 | No | 3 | TREM2 | No | Forms a receptor signaling complex with TYROBP and triggers activation of the immune responses in macrophages and dendritic cells. Alternative splicing results in multiple transcript variants encoding different isoforms | ρ = 0.06 (0.31) | |
| cg24211388 | -0.041 | 6 | No | 196 | AIF1 | No | Involved in vascular inflammation and phagocytosis and thought to be involved in negative regulation vascular smooth muscle cell growth. Overexpressed in recurrent ovarian tumors (Jinawath et al. [2010]). Three transcript variants encoding different isoforms have been found |
ρ = 0.07 (0.29) | |
| cg00974864 | -0.039 | 1 | No | 152 | FCGR3B | No | Low affinity receptor for the Fc region of gamma immunoglobulins (IgG). Several transcript variants encoding different isoforms have been found | ρ = -0.09 (0.13) | |
| cg06872381 | -0.046 | 12 | Yes | 13 | DPPA3 | No | Involved in epigenetic chromatin reprogramming in the zygote | ρ = 0.15 (0.01) | |
| cg06543018 | +0.020 | 3 | Yes | 332 | RBP1 | No | Involved in the transport of retinol from the liver storage site to peripheral tissue. Decreased expression has been associated with malignant transformation of ovarian epithelium (Roberts D et al. [2002]). Multiple transcript variants encoding different isoforms have been found | ρ = -0.14 (0.02) | |
| cg26205131 | +0.020 | 5 | Yes | 667 | SLC6A3 | No | Dopamine transporter with reported SNP associations with body mass index (Azzato EM et al. [2009]) |
ρ = -0.23 (0.0001) | |
| cg12910797 | -0.055 | 17 | Yes | 88 | HOXB3 | Yes | Homeobox protein involved in developmental control | ρ = -0.23 (0.0002) | |
| cg19910382 | -0.050 | 2 | No | 17 | FABP1 | No | Involved in fatty acid uptake, transport, and metabolism | ρ = 0.10 (0.11) | |
| cg00645579 | -0.037 | 11 | Yes | 1213 | IRF7 | Yes | Key regulator of type I interferon (IFN)-dependent immune responses. Has been identified as a BRCA1 transcriptional target (Buckley NE et al. [2007]). Multiple transcript variants have been identified | ρ = -0.17 (0.005) | |
| cg09964921 | -0.034 | 21 | No | 1194 | KCNE1 | No | Involved in potassium ion channel formation. Alternative transcript variants have been identified |
ρ = 0.10 (0.12) | |
a Absolute distance in bases from transcription start site; bDescription adapted from www.GeneCards.org; cMethylation-expression Spearman correlation coefficients (ρ) for tumor were calculated using Infinium 450K HumanMethylation and mRNA-Seq v2 data publicly available through The Cancer Genome Atlas (http://cancergenome.nih.gov/). Paired methylation-expression data was available for 165 bladder cancers, 133 HNSCC, and 258 ovarian cancers.
Motivated by the hypothesis that altered DNA methylation observed in cases may be specifically targeting genes with common sequence features, we evaluated the DNA sequences within 1kb of the CpG dinucleotides associated with altered DNA methylation (after leukocyte adjustment) for the presence of transcription factor binding sites (TFBS). There were 29 TFBS that were overrepresented in the bladder cancer study, 27 for the HNSCC study, and 33 for the ovarian cancer study. Seventeen TFBS were overrepresented in at least two of the studies, while one of the overrepresented TFBS was common to all three cancers (EVI1). The entirety of observed overrepresented TFBS proximal to cancer-associated loci is presented in Figure 4.

Figure 4. Overrepresentation of transcription factor binding sites (TFBS) within 1kb of cancer-associated CpG loci (after leukocyte-adjustment) in the blood of patients with bladder, head and neck (HNSCC), and ovarian cancer. Each circle depicts overrepresented TFBS motifs for each respective cancer; those listed in overlapping regions indicate overrepresented TFBS common to two or more of the cancer types.
Discussion
We have presented an approach for conducting blood-based EWAS in solid tumor studies that, along with the approaches recently described by Liu et al.38 and Elliott et al.44 for non-cancer associated disease and smoking exposure, respectively, offers a practical solution for dealing with cellular heterogeneity of blood samples in EWAS, advancing the methodology in this field. In applying these methods, we have provided support for the existence of cancer-specific methylation patterns in the blood of bladder cancer, HNSCC, and ovarian cancer patients that were independent of differences in leukocyte profile. Our observation of cancer-specific DNA methylation profiles in blood is in line with other reports of such findings in the literature.8,30-34,36,37 Importantly, our results suggest that differential methylation patterns in the blood of patients with solid tumors may arise as a result of biological mechanisms beyond what can be attributed to differential methylation between the major normal leukocyte cell types. It should be noted that it is presently unknown to what degree the observed differences in blood reflect the methylation patterns of the tumor. Further, while these findings are provocative, the cross-sectional measurement of blood methylation at diagnosis does not allow us to draw any definitive conclusions on the temporality of the observed epigenetic marks, although a recent prospective study identified differentially methylated loci in the blood of breast cancer patients that preceded diagnosis.45 As such, the source of the observed cancer-specific methylation differences remains unclear at the present.
While it is accepted that circulating tumor DNA is detectable in the blood stream of a subset of patients with solid tumors, including bladder cancer,46-51 HNSCC,52-55 and ovarian cancer,56-59 it is improbable that the observed methylation differences in peripheral blood between cancer cases and controls are driven by this phenomenon, particularly in the absence of enrichment for free DNA in the plasma, as circulating tumor DNA, when present, occurs at an extremely low concentration in blood60 and, thus, would have a negligible impact on the methylation signature of peripheral blood.
The analytic applications presented here required high-density epigenome-wide methylation data from sorted non-pathologic human leukocyte samples for the identification of putative leukocyte lineage-specific DMPs, along with corresponding epigenomic data for the peripheral blood samples from the case-control studies for conduct of the EWAS. Even though our analyses and epigenetic library were derived from the Infinium 27K BeadArray, approximately 94% of the loci from the 27K platform are featured on the Infinium 450K BeadArray platform, and any of the loci derived from the array could be extended to sequencing data based on their annotated genomic coordinates. Our findings were bolstered by the population-based nature of the case-control studies, strengthening the generalizability, and by the availability of well-characterized epidemiologic data, particularly for the bladder cancer and HNSCC study populations. These attributes made it possible to control for possible confounding by exposure and/or personal characteristics related to the cancer of interest.
Among the potential limitations of the study was the cross-sectional procurement of blood samples upon study enrollment. While the case-control study design allows us to identify cancer-associated DNA methylation in blood, the lack of temporality precludes us from knowing whether the observed differences in methylation preceded the development of the disease or if they occurred as a direct or indirect result of the cancer itself. Prospective studies are needed to establish temporality and determine the time frame for which epigenetic alterations occur in blood during the carcinogenic process of solid tumors. Additionally, the moderate sample size may have limited our statistical power to detect associations (particularly for the HNSCC study). Further, while the focus of this manuscript was the application of a practical EWAS approach to account for cellular heterogeneity, independent validation of the findings reported herein for each of the three solid tumor types should be performed in future studies. Further, although we have identified differentially methylated loci across major leukocyte types (i.e., CD4+ T cells, CD8+ T cells, NK cells, B cells, monocytes, and granulocytes), we cannot rule out additional epigenetic distinctions, either from unmeasured subpopulations or activated versions of measured cell types (including possible cancer-specific immune subtypes or compartments) as a source of residual confounding. While several of the top loci are situated within cancer-associated genes, many others lie within genes with known immune function, raising the prospect that they are unaccounted DMRs in unmeasured leukocyte subtypes. This may be best exemplified by the presence of a top 10 loci for ovarian cancer that is associated with TREM2, a gene with known involvement in immune activation of macrophages and dendritic cells.61 It is also important to note that cancer-specific changes that occur solely in a minor subtype may not be detected using this method. However, these would also not likely be detected in unadjusted assessments in the absence of isolation and purification of the single subtype for all study subjects. Finally, the vast majority of participants for the three studies presented herein were Caucasian, which may affect the generalizability of our findings to other races/ethnicities. Future studies should seek to incorporate additional racial/ethnic subgroups to determine if these findings apply across different populations, as well as extending similar research to other solid tumor types, particularly those with reported alterations in blood methylation.
Our findings underscore the importance of adjusting for interpersonal leukocyte variability in blood-based EWAS. The methods described herein present new opportunities for researchers to account for such variations for the conduct of these studies, particularly as large international consortiums are underway to establish epigenetic roadmaps of human cells,62 from which similar information on DMPs can be derived. This is particularly important, as blood is commonly collected and archived as part of many epidemiologic studies, and thus the ability to accurately analyze disease associated DNA methylation modifications may open new doors for biomarker discovery or individual risk assessment. Elucidation of the mechanisms driving cancer-associated methylation changes in peripheral blood and their impact on cancer risk will enhance our understanding of the epigenetic contribution to the carcinogenic process and will better inform biomedical scientists and clinicians as to how to use and interpret blood-based methylation data.
Methods
Study populations
Epigenome-wide methylation analyses were performed on three separate population-based case-control cancer studies. The first included 223 incident bladder cancer cases, identified from the NH State Cancer Registry from July 1, 1994 through June 30, 1998 as part of a previously described study,39,40 with available blood samples and 205 healthy population-based control subjects identified through public records, also with available blood samples and no prior history of cancer (Gene Expression Omnibus [GEO] accession number: GSE50409). Upon enrollment, consenting subjects underwent personal interviews furnishing sociodemographic, health behavior and exposure information. Blood was collected for cases on average within 1-y of diagnosis; however only 24.7% of cases (n = 55) received any treatment beyond surgery (i.e., radiation, BCG, or chemotherapy). The second study consisted of 92 incident head and neck squamous cell carcinoma (HNSCC) cases and 92 cancer-free control subjects with available blood samples (GEO: GSE30229), randomly selected from a previously described case-control study of head and neck cancer.35,41 Briefly, the study was comprised of incident head and neck cancer patients from the greater Boston area and population-based controls from the same region with no prior history of cancer, frequency-matched on age (+/− 3 y), gender, and town of residence. Subjects completed a self-administered questionnaire that provided data on sociodemographic, health behavior and exposure history. All case blood draws were performed prior to initiation of radiation or chemotherapy. Serologic HPV16 testing for E6, E7, and L1 viral proteins was performed on all subjects using sandwich ELISA assays for antibody detection, as previously described.63,64 The third study was obtained from a publicly available Infinium HumanMethylation27 blood methylation data set (GEO: GSE19711) and included 266 postmenopausal women diagnosed with primary epithelial ovarian cancer (131 cases collected pre-treatment and 135 post-treatment) and 274 postmenopausal cancer-free controls collected as part of the United Kingdom Ovarian Cancer Population Study (UKOPS).32 Of the ovarian cancer cases, only the 131 pre-treatment cases were used in our analyses; all cases with samples collected post-treatment were excluded.
All cases and controls enrolled in the study provided written informed consent as approved by the Institutional Review Boards of the participating institutions.
DNA methylation array
DNA was extracted from peripheral blood samples from the bladder cancer and HNSCC case-control studies using the DNeasy Blood and Tissue Kit (Qiagen) and sodium bisulfite-converted using the EZ DNA methylation kit (Zymo Research) according to manufacturer’s protocols. Methylation was measured using the Infinium HumanMethylation27 BeadArray (Illumina), which interrogates 27 578 CpG loci across 14 495 genes, at the University of California San Francisco Institute for Human Genomics Core Facility. Outliers were detected using array control probes supplied by Illumina to diagnose problems such as poor bisulfite conversion, batch or beadchip effect or color-specific problems. Specifically, Mahalanobis distances between arrays were determined based on fitted mean vector and variance-covariance matrix, and arrays with large distances inconsistent with multivariate normality were discarded. The methylation status for each individual CpG locus was calculated as the ratio of fluorescent signals, ranging from 0 to 1, using the average probe intensity for the methylated (M) and unmethylated (U) alleles.
Statistical analysis
Identification of putative leukocyte DMPs
Putative leukocyte-specific DMPs were determined using a series of linear mixed effects models fit to each of the 26,486 autosomal CpG loci from methylation data for previously obtained samples of sorted leukocytes6 (GEO: GSE39981), with a random intercept for beadchip to adjust for possible plate effect. Briefly, sorted non-pathological human peripheral blood leukocytes (n = 47) were previously obtained from different anonymous individuals through AllCells, LLC. Leukocytes were isolated by magnetic activated cell sorting (Miltenyi Biotec Inc.) and purity was confirmed by fluorescence activated cell sorting. The major cell types obtained included NK cells (n = 9), B cells (n = 5), CD4+ T cells (n = 8), CD8+ T cells (n = 2), “other” pan-T cells (n = 6), monocytes (n = 5), and granulocytes (n = 12). This yielded an F-statistic for differences across leukocyte types for each CpG locus.
Differential methylation
Differential methylation between cases and controls was examined among all autosomal loci (26 486 CpG sites) and for CpGs that did not significantly vary between leukocyte cell types (leukocyte-specific DMPs). To maximize the likelihood that all leukocyte-specific DMPs were excluded from the latter analysis, all CpG loci with an unadjusted P ≤ 0.05 were considered as putative DMPs (n = 9849), leaving 16 637 autosomal CpG loci that did not vary between leukocyte cell types (henceforth referred to as non-DMP CpGs) for the subsequent differential methylation analyses of the bladder cancer, HNSCC, and ovarian cancer case-control studies. Differentially methylated CpGs were identified by subtracting the median methylation β-value of controls from that of cases (Δβ) for each locus. To account for multiple testing, false discovery rate (FDR) estimation and Q-values were computed using the methods of Benjamini and Hochberg.42 CpGs were considered to be hypermethylated in cases if median β was greater in cases than controls (Δβ > 0) and hypomethylated if lower (Δβ < 0). Differential methylation was considered to be significant where Q ≤ 0.05.
Leukocyte-adjusted epigenome-wide association studies
The subject-specific leukocyte distribution (ωi, where i = 1,…,Np), a d0 × 1vector such that , was estimated using constrained projections based on peripheral blood methylation profiles, as previously described by Houseman et al.6 Here Np, p ∈ {1, 2, 3}, indicates the number of samples in the three cancer case-control data sets considered, and d0 indicates the number of unique cell-types (e.g., CD4+ T-lymphocytes, CD8+ T-lymphocytes, B cells, etc.). Several regression parameters were estimated: βj, representing the association of case-status and methylation β value at CpG j, unadjusted for ω; αj, representing the corresponding association adjusted for ω; and Г, the association of case-status and ω. Г, β and α were each adjusted for age, sex and smoking for bladder cancer and HNSCC, and age for ovarian cancer. Statistical inference was achieved by permutation, where null distributions were obtained by permuting case-status with respect to methylation values and other covariates, using an omnibus test adjusted for multiple-comparisons, constructed by comparing the observed average F-statistic across all CpGs to the corresponding quantity obtained from the permutation distribution. Volcano plots were generated for each adjusted and unadjusted EWAS by plotting the –log10 of the P value (y-axis) from the regression model against the case-coefficient from the respective case-coefficient. Volcano plots for the leukocyte-adjusted (represented by black diamonds) and -unadjusted (represented by blue open circles) models were overlaid for each respective case-control study to contrast the results.
To explore the biological relevance of our findings, we performed a gene set enrichment-based analysis to examine overrepresentation of any of 258 putative transcription factor binding site motifs (TFBS) located within 1kb of cancer-associated methylation of loci in blood CpG loci. TFBS data was obtained from the tfbsConsSites track of the UCSC Genomes Browser site (TFBS Z-score > 2). Overrepresented TFBS were determined using a Kolmogorov-Smirnov test, stratified by CpG island status of the target CpG. Stratification was implemented using a weighted chi-square statistic, with weights based on the number of CpGs within each of the two CpG island strata, individual stratum-specific chi-squares obtained by inverting the individual Kolomogorov-Smirnov P-value, and null distribution determined by simulation (107 samples). Significance was considered where the summary P ≤ 0.05.
All statistical analyses were conducted using the R statistical package (v. 2.11.1).
Supplementary Material
Disclosure of Potential Conflicts of Interest
K.T.K, E.A.H., C.J.M, and J.K.W. have applied for a patent covering DNA methylation arrays as biomarkers of immune cell distributions.
Acknowledgments
We thank Dr Shichun Zheng, George Hsuang, and Helen Hansen for technical assistance.
This work was supported by the National Cancer Institute (R01CA121147, R01CA100679, and R01CA078609 to K.T.K., R01CA057494 to M.R.K., R01CA082354 to H.H.N., R01 CA126831 J.K.W., and K22CA172358 to S.M.L.); and National Institute of Environmental Health Sciences (P42ES007373 to M.R.K. and T32ES07272 to S.M.L.).
References
- 1.Lander ES. Initial impact of the sequencing of the human genome. Nature. 2011;470:187–97. doi: 10.1038/nature09792. [DOI] [PubMed] [Google Scholar]
- 2.Stadler ZK, Vijai J, Thom P, Kirchhoff T, Hansen NA, Kauff ND, Robson M, Offit K. Genome-wide association studies of cancer predisposition. Hematol Oncol Clin North Am. 2010;24:973–96. doi: 10.1016/j.hoc.2010.06.009. [DOI] [PubMed] [Google Scholar]
- 3.Rakyan VK, Down TA, Balding DJ, Beck S. Epigenome-wide association studies for common human diseases. Nat Rev Genet. 2011;12:529–41. doi: 10.1038/nrg3000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Dawson MA, Kouzarides T. Cancer epigenetics: from mechanism to therapy. Cell. 2012;150:12–27. doi: 10.1016/j.cell.2012.06.013. [DOI] [PubMed] [Google Scholar]
- 5.Accomando WP, Wiencke JK, Houseman EA, Butler RA, Zheng S, Nelson HH, Kelsey KT. Decreased NK cells in patients with head and neck cancer determined in archival DNA. Clin Cancer Res. 2012;18:6147–54. doi: 10.1158/1078-0432.CCR-12-1008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Houseman EA, Accomando WP, Koestler DC, Christensen BC, Marsit CJ, Nelson HH, Wiencke JK, Kelsey KT. DNA methylation arrays as surrogate measures of cell mixture distribution. BMC Bioinformatics. 2012;13:86. doi: 10.1186/1471-2105-13-86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Ivascu C, Wasserkort R, Lesche R, Dong J, Stein H, Thiel A, Eckhardt F. DNA methylation profiling of transcription factor genes in normal lymphocyte development and lymphomas. Int J Biochem Cell Biol. 2007;39:1523–38. doi: 10.1016/j.biocel.2007.02.006. [DOI] [PubMed] [Google Scholar]
- 8.Koestler DC, Marsit CJ, Christensen BC, Accomando W, Langevin SM, Houseman EA, Nelson HH, Karagas MR, Wiencke JK, Kelsey KT. Peripheral blood immune cell methylation profiles are associated with nonhematopoietic cancers. Cancer Epidemiol Biomarkers Prev. 2012;21:1293–302. doi: 10.1158/1055-9965.EPI-12-0361. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Reinius LE, Acevedo N, Joerink M, Pershagen G, Dahlén SE, Greco D, Söderhäll C, Scheynius A, Kere J. Differential DNA methylation in purified human blood cells: implications for cell lineage and studies on disease susceptibility. PLoS One. 2012;7:e41361. doi: 10.1371/journal.pone.0041361. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Wieczorek G, Asemissen A, Model F, Turbachova I, Floess S, Liebenberg V, Baron U, Stauch D, Kotsch K, Pratschke J, et al. Quantitative DNA methylation analysis of FOXP3 as a new method for counting regulatory T cells in peripheral blood and solid tissue. Cancer Res. 2009;69:599–608. doi: 10.1158/0008-5472.CAN-08-2361. [DOI] [PubMed] [Google Scholar]
- 11.Wiencke JK, Accomando WP, Zheng S, Patoka J, Dou X, Phillips JJ, Hsuang G, Christensen BC, Houseman EA, Koestler DC, et al. Epigenetic biomarkers of T-cells in human glioma. Epigenetics. 2012;7:1391–402. doi: 10.4161/epi.22675. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Cedar H, Bergman Y. Epigenetics of haematopoietic cell development. Nat Rev Immunol. 2011;11:478–88. doi: 10.1038/nri2991. [DOI] [PubMed] [Google Scholar]
- 13.Lee ST, Xiao Y, Muench MO, Xiao J, Fomin ME, Wiencke JK, Zheng S, Dou X, de Smith A, Chokkalingam A, et al. A global DNA methylation and gene expression analysis of early human B-cell development reveals a demethylation signature and transcription factor network. Nucleic Acids Res. 2012;40:11339–51. doi: 10.1093/nar/gks957. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Christensen BC, Houseman EA, Marsit CJ, Zheng S, Wrensch MR, Wiemels JL, Nelson HH, Karagas MR, Padbury JF, Bueno R, et al. Aging and environmental exposures alter tissue-specific DNA methylation dependent upon CpG island context. PLoS Genet. 2009;5:e1000602. doi: 10.1371/journal.pgen.1000602. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Christensen BC, Marsit CJ. Epigenomics in environmental health. Front Genet. 2011;2:84. doi: 10.3389/fgene.2011.00084. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Feil R, Fraga MF. Epigenetics and the environment: emerging patterns and implications. Nat Rev Genet. 2011;13:97–109. doi: 10.1038/nrg3142. [DOI] [PubMed] [Google Scholar]
- 17.Cheng CK, Chan J, Cembrowski GS, van Assendelft OW. Complete blood count reference interval diagrams derived from NHANES III: stratification by age, sex, and race. Lab Hematol. 2004;10:42–53. doi: 10.1532/LH96.04010. [DOI] [PubMed] [Google Scholar]
- 18.Oertelt-Prigione S. The influence of sex and gender on the immune response. Autoimmun Rev. 2012;11:A479–85. doi: 10.1016/j.autrev.2011.11.022. [DOI] [PubMed] [Google Scholar]
- 19.Mehta H, Nazzal K, Sadikot RT. Cigarette smoking and innate immunity. Inflamm Res. 2008;57:497–503. doi: 10.1007/s00011-008-8078-6. [DOI] [PubMed] [Google Scholar]
- 20.Federico A, D’Aiuto E, Borriello F, Barra G, Gravina AG, Romano M, De Palma R. Fat: a matter of disturbance for the immune system. World J Gastroenterol. 2010;16:4762–72. doi: 10.3748/wjg.v16.i38.4762. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Kiecolt-Glaser JK, Gouin JP, Hantsoo L. Close relationships, inflammation, and health. Neurosci Biobehav Rev. 2010;35:33–8. doi: 10.1016/j.neubiorev.2009.09.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Szabo G, Mandrekar P. A recent perspective on alcohol, immunity, and host defense. Alcohol Clin Exp Res. 2009;33:220–32. doi: 10.1111/j.1530-0277.2008.00842.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Blum KS, Pabst R. Lymphocyte numbers and subsets in the human blood. Do they mirror the situation in all organs? Immunol Lett. 2007;108:45–51. doi: 10.1016/j.imlet.2006.10.009. [DOI] [PubMed] [Google Scholar]
- 24.Handin RI, Lux SE, Stossel TP, eds. Blood: Principles and Practice of Hematology. Philadelphia: Lippincott Williams & Wilkins, 2003. [Google Scholar]
- 25.Roussel M, Davis BH, Fest T, Wood BL, International Council for Standardization in Hematology (ICSH) Toward a reference method for leukocyte differential counts in blood: comparison of three flow cytometric candidate methods. Cytometry A. 2012;81:973–82. doi: 10.1002/cyto.a.22092. [DOI] [PubMed] [Google Scholar]
- 26.van de Geijn GJ, van Rees V, van Pul-Bom N, Birnie E, Janssen H, Pegels H, Beunis M, Njo T. Leukoflow: multiparameter extended white blood cell differentiation for routine analysis by flow cytometry. Cytometry A. 2011;79:694–706. doi: 10.1002/cyto.a.21105. [DOI] [PubMed] [Google Scholar]
- 27.Cherian S, Levin G, Lo WY, Mauck M, Kuhn D, Lee C, Wood BL. Evaluation of an 8-color flow cytometric reference method for white blood cell differential enumeration. Cytometry B Clin Cytom. 2010;78:319–28. doi: 10.1002/cyto.b.20529. [DOI] [PubMed] [Google Scholar]
- 28.Björnsson S, Wahlström S, Norström E, Bernevi I, O’Neill U, Johansson E, Runström H, Simonsson P. Total nucleated cell differential for blood and bone marrow using a single tube in a five-color flow cytometer. Cytometry B Clin Cytom. 2008;74:91–103. doi: 10.1002/cyto.b.20382. [DOI] [PubMed] [Google Scholar]
- 29.Koestler DC, Christensen B, Karagas MR, Marsit CJ, Langevin SM, Kelsey KT, Wiencke JK, Houseman EA. Blood-based profiles of DNA methylation predict the underlying distribution of cell types: a validation analysis. Epigenetics. 2013;8:816–26. doi: 10.4161/epi.25430. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Widschwendter M, Apostolidou S, Raum E, Rothenbacher D, Fiegl H, Menon U, Stegmaier C, Jacobs IJ, Brenner H. Epigenotyping in peripheral blood cell DNA and breast cancer risk: a proof of principle study. PLoS One. 2008;3:e2656. doi: 10.1371/journal.pone.0002656. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Liggett TE, Melnikov A, Yi Q, Replogle C, Hu W, Rotmensch J, Kamat A, Sood AK, Levenson V. Distinctive DNA methylation patterns of cell-free plasma DNA in women with malignant ovarian tumors. Gynecol Oncol. 2011;120:113–20. doi: 10.1016/j.ygyno.2010.09.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Teschendorff AE, Menon U, Gentry-Maharaj A, Ramus SJ, Gayther SA, Apostolidou S, Jones A, Lechner M, Beck S, Jacobs IJ, et al. An epigenetic signature in peripheral blood predicts active ovarian cancer. PLoS One. 2009;4:e8274. doi: 10.1371/journal.pone.0008274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Pedersen KS, Bamlet WR, Oberg AL, de Andrade M, Matsumoto ME, Tang H, Thibodeau SN, Petersen GM, Wang L. Leukocyte DNA methylation signature differentiates pancreatic cancer patients from healthy controls. PLoS One. 2011;6:e18223. doi: 10.1371/journal.pone.0018223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Marsit CJ, Koestler DC, Christensen BC, Karagas MR, Houseman EA, Kelsey KT. DNA methylation array analysis identifies profiles of blood-derived DNA methylation associated with bladder cancer. J Clin Oncol. 2011;29:1133–9. doi: 10.1200/JCO.2010.31.3577. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Langevin SM, Koestler DC, Christensen BC, Butler RA, Wiencke JK, Nelson HH, Houseman EA, Marsit CJ, Kelsey KT. Peripheral blood DNA methylation profiles are indicative of head and neck squamous cell carcinoma: an epigenome-wide association study. Epigenetics. 2012;7:291–9. doi: 10.4161/epi.7.3.19134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Cassinotti E, Melson J, Liggett T, Melnikov A, Yi Q, Replogle C, Mobarhan S, Boni L, Segato S, Levenson V. DNA methylation patterns in blood of patients with colorectal cancer and adenomatous colorectal polyps. Int J Cancer. 2012;131:1153–7. doi: 10.1002/ijc.26484. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Wang L, Aakre JA, Jiang R, Marks RS, Wu Y, Chen J, Thibodeau SN, Pankratz VS, Yang P. Methylation markers for small cell lung cancer in peripheral blood leukocyte DNA. J Thorac Oncol. 2010;5:778–85. doi: 10.1097/JTO.0b013e3181d6e0b3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Liu Y, Aryee MJ, Padyukov L, Fallin MD, Hesselberg E, Runarsson A, Reinius L, Acevedo N, Taub M, Ronninger M, et al. Epigenome-wide association data implicate DNA methylation as an intermediary of genetic risk in rheumatoid arthritis. Nat Biotechnol. 2013;31:142–7. doi: 10.1038/nbt.2487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Wallace K, Kelsey KT, Schned A, Morris JS, Andrew AS, Karagas MR. Selenium and risk of bladder cancer: a population-based case-control study. Cancer Prev Res (Phila) 2009;2:70–3. doi: 10.1158/1940-6207.CAPR-08-0046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Karagas MR, Tosteson TD, Blum J, Morris JS, Baron JA, Klaue B. Design of an epidemiologic study of drinking water arsenic exposure and skin and bladder cancer risk in a U.S. population. Environ Health Perspect. 1998;106(Suppl 4):1047–50. doi: 10.1289/ehp.98106s41047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Applebaum KM, McClean MD, Nelson HH, Marsit CJ, Christensen BC, Kelsey KT. Smoking modifies the relationship between XRCC1 haplotypes and HPV16-negative head and neck squamous cell carcinoma. Int J Cancer. 2009;124:2690–6. doi: 10.1002/ijc.24256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Benjamini Y, Hochberg Y. Controlling the False Discovery Rate - a Practical and Powerful Approach to Multiple Testing. J R Stat Soc, B. 1995;57:289–300. [Google Scholar]
- 43.Payer B, Saitou M, Barton SC, Thresher R, Dixon JP, Zahn D, Colledge WH, Carlton MB, Nakano T, Surani MA. Stella is a maternal effect gene required for normal early development in mice. Curr Biol. 2003;13:2110–7. doi: 10.1016/j.cub.2003.11.026. [DOI] [PubMed] [Google Scholar]
- 44.Elliott HR, Tillin T, McArdle WL, Ho K, Duggirala A, Frayling TM, Davey Smith G, Hughes AD, Chaturvedi N, Relton CL. Differences in smoking associated DNA methylation patterns in South Asians and Europeans. Clin Epigenetics. 2014;6:4. doi: 10.1186/1868-7083-6-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Xu Z, Bolick SC, DeRoo LA, Weinberg CR, Sandler DP, Taylor JA. Epigenome-wide association study of breast cancer using prospectively collected sister study samples. J Natl Cancer Inst. 2013;105:694–700. doi: 10.1093/jnci/djt045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Gallagher DJ, Milowsky MI, Ishill N, Trout A, Boyle MG, Riches J, Fleisher M, Bajorin DF. Detection of circulating tumor cells in patients with urothelial cancer. Ann Oncol. 2009;20:305–8. doi: 10.1093/annonc/mdn627. [DOI] [PubMed] [Google Scholar]
- 47.Ribal MJ, Mengual L, Marín M, Algaba F, Ars E, Fernández PL, Oliva R, Villavicencio H, Alcaraz A. Molecular staging of bladder cancer with RT-PCR assay for CK20 in peripheral blood, bone marrow and lymph nodes: comparison with standard histological staging. Anticancer Res. 2006;26(1A):411–9. [PubMed] [Google Scholar]
- 48.Karl A, Tritschler S, Hofmann S, Stief CG, Schindlbeck C. Perioperative search for circulating tumor cells in patients undergoing radical cystectomy for bladder cancer. Eur J Med Res. 2009;14:487–90. doi: 10.1186/2047-783X-14-11-487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Flaig TW, Wilson S, van Bokhoven A, Varella-Garcia M, Wolfe P, Maroni P, Genova EE, Morales D, Lucia MS. Detection of circulating tumor cells in metastatic and clinically localized urothelial carcinoma. Urology. 2011;78:863–7. doi: 10.1016/j.urology.2011.05.045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Okegawa T, Hayashi K, Hara H, Nutahara K, Higashihara E. Immunomagnetic quantification of circulating tumor cells in patients with urothelial cancer. Int J Urol. 2010;17:254–8. doi: 10.1111/j.1442-2042.2010.02454.x. [DOI] [PubMed] [Google Scholar]
- 51.Naoe M, Ogawa Y, Morita J, Omori K, Takeshita K, Shichijyo T, Okumura T, Igarashi A, Yanaihara A, Iwamoto S, et al. Detection of circulating urothelial cancer cells in the blood using the CellSearch System. Cancer. 2007;109:1439–45. doi: 10.1002/cncr.22543. [DOI] [PubMed] [Google Scholar]
- 52.Hristozova T, Konschak R, Stromberger C, Fusi A, Liu Z, Weichert W, Stenzinger A, Budach V, Keilholz U, Tinhofer I. The presence of circulating tumor cells (CTCs) correlates with lymph node metastasis in nonresectable squamous cell carcinoma of the head and neck region (SCCHN) Ann Oncol. 2011;22:1878–85. doi: 10.1093/annonc/mdr130. [DOI] [PubMed] [Google Scholar]
- 53.Pajonk F, Schlessmann S, Guttenberger R, Henke M. Epithelial cells in the peripheral blood of patients with cancer of the head and neck: incidence, detection and possible clinical significance. Radiother Oncol. 2001;59:213–7. doi: 10.1016/S0167-8140(00)00315-7. [DOI] [PubMed] [Google Scholar]
- 54.Nunes DN, Kowalski LP, Simpson AJ. Circulating tumor-derived DNA may permit the early diagnosis of head and neck squamous cell carcinomas. Int J Cancer. 2001;92:214–9. doi: 10.1002/1097-0215(200102)9999:9999<::AID-IJC1176>3.0.CO;2-C. [DOI] [PubMed] [Google Scholar]
- 55.Winter SC, Stephenson SA, Subramaniam SK, Paleri V, Ha K, Marnane C, Krishnan S, Rees G. Long term survival following the detection of circulating tumour cells in head and neck squamous cell carcinoma. BMC Cancer. 2009;9:424. doi: 10.1186/1471-2407-9-424. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.He W, Kularatne SA, Kalli KR, Prendergast FG, Amato RJ, Klee GG, Hartmann LC, Low PS. Quantitation of circulating tumor cells in blood samples from ovarian and prostate cancer patients using tumor-specific fluorescent ligands. Int J Cancer. 2008;123:1968–73. doi: 10.1002/ijc.23717. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Fan T, Zhao Q, Chen JJ, Chen WT, Pearl ML. Clinical significance of circulating tumor cells detected by an invasion assay in peripheral blood of patients with ovarian cancer. Gynecol Oncol. 2009;112:185–91. doi: 10.1016/j.ygyno.2008.09.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Aktas B, Kasimir-Bauer S, Heubner M, Kimmig R, Wimberger P. Molecular profiling and prognostic relevance of circulating tumor cells in the blood of ovarian cancer patients at primary diagnosis and after platinum-based chemotherapy. Int J Gynecol Cancer. 2011;21:822–30. doi: 10.1097/IGC.0b013e318216cb91. [DOI] [PubMed] [Google Scholar]
- 59.Obermayr E, Castillo-Tong DC, Pils D, Speiser P, Braicu I, Van Gorp T, et al. Molecular characterization of circulating tumor cells in patients with ovarian cancer improves their prognostic significance - A study of the OVCAD consortium. Gynecol Oncol. 2013;128:15, 21;. doi: 10.1016/j.ygyno.2012.09.021. [DOI] [PubMed] [Google Scholar]
- 60.Nezos A, Pissimisis N, Lembessis P, Sourla A, Dimopoulos P, Dimopoulos T, Tzelepis K, Koutsilieris M. Detection of circulating tumor cells in bladder cancer patients. Cancer Treat Rev. 2009;35:272–9. doi: 10.1016/j.ctrv.2008.11.003. [DOI] [PubMed] [Google Scholar]
- 61.Bouchon A, Hernández-Munain C, Cella M, Colonna M. A DAP12-mediated pathway regulates expression of CC chemokine receptor 7 and maturation of human dendritic cells. J Exp Med. 2001;194:1111–22. doi: 10.1084/jem.194.8.1111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Shiiba M, Nomura H, Shinozuka K, Saito K, Kouzu Y, Kasamatsu A, Sakamoto Y, Murano A, Ono K, Ogawara K, et al. Down-regulated expression of SERPIN genes located on chromosome 18q21 in oral squamous cell carcinomas. Oncol Rep. 2010;24:241–9. doi: 10.3892/or_00000852. [DOI] [PubMed] [Google Scholar]
- 63.Meschede W, Zumbach K, Braspenning J, Scheffner M, Benitez-Bribiesca L, Luande J, Gissmann L, Pawlita M. Antibodies against early proteins of human papillomaviruses as diagnostic markers for invasive cervical cancer. J Clin Microbiol. 1998;36:475–80. doi: 10.1128/jcm.36.2.475-480.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Sehr P, Müller M, Höpfl R, Widschwendter A, Pawlita M. HPV antibody detection by ELISA with capsid protein L1 fused to glutathione S-transferase. J Virol Methods. 2002;106:61–70. doi: 10.1016/S0166-0934(02)00134-9. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.


