Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Feb 1.
Published in final edited form as: Cancer Epidemiol Biomarkers Prev. 2012 Dec 14;22(2):189–200. doi: 10.1158/1055-9965.EPI-12-1263

Opportunities and Challenges for Selected Emerging Technologies in Cancer Epidemiology: Mitochondrial, Epigenomic, Metabolomic, and Telomerase Profiling

Mukesh Verma 1,*, Muin J Khoury 1, John PA Ioannidis 2
PMCID: PMC3565041  NIHMSID: NIHMS427307  PMID: 23242141

Abstract

Remarkable progress has been made in the last decade in new methods for biological measurements using sophisticated technologies that go beyond the established genome, proteome, and gene expression platforms. These methods and technologies create opportunities to enhance cancer epidemiologic studies. In this article, we describe several emerging technologies and evaluate their potential in epidemiologic studies. We review the background, assays, methods, and challenges, and offer examples of the use of mitochondrial DNA and copy number assessments, epigenomic profiling (including methylation, histone modification, microRNAs (miRNAs), and chromatin condensation), metabolite profiling (metabolomics), and telomere measurements. We map the volume of literature referring to each one of these measurement tools and the extent to which efforts have been made at knowledge integration (e.g. systematic reviews and meta-analyses). We also clarify strengths and weaknesses of the existing platforms and the range of type of samples that can be tested with each of them. These measurement tools can be used in identifying at-risk populations and providing novel markers of survival and treatment response. Rigorous analytical and validation standards, transparent availability of massive data, and integration in large-scale evidence are essential in fulfilling the potential of these technologies.

Keywords: Epigenetics, methylation, mitochondria, risk assessment, telomerase

Introduction

Tremendous progress has been made recently in the development and use of sophisticated technologies for enhancing biological measurements beyond the classic platforms of genomics, proteomics, and gene expression profiling. The advent of these tools offers unique opportunities and challenges for their use in human studies, and cancer epidemiology may benefit from incorporating such measurements. In this review, we assess the landscape of this emerging literature, and discuss several of these methods. We specifically address mitochondrial DNA and copy number assessments, epigenomic profiling (including assessments of methylation patterns, histone modification, microRNAs (miRNAs), and chromatin condensation), metabolite profiling (metabolomics), and telomere measurements. For each measurement platform we offer a background introduction, describe the main assays and methods, and list the main remaining challenges. Finally, we overview the use of these methods in the cancer epidemiology literature, the types of samples they can be used on, and their overall strengths and weaknesses.

Overview of the literature landscape

Table 1 shows the advent of these measurement platforms in the overall literature, and also focused on cancer, human studies, and specific types of designs. As shown, the volume of publications is still relatively limited compared with the massive literature on genomics/genetics and gene expression profiling, but many of these measurements already have as large literatures as proteomics with several tens of thousands of papers overall, and several thousands of papers focused on cancer in particular. Methylation and telomere-related papers have an especially strong cancer focus, with approximately 40% of the literature focusing on cancer (as compared with 13% of the overall PubMed). Moreover, 78–85% of the cancer literature on all these platforms is on humans. Their use in traditional epidemiologic studies is still relatively limited, accounting for a small fraction of this rapidly expanding literature, with only methylation-related epidemiologic studies exceeding 1,000. Many systematic reviews have also started being published, but meta-analyses remain uncommon, with only a few dozen being available. Most of these meta-analyses focus on single markers, and they almost ubiquitously depend on published summary data. This raises concerns about the breadth of coverage of the evidence and the reliability of inferences.

Table 1.

Landscape of the literature on emerging types of measurements

Measurements All Cancer Human Case control OR cohort SR MA
Emerging
Mitochondrial DNA 49990 5294 4211 134 8 0
Methylation 63132 20745 16604 1274 137 25
Histone modification 18002 5862 4643 74 27 0
miRNA 19886 7844 6089 405 77 19
Chromatin condensation 5649 1300 995 10 1 0
Metabolomics 7533 948 673 86 8 0
Telomerase 25074 9765 8332 326 43 12
Classic
Genome OR genomic*OR genetic 1529835 278971 224763 16488 2654 1080
Proteom* 54664 10108 8046 454 149 7
Gene expression 883585 201768 159270 5975 968 193

SR: systematic review; MA: meta-analysis.

Based on PubMed searches performed in November 2012; the last three columns are limited to studies that are already retrieved by Cancer and Human filters; the Human column is limited to studies that are already retrieved by the Cancer filter. Searches for the different measurements are: “mitochondrial DNA”, “methylation OR hypermethylation”, “histone AND (modification OR methylation OR acetylation)”, “miRNA OR microRNA”,”chromatin condensation”, “metabolomic* OR metabolome OR metabonomic*”, “telomere*”, “genome OR genomic* OR genetic”, “proteome*”, “gene expression”.

Mitochondrial DNA

Background

Mitochondria play an important role in cellular energy metabolism, free-radical generation, and apoptosis. During neoplastic transformation the mitochondrial genome may be damaged with accumulation of somatic mutations in the mitochondrial DNA (mtDNA). These mutations could represent a means for tracking tumor progression. Mitochondria contain their own genome (16.5 kb), along with transcription, translation, and protein assembly machinery and maintain genomic independence from the nucleus (1, 2). Both germline and somatic alterations in mtDNA have been observed in cancer and other diseases (36). For example, the polymorphism G10398A within the NADH dehydrogenase (ND3) subunit of Complex I has been probed for association with breast cancer, neurodegerative diseases, Alzheimer’s disease, Friedreich’s ataxia, longevity, and amyotropic lateral sclerosis (7). Somatic mitochondrial mutations have been detected in different tumor types, including in breast, colon, esophageal, endometrial, head and neck, liver, kidney, lung, oral, ovarian, prostate, and thyroid cancers, leukemia and melanoma (310). Most somatic mutations are homoplasmic in nature (i.e., all mitochondria carry the same mutations), with mutant mtDNA becoming dominant in tumor cells. Furthermore, the number of copies of mtDNA per cell can vary in normal and disease states (8). The mitochondrial genome lacks introns and is organized in 21 major haplogroups named after the letters of the alphabet (4, 912). Some haplogroups have been associated with specific cancers in specific populations (3, 4). Tools for characterizing and measuring mtDNA characteristics (including MitoChip) are available and are sufficiently high-throughput for assessing large numbers of epidemiological samples (13, 14). Numerous epidemiologic studies have been conducted using mitochondria information to examine cancer risk factors, natural history, screening markers, response to therapy and/or long term outcomes.

Assays and methods

Tissues, blood cells, exfoliated cells, and biofluids are a good source of mtDNA. To measure alterations in mtDNA (deletions, single nucleotide polymorphisms [SNPs], mutations, copy number), total DNA is usually isolated, followed by polymerase chain reaction (PCR) and nucleotide sequencing. The entire mitochondrial genome is amplified first in two long-range PCR reactions, followed by sequencing. Using MitoChip, mtDNA fragments are amplified and prepared for array hybridization according to the Affymetrix protocol for the GeneChip Customseq array (15). Investigators also have used restriction fragment length polymorphism (RFLP) analysis for mtDNA variations in tissue samples (16).

For haplogroup analysis, a hierarchical system combines multiplex PCR amplification, multiple single-base primer extensions, and capillary-based electrophoretic separation (17). The output of the GeneChip DNA analysis generates a report of the individual and total numbers of SNPs. Sequence variations are verified against reference mtDNA. Typically samples with call rates <95% are discarded. mtDNA molecules and the virtual number of mitochondria per cell are calculated with reference to a nuclear housekeeping gene (18). Laser capture microdissection can be used to separate different cell types, e.g. epithelial cells from stroma in for ovarian cancer (19, 20). A transparent thermoplastic film is attached to the tissue on the histopathology slide and cells are localized by microscopy. Different cell types are identified and targeted through the microscope with a 15–30 μm carbon dioxide laser beam pulse. The strong focal adhesion allows selective procurement of targeted cells suitable for mtDNA isolation and characterization.

Challenges

Determining an accurate mtDNA copy number is difficult, because in some situations mtDNA becomes integrated into the nuclear genome at nonspecific sites (8, 2123). Another challenge is the simultaneous characterization of nuclear and mtDNA in cases and controls. Although technically possible, such studies have not yet been conducted within large epidemiologic studies. Selection of sample source is another problem. When mutations in blood DNA were compared with mutations in breast cancer tissue from the same patient, the mutations did not match. This suggests that blood might not be the most appropriate biospecimen (24).

Epigenomics

Background

Epigenetics may affect gene expression without changing the nucleotide sequence. The four major components of epigenetic machinery include DNA methylation, histone modification, microRNA (miRNA) expression and processing, and chromatin condensation (25, 26). Methylation and histone markers have been used in studies trying to determine the etiology of breast, colon, esophageal, gastric, liver, lung, pancreas, ovary, prostate, renal, and other cancers (2531).

miRNA profiling has been used in cases and controls in some epidemiologic studies (e.g. disease survival in lung cancer and therapy outcome in bladder cancer) (3235). High-throughput miRNA quantification technologies such as the miRNA microarray (3641), bead-based flow cytometry (42), and real-time (RT)-PCR-based Taqman miRNA assay (43, 44) can be used for miRNA profiling.

Epigenetic biomarkers may offer advantages over other types of biomarkers because they reflect a person’s genetic background plus environmental exposures. Most epigenetic events occur early in cancer development and thus can be used for early detection. Epigenetic alterations also respond to environmental changes, and technologies are available to measure these changes (45, 46). Altered epigenomic profiling can be seen in response to toxins and environmental pollutants (4750). Different environmental exposures may affect different components of the epigenetic machinery. For example, exposure to metal carcinogens such as nickel, chromate, arsenite, and cadmium has increased recently because of occupational exposures, the massive growth of manufacturing activities, increased consumption of nonferrous materials, and disposal of waste products (51). These metals are potentially weak carcinogens: although they do not damage DNA directly (as does radiation), they may exert carcinogenic effects by epigenetic mechanisms, especially after chronic exposure (50).

Epigenetic alterations can be reversed by chemicals and can activate gene expression. Thus multiple potential uses have been proposed for epigenetic biomarkers in cancer intervention and treatment (25, 26, 29, 30, 42, 5264). Observational, experimental, and clinical studies in different diseases, especially cancer, have shown that nutrients may influence epigenetic regulation, e.g. folic acid can supply methyl groups (57, 59, 6568). Ingredients in some natural foods show properties similar to the inhibitors of histone acetylation.

Epidemiologic studies have been conducted in bladder (30), breast (69, 70), cervical (71), colon (72), gastric (26, 73, 74), head and neck (55, 75), liver (25, 52, 76), and renal (7779) cancers using methylation profiling and/or polymorphisms in genes involved in initiating or maintaining methylation (53, 54, 78, 80, 81). These studies have suggested associations between methylation markers and cancer development that need further validation. In most studies blood rather than tissue was used for analysis.

Assays and methods

Both tissues and biofluids have been used for epigenetic analysis. MethyLight technology, pyrosequencing, and chromatin immunoprecipitation-on-chip (ChIP-on-chip) can measure epigenetic alterations in cancer (82, 83). For methylation profiling, quantitative methylation-specific polymerase chain reaction (QMSP) assays are performed, followed by pyrosequencing (84). All assays use sodium bisulfite followed by alkali treatment (85). Bisulfite reacts with unmethylated cytosines and converts them to uracil. Methylated cytosines and other bases are not affected by bisulfite treatment. In the PCR reaction, all converted cytosines behave like uracils. MethyLight is the most common method used to determine the methylation profile in real-time (82, 8688). MethyLight is a high-throughput, quantitative methylation assay that utilizes fluorescence-based, real-time PCR technology and requires no manipulation after the PCR reaction. It can detect a methylation allele among 1,000 unmethylated alleles.

The most common method for miRNA profiling in cancer samples is the GeneChip microarray technology developed by Affymetrix. For histone profiling, monoclonal antibodies against specific histone modifications are used for chromatin immunoprecipitation (89, 90). Another popular epigenetics technique is the ChIP assay followed by next-generation sequencing (ChIP-seq) analysis, which can detect genome-wide histone modifications and methylation (91).

Challenges

Unlike the genome, which is the same for all types of cells, the epigenome is dynamic and changes with cell type and age. Therefore, the epigenome should be evaluated several times to follow cancer-associated alterations. The biggest challenge is the choice of sample (tissue vs. blood). Blood, which is collected in most epidemiologic studies, may not be an adequate sample, because epigenetic profiles and alterations of blood cells do not match those of tissue. Use of blood cells is also problematic because blood is a mixture of cells with different half-lives, ranging from 6 hours for neutrophils to months and years for macrophages and memory cells. Epigenetic changes are dynamic and continuously evolve during cancer development. Epigenetic changes are tissue-specific and cell type-specific. The research question itself determines the most appropriate tissue to be selected for epigenetic analyses.

Histone profiling uses ChIP assays that employ antibodies against post-translational modifications of histones (9294). Obtaining high-quality monoclonal antibodies for use against cancer-associated histone modifications is challenging, because monoclonal antibodies show batch effects (92). A central resource of large amounts of high-affinity, high-quality monoclonal antibodies is needed.

Proteins that bind to the methylated regions have been characterized, along with methylation patterns. These proteins are identified by methylated DNA immunoprecipitation (methyl DIP), which involves the hybridization of immunoprecipitated methylated DNA to microarrays or deep sequencing of the DNA in the immunoprecipitated DNA complex (95). Improvements are required, however, to adapt this process for large-scale use in addressing such problems as low resolution when using microarrays, difficulty in obtaining sufficient coverage when deep sequencing is used, and high false-discovery rates.

Taking precautions while collecting and storing samples for miRNA analysis can be challenging in epidemiologic studies. Ideally tissue samples are snap-frozen and stored at −70°C (96, 97). Fixed tissues can be problematic for miRNA analyses if proper protocols are not applied (98, 99). In miRNA analysis, different control RNAs are run simultaneously. During miRNA profiling, primers to the internal controls should be included to avoid false-positive results (100).

Metabolomics

Background

The metabolome measures directly the output of biological pathways and thus may be more representative of the functional state of cells than other “omics” measures. Metabolomics is the study of low molecular weight molecules or metabolites produced within cells and biological systems. Metabolomic profiling may help discover new disease-risk, screening, diagnostic, and prognostic biomarkers. This technology also provides novel insights into disease mechanisms (101103). The metabolome reflects cellular activity at the functional level and, hence, can be used to discern mechanistic information during normal and disease states (104107). In clinical samples (serum, urine), metabolites are more stable than proteins or RNA. The number of epidemiologic studies that use metabolomic profiling is still small compared with other technologies (Table 1), but applications are developing quickly (103, 104, 108) and validation studies are expected in the near future.

Assays and Methods

Metabolomic profiling is performed in blood or urine. Metabolomics involves two major technologies—mass spectrometry (MS) and nuclear magnetic resonance spectroscopy (NMR)—that can measure hundreds to thousands of unique chemical entities (101). The advantages of NMR include comprehensive generation of metabolite profiles by a single nondestructive method, full automation with high-throughput capacity, a well-established mathematical and statistical tool box, and very high analytical reproducibility (104). Disadvantages of NMR are its relative insensitivity in detecting metabolites with concentrations in the micromole range and above; and dependence on the quality of sample collection and handling, and on the available metadata. MS-based metabolomics typically consist of three basic components: (i) the “front end” fractionation of complex mixtures, (ii) mass spectral data acquisition, and (iii) metabolite identification and characterization by database searching. Advantages of MS include that the technique is highly sensitive and can detect metabolites with picomole concentrations, it requires small biospecimen volumes, separation by chromatography enables metabolites to be individually identified and quantified, and high-throughput automation is feasible (109, 110). Disadvantages of MS include expensive consumables, relatively lower analytical reproducibility, poor representation of highly polar metabolites when using standard chromatography protocols, and more complex software and algorithms required for routine data analysis (111, 112).

Challenges

Special attention must be paid to optimize protocols for maximizing the reproducibility, sensitivity, and quantitative reliability of metabolomics analysis. Furthermore, multivariate statistical modeling approaches are needed for better visualization and analysis of data. False-positive results can make interpretation difficult unless multiplicity is properly accounted for. Advancements in automatic sample preparation and handling, robotic sample delivery systems, automatic data processing, and multivariate statistical approaches can help streamline and standardize the process, but there are a number of different platforms (113120) and familiarity is required for their proper use.

Despite early promise, the full potential of metabolomics cannot be fully realized at the present time. Challenges include the limited availability of high-quality metabolite reference standards and of facilities that provide high-quality metabolomics services. To characterize unknown metabolites, standard, well-characterized metabolites are spiked with the clinical samples. The idea is to develop both isotopically labeled (i.e., 15N, 13C, or 2H) and unlabeled metabolite standards for use with MS and/or NMR, respectively. Compounds need to be synthesized in GLP laboratories with ISO 9000 certification and purified either by chromatographic methods or crystallization to > 95% purity. Classes of metabolites that require reference standards for metabolite identification include but are not limited to glycolytic and other energy intermediates, amino acid metabolism, lipids (phospholipids, glycerolipids, sphingolipids, glycolipids, oxylipins), acylcarnitines and acylglycines, secondary drug metabolites, secondary food metabolites, and fatty acids. The lack of widely-used robust automation tools and techniques in MS-based platforms remains a major limiting factor in high-throughput discovery and in transitioning such platforms to clinical chemistry laboratories (121)

Telomerase

Background

Telomeres, the ends of chromosomes, are specialized nucleoprotein structures that consist of guanine (G)-rich repetitive DNA sequences complexed with proteins (122124). Telomeres are required for maintenance, proper replication, and segregation of chromosomes. Without telomerase caps, human chromosomes undergo end-to-end fusion, forming dicentric and multicentric chromosomes that break during mitosis, leading to the activation of DNA damage checkpoints and initiation of the p53 pathway with growth arrest and cell death (125). Somatic cell telomeres shorten by 50–200 bp with each cell division, leading to replicative senescence and irreversible growth arrest. Telomere length is maintained by the protein telomerase, which adds TTAGGG repeats at the ends of chromosomes (126). Telomerase encompasses a catalytic subunit with telomerase reverse transcriptase (TERT) activity, a telomerase RNA component (TERC) that acts as a template for DNA synthesis, and the protein dyskerin (Dkc1), which binds and stabilizes TERC. Telomerase protects the chromosome ends from unscheduled DNA repair and degradation. Both the length of the telomere repeats and the integrity of telomere-binding proteins are important for telomere protection. Telomere shortening below a certain threshold length and/or alterations in the functionality of telomere-binding proteins can result in loss of telomere protection, leading eventually to apoptosis (127). Telomere dysfunction has been hypothesized to promote the acquisition of genetic lesions essential to cancer progression. Several epidemiologic studies have examined the average relative telomere length (RTL) as a potential biomarker for predisposition to bladder, colon, head and neck, lung, renal, and skin cancers (126, 128, 129). Biospecimen collection response rates are greater for buccal cells than for blood samples. PCR-based assays have been developed to measure telomerase activity in epidemiologic samples (130). In addition, the area around the TERT gene has been hypothesized to be a cancer polymorphism “hot spot” in different cancers (131134).

Assays and Methods

DNA from any type of cells is suitable for telomerase assays and can be isolated as described in reference (130). The PCR-based assay includes controls for inter-plate and intra-plate variability of threshold cycle values. RTL is calculated as the ratio of telomere repeat copy number to single-gene copy number in samples, compared with the reference DNA sample. Telomere length also can be determined by quantitative fluorescent in situ hybridization (TQ-FISH) (135, 136) where paraffin-embedded tissues are hybridized with fluorescence-tagged telomere probes.

Challenges

When studying the association between disease risk and telomere length, it is critical to determine the telomere length accurately. Discrepancies have been reported between telomere length-based studies and telomerase activity-based studies. In contrast to the belief that reduced telomere length reflects a risk of cancer, contradictory results were obtained by different investigators (134, 137139). Nonsignificant RTL shortening was observed in a breast cancer nested case-control study (130, 138). Study limitations that affect all epidemiologic observational studies, such as subject selection procedures, confounding, measurement errors, analysis, or selective reporting, might explain discrepancies.

Comments and Conclusions

Table 2 summarizes some strengths and weaknesses for each of the methods discussed above. Not all samples are suitable for these methods and technologies. A list of biospecimens and the appropriate technology for analyzing samples is provided in Table 3. Selected examples where technologies described in this article are applied for different epidemiologic studies are given in Table 4.

Table 2.

Comparison of selected emerging methods and technologies for use in cancer epidemiology

Method/Technology Strengths Weaknesses
mtDNA polymorphism Whole genome can be sequenced for a large number of samples because of the small size of the mt genome (16.5 kb) mtDNA sometimes can become integrated into the nuclear genome, and identifying the integrated mt genome in the nuclear genome is tedious
mtDNA copy number May provides additional information for identifying risk- and survival-associated biomarkers Experiments should be done very carefully because the number of copies varies during the disease development
Methylation profiling Provides a mechanism for studying gene activation/inactivation without a change in the genome Careful selection of the method is key to avoiding false- negative and false- positive results; tissue specificity can be a concern
miRNA profiling Requires a small amount of sample and provides additional information to understand epigenetically mediated gene regulation; information can be used in targeted intervention studies Tissue specificity can be a concern
Nuclear magnetic resonance (NMR) Quantitation of analytes is accurate with full automation and high-througput capacity and high reproducibility Identifying products can be challenging because of its insensitivity in detecting metabolites with concentrations in micromole range
Mass spectroscopy (MS) Extremely sensitive, can detect analytes at picomole range; requires small biospecimen volumes Requires expensive consumables; poor representation of highly polar metabolites
Metabolite profiling Can be done easily in patient biofluids Standards for all metabolites are not available
Telomerase activity and telomere size variation Suitable for paraffin-embedded tissue samples Since the length of telomere changes with age, subject selection is very critical

mt: mitochondrial

Table 3.

Sample types appropriate for selected methods and technologies that can be used in cancer risk assessment, detection, prognosis, and survival

Sample Method/Technology Typical Potential Applications in Epidemiologic Studies
Blood Methylation analysis (29, 75, 153155), miRNA analysis (156159), mtDNA analysis (160), metabolite profiling (161, 162), telomerase assay (134, 163, 164), Cancer detection, screening, survival, prognosis, risk assessment
Buccal cells Methylation analysis (165, 166), mtDNA analysis (167), mtDNA copy number analysis (8) Cancer detection
Circulating tumor cells Telomerase (168) Cancer detection and prognosis
Circulating plasma DNA Methylation analysis (31) Cancer prognosis
Exfoliated cells from cervix Methylation analysis (169, 170), mtDNA analysis (171) Cancer detection
Exfoliated cells from urine Methylation analysis (172, 173), mtDNA analysis (172, 173) Cancer detection
Nipple aspirate Methylation analysis (174, 175), mtDNA analysis (176178), metabolite profiling (179, 180) Cancer detection and prognosis
Serum miRNA profiling (32) Cancer prognosis
Tissue samples All methods [epigenomics (25, 26, 30, 33, 181184), telomerase (185), mtDNA (19, 20, 186)] Cancer detection and prognosis
Urine Metabolomic profiling (187, 188) Cancer detection, identification of lifestyle factors contributing to disease, factors contributing to cancer prognosis

Table 4.

Selected examples where mitochondrial, metabolomic, epigenomic, and telomerase profiling was used in screening, risk assessment, and prognosis (e.g. survival, disease aggression or recurrence)

Measurement Examples of Applications in epidemiology
Mitochondrial DNA alterations Germline mtDNA for screening populations for breast cancer (3, 5, 189, 190), breast and esophageal cancer (4), kidney cancer (191), pancreatic cancer (15); to evaluate long term survival in breast cancer (192), somatic mtDNA mutation to identify risk of lung cancer (193, 194)
Epigenetic profiling Methylation profiling to evaluate cancer survival in breast cancer (69), head and neck cancer (195), and esophageal squamous cell carcinoma (31); to evaluate disease aggressiveness in bladder cancer (30, 196); to detect disease and disease stratification in head and neck cancer (75) and kidney cancer (184); histone profiling and chromatin condensation in disease recurrence in lung cancer (183), colon cancer (25) and gastric cancer (26); miRNA profiling to evaluate survival in lung cancer (32, 33), miRNA polymorphism to evaluate therapy outcome in bladder and breast cancer (34)
Metabolomic profiling Metabolomic profiling in disease detection and stratification in ovarian cancer (102, 103); to evaluate nutritional factors and disease development (197, 198); life-style factors that contribute to lung cancer development (187); endogenous factors that contribute to endometrial cancer (199); and prostate cancer aggressiveness (200)
Telomerase profiling Association between TERT locus polymorphisms and predisposition to cancer (201, 202); TERT polymorphism in detecting risk of bladder cancer (203), ovarian cancer (134), and lung cancer (204, 205); tandem repeat minisatellite of telomerase as a risk factor for colorectal cancer (128); telomere length to assess breast cancer treatment outcome (206)

We have described the advent of several new biological measurement methods that may be of use in cancer epidemiology and beyond. We make some final comments here about the evolution of this evidence.

First, while we discussed each platform in isolation, it is possible that information obtained from multiple markers and multiple platforms may be most informative in some circumstances. Detecting multiple markers in cancer epidemiology has been suggested from time to time (140143). For example, El-Tayeh et al. (141) suggested evaluating alpha-fetoprotein (AFP), alpha-L-fucosidase (AFU), transforming growth factors alpha and beta (TGF-α and TGF-β), and interleukin-8 (IL-8) simultaneously to enhance the sensitivity and specificity of hepatocellular carcinoma. Large-scale assessment at multiple times of the genome, proteome, transcriptome, and metabolome has been recently described (144), and as platforms become less expensive, such combined assessments may become feasible in larger samples of patients. Selecting between complexity and parsimony remains a prominent challenge.

Second, for most of the platforms that we described, most of the ongoing research is discovery-oriented and replication efforts are still at their infancy. Not surprisingly, no meta-analysis to-date is available on any mtDNA topic and only few have been performed on epigenetic or telomere markers. This poses challenges in interpreting the reliability of the published results. Validation efforts should include not only cross-validation or bootstrapping on the same samples and datasets, but also external validation in independent diverse datasets, preferably also by different teams of investigators (145148). Reporting of these complex studies is also not standardized and would benefit from adoption of relevant reporting guidelines (149151).

Third, handling complex omics and related data collected in cancer epidemiology presents another challenge. The vast amount of data and biases that are introduced create a need for fast and effective computer analysis programs and for transparent large-scale data repositories. Most studies using the discussed platforms are done by single teams, but there is an increasing interest in larger coalitions of teams and consortia. Public availability of raw data, protocols, and analysis codes for these complex investigations could go a long way towards improving the transparency, reliability, and reproducibility of this research (145, 152).

In summary, progress continues to be made in emerging technologies in the cancer epigenetics and epidemiology fields, and some of the technologies are ready to be used in larger scale while others need improvements in analytical validity, high-throughput performance and sensitivity of detection. In the coming years, we expect that these emerging technologies may be used for different epidemiologic studies to contribute to a more comprehensive understanding of cancer risk factors, understand natural history and evaluate screening markers, and understand responses to therapy and/or evaluate longer term outcomes. Epidemiologic studies may also inform future randomized controlled trials to explore clinical utility for different applications in practice.

Acknowledgments

We are thankful to Andrew Freedman, Elizabeth Gillanders, Somdat Mahabir, Britt Reid, Sheri Schully, and Daniela Seminara for reading the manuscript and providing useful comments.

Abbreviations

miRNA

microRNA

mtDNA

mitochondrial DNA

RTL

Relative Telomerase Length

TERT

Telomerase reverse transcriptase

References

  • 1.Czarnecka AM, Golik P, Bartnik E. Mitochondrial DNA mutations in human neoplasia. J Appl Genet. 2006;47(1):67–78. doi: 10.1007/BF03194602. [DOI] [PubMed] [Google Scholar]
  • 2.Czarnecka AM, Czarnecki JS, Kukwa W, Cappello F, Scinska A, Kukwa A. Molecular oncology focus - is carcinogenesis a ‘mitochondriopathy’? J Biomed Sci. 2010;17:31. doi: 10.1186/1423-0127-17-31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Canter JA, Kallianpur AR, Parl FF, Millikan RC. Mitochondrial DNA G10398A polymorphism and invasive breast cancer in African-American women. Cancer Res. 2005;65(17):8028–33. doi: 10.1158/0008-5472.CAN-05-1428. [DOI] [PubMed] [Google Scholar]
  • 4.Darvishi K, Sharma S, Bhat AK, Rai E, Bamezai RN. Mitochondrial DNA G10398A polymorphism imparts maternal Haplogroup N a risk for breast and esophageal cancer. Cancer Lett. 2007;249(2):249–55. doi: 10.1016/j.canlet.2006.09.005. [DOI] [PubMed] [Google Scholar]
  • 5.Mims MP, Hayes TG, Zheng S, Leal SM, Frolov A, Ittmann MM, et al. Mitochondrial DNA G10398A polymorphism and invasive breast cancer in African-American women. Cancer Res. 2006;66(3):1880. doi: 10.1158/0008-5472.CAN-05-3774. author reply 1880–1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Yu M. Somatic mitochondrial DNA mutations in human cancers. Adv Clin Chem. 2012;57:99–138. doi: 10.1016/b978-0-12-394384-2.00004-8. [DOI] [PubMed] [Google Scholar]
  • 7.Ross OA, McCormack R, Curran MD, Duguid RA, Barnett YA, Rea IM, et al. Mitochondrial DNA polymorphism: its role in longevity of the Irish population. Exp Gerontol. 2001;36(7):1161–78. doi: 10.1016/s0531-5565(01)00094-8. [DOI] [PubMed] [Google Scholar]
  • 8.Yu M. Generation, function and diagnostic value of mitochondrial DNA copy number alterations in human cancers. Life Sci. 2011;89(3–4):65–71. doi: 10.1016/j.lfs.2011.05.010. [DOI] [PubMed] [Google Scholar]
  • 9.Fang H, Shen L, Chen T, He J, Ding Z, Wei J, et al. Cancer type-specific modulation of mitochondrial haplogroups in breast, colorectal and thyroid cancer. BMC Cancer. 2010;10:421. doi: 10.1186/1471-2407-10-421. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Ebner S, Lang R, Mueller EE, Eder W, Oeller M, Moser A, et al. Mitochondrial haplogroups, control region polymorphisms and malignant melanoma: a study in middle European Caucasians. PLoS One. 2011;6(12):e27192. doi: 10.1371/journal.pone.0027192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Verma M, Kagan J, Sidransky D, Srivastava S. Proteomic analysis of cancer-cell mitochondria. Nat Rev Cancer. 2003;3(10):789–95. doi: 10.1038/nrc1192. [DOI] [PubMed] [Google Scholar]
  • 12.Verma M, Naviaux RK, Tanaka M, Kumar D, Franceschi C, Singh KK. Meeting report: mitochondrial DNA and cancer epidemiology. Cancer Res. 2007;67(2):437–9. doi: 10.1158/0008-5472.CAN-06-4119. [DOI] [PubMed] [Google Scholar]
  • 13.Maitra A, Cohen Y, Gillespie SE, Mambo E, Fukushima N, Hoque MO, et al. The Human MitoChip: a high-throughput sequencing microarray for mitochondrial mutation detection. Genome Res. 2004;14(5):812–9. doi: 10.1101/gr.2228504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Kassauei K, Habbe N, Mullendore ME, Karikari CA, Maitra A, Feldmann G. Mitochondrial DNA mutations in pancreatic cancer. Int J Gastrointest Cancer. 2006;37(2–3):57–64. doi: 10.1007/s12029-007-0008-2. [DOI] [PubMed] [Google Scholar]
  • 15.Lam ET, Bracci PM, Holly EA, Chu C, Poon A, Wan E, et al. Mitochondrial DNA sequence variation and risk of pancreatic cancer. Cancer Res. 2012;72(3):686–95. doi: 10.1158/0008-5472.CAN-11-1682. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Ye C, Shu XO, Pierce L, Wen W, Courtney R, Gao YT, et al. Mutations in the mitochondrial DNA D-loop region and breast cancer risk. Breast Cancer Res Treat. 2010;119(2):431–6. doi: 10.1007/s10549-009-0397-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Wiesbauer M, Meierhofer D, Mayr JA, Sperl W, Paulweber B, Kofler B. Multiplex primer extension analysis for rapid detection of major European mitochondrial haplogroups. Electrophoresis. 2006;27(19):3864–8. doi: 10.1002/elps.200600086. [DOI] [PubMed] [Google Scholar]
  • 18.Robin ED, Wong R. Mitochondrial DNA molecules and virtual number of mitochondria per cell in mammalian cells. J Cell Physiol. 1988;136(3):507–13. doi: 10.1002/jcp.1041360316. [DOI] [PubMed] [Google Scholar]
  • 19.Fukino K, Shen L, Matsumoto S, Morrison CD, Mutter GL, Eng C. Combined total genome loss of heterozygosity scan of breast cancer stroma and epithelium reveals multiplicity of stromal targets. Cancer Res. 2004;64(20):7231–6. doi: 10.1158/0008-5472.CAN-04-2866. [DOI] [PubMed] [Google Scholar]
  • 20.Kurose K, Hoshaw-Woodard S, Adeyinka A, Lemeshow S, Watson PH, Eng C. Genetic model of multi-step breast carcinogenesis involving the epithelium and stroma: clues to tumour-microenvironment interactions. Hum Mol Genet. 2001;10(18):1907–13. doi: 10.1093/hmg/10.18.1907. [DOI] [PubMed] [Google Scholar]
  • 21.Lee HC, Li SH, Lin JC, Wu CC, Yeh DC, Wei YH. Somatic mutations in the D-loop and decrease in the copy number of mitochondrial DNA in human hepatocellular carcinoma. Mutat Res. 2004;547(1–2):71–8. doi: 10.1016/j.mrfmmm.2003.12.011. [DOI] [PubMed] [Google Scholar]
  • 22.Yin PH, Lee HC, Chau GY, Wu YT, Li SH, Lui WY, et al. Alteration of the copy number and deletion of mitochondrial DNA in human hepatocellular carcinoma. Br J Cancer. 2004;90(12):2390–6. doi: 10.1038/sj.bjc.6601838. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Yamada S, Nomoto S, Fujii T, Kaneko T, Takeda S, Inoue S, et al. Correlation between copy number of mitochondrial DNA and clinico-pathologic parameters of hepatocellular carcinoma. Eur J Surg Oncol. 2006;32(3):303–7. doi: 10.1016/j.ejso.2006.01.002. [DOI] [PubMed] [Google Scholar]
  • 24.Cai FF, Kohler C, Zhang B, Chen WJ, Barekati Z, Garritsen HS, et al. Mutations of mitochondrial DNA as potential biomarkers in breast cancer. Anticancer Res. 2011;31(12):4267–71. [PubMed] [Google Scholar]
  • 25.Tamagawa H, Oshima T, Shiozawa M, Morinaga S, Nakamura Y, Yoshihara M, et al. The global histone modification pattern correlates with overall survival in metachronous liver metastasis of colorectal cancer. Oncol Rep. 2012;27(3):637–42. doi: 10.3892/or.2011.1547. [DOI] [PubMed] [Google Scholar]
  • 26.Park YS, Jin MY, Kim YJ, Yook JH, Kim BS, Jang SJ. The global histone modification pattern correlates with cancer recurrence and overall survival in gastric adenocarcinoma. Ann Surg Oncol. 2008;15(7):1968–76. doi: 10.1245/s10434-008-9927-9. [DOI] [PubMed] [Google Scholar]
  • 27.Vineis P, Chuang SC, Vaissiere T, Cuenin C, Ricceri F, Johansson M, et al. DNA methylation changes associated with cancer risk factors and blood levels of vitamin metabolites in a prospective study. Epigenetics. 2011;6(2):195–201. doi: 10.4161/epi.6.2.13573. [DOI] [PubMed] [Google Scholar]
  • 28.Brennan K, Flanagan JM. Epigenetic epidemiology for cancer risk: harnessing germline epigenetic variation. Methods Mol Biol. 2012;863:439–65. doi: 10.1007/978-1-61779-612-8_27. [DOI] [PubMed] [Google Scholar]
  • 29.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(1):108–14. doi: 10.1158/1055-9965.EPI-06-0636. [DOI] [PubMed] [Google Scholar]
  • 30.Yan C, Kim YW, Ha YS, Kim IY, Kim YJ, Yun SJ, et al. RUNX3 methylation as a predictor for disease progression in patients with non-muscle-invasive bladder cancer. J Surg Oncol. 2012;105(4):425–30. doi: 10.1002/jso.22087. [DOI] [PubMed] [Google Scholar]
  • 31.Ling ZQ, Zhao Q, Zhou SL, Mao WM. MSH2 promoter hypermethylation in circulating tumor DNA is a valuable predictor of disease-free survival for patients with esophageal squamous cell carcinoma. Eur J Surg Oncol. 2012;38(4):326–32. doi: 10.1016/j.ejso.2012.01.008. [DOI] [PubMed] [Google Scholar]
  • 32.Hu Z, Chen X, Zhao Y, Tian T, Jin G, Shu Y, et al. Serum microRNA signatures identified in a genome-wide serum microRNA expression profiling predict survival of non-small-cell lung cancer. J Clin Oncol. 2010;28(10):1721–6. doi: 10.1200/JCO.2009.24.9342. [DOI] [PubMed] [Google Scholar]
  • 33.Landi MT, Zhao Y, Rotunno M, Koshiol J, Liu H, Bergen AW, et al. MicroRNA expression differentiates histology and predicts survival of lung cancer. Clin Cancer Res. 2010;16(2):430–41. doi: 10.1158/1078-0432.CCR-09-1736. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Teo MT, Landi D, Taylor CF, Elliott F, Vaslin L, Cox DG, et al. The role of microRNA-binding site polymorphisms in DNA repair genes as risk factors for bladder cancer and breast cancer and their impact on radiotherapy outcomes. Carcinogenesis. 2012;33(3):581–6. doi: 10.1093/carcin/bgr300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Nair VS, Maeda LS, Ioannidis JP. Clinical outcome prediction by microRNAs in human cancer: a systematic review. J Natl Cancer Inst. 2012;104(7):528–40. doi: 10.1093/jnci/djs027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Castoldi M, Schmidt S, Benes V, Hentze MW, Muckenthaler MU. miChip: an array-based method for microRNA expression profiling using locked nucleic acid capture probes. Nat Protoc. 2008;3(2):321–9. doi: 10.1038/nprot.2008.4. [DOI] [PubMed] [Google Scholar]
  • 37.Castoldi M, Benes V, Hentze MW, Muckenthaler MU. miChip: a microarray platform for expression profiling of microRNAs based on locked nucleic acid (LNA) oligonucleotide capture probes. Methods. 2007;43(2):146–52. doi: 10.1016/j.ymeth.2007.04.009. [DOI] [PubMed] [Google Scholar]
  • 38.Castoldi M, Schmidt S, Benes V, Noerholm M, Kulozik AE, Hentze MW, et al. A sensitive array for microRNA expression profiling (miChip) based on locked nucleic acids (LNA) RNA. 2006;12(5):913–20. doi: 10.1261/rna.2332406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Goff LA, Yang M, Bowers J, Getts RC, Padgett RW, Hart RP. Rational probe optimization and enhanced detection strategy for microRNAs using microarrays. RNA Biol. 2005;2(3):93–100. doi: 10.4161/rna.2.3.2059. [DOI] [PubMed] [Google Scholar]
  • 40.Liu CG, Calin GA, Meloon B, Gamliel N, Sevignani C, Ferracin M, et al. An oligonucleotide microchip for genome-wide microRNA profiling in human and mouse tissues. Proc Natl Acad Sci U S A. 2004;101(26):9740–4. doi: 10.1073/pnas.0403293101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Thomson JM, Parker J, Perou CM, Hammond SM. A custom microarray platform for analysis of microRNA gene expression. Nat Methods. 2004;1(1):47–53. doi: 10.1038/nmeth704. [DOI] [PubMed] [Google Scholar]
  • 42.Lu J, Getz G, Miska EA, Alvarez-Saavedra E, Lamb J, Peck D, et al. MicroRNA expression profiles classify human cancers. Nature. 2005;435(7043):834–8. doi: 10.1038/nature03702. [DOI] [PubMed] [Google Scholar]
  • 43.Chen C, Ridzon DA, Broomer AJ, Zhou Z, Lee DH, Nguyen JT, et al. Real-time quantification of microRNAs by stem-loop RT-PCR. Nucleic Acids Res. 2005;33(20):e179. doi: 10.1093/nar/gni178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Jiang J, Lee EJ, Gusev Y, Schmittgen TD. Real-time expression profiling of microRNA precursors in human cancer cell lines. Nucleic Acids Res. 2005;33(17):5394–403. doi: 10.1093/nar/gki863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Arita A, Costa M. Epigenetics in metal carcinogenesis: nickel, arsenic, chromium and cadmium. Metallomics. 2009;1(3):222–8. doi: 10.1039/b903049b. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Sutherland JE, Costa M. Epigenetics and the environment. Ann N Y Acad Sci. 2003;983:151–60. doi: 10.1111/j.1749-6632.2003.tb05970.x. [DOI] [PubMed] [Google Scholar]
  • 47.Bollati V, Baccarelli A, Sartori S, Tarantini L, Motta V, Rota F, et al. Epigenetic effects of shiftwork on blood DNA methylation. Chronobiol Int. 2010;27(5):1093–104. doi: 10.3109/07420528.2010.490065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Zhu ZZ, Hou L, Bollati V, Tarantini L, Marinelli B, Cantone L, et al. Predictors of global methylation levels in blood DNA of healthy subjects: a combined analysis. Int J Epidemiol. 2012;41(1):126–39. doi: 10.1093/ije/dyq154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Costa BM, Caeiro C, Guimaraes I, Martinho O, Jaraquemada T, Augusto I, et al. Prognostic value of MGMT promoter methylation in glioblastoma patients treated with temozolomide-based chemoradiation: a Portuguese multicentre study. Oncol Rep. 2010;23(6):1655–62. doi: 10.3892/or_00000808. [DOI] [PubMed] [Google Scholar]
  • 50.Arita A, Niu J, Qu Q, Zhao N, Ruan Y, Nadas A, et al. Global levels of histone modifications in peripheral blood mononuclear cells of subjects with exposure to nickel. Environ Health Perspect. 2012;120(2):198–203. doi: 10.1289/ehp.1104140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Arita A, Shamy MY, Chervona Y, Clancy HA, Sun H, Hall MN, et al. The effect of exposure to carcinogenic metals on histone tail modifications and gene expression in human subjects. J Trace Elem Med Biol. 2012;26(2–3):174–8. doi: 10.1016/j.jtemb.2012.03.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Rivenbark AG, Coleman WB. The use of epigenetic biomarkers for preclinical detection of hepatocellular carcinoma: potential for noninvasive screening of high-risk populations. Clin Cancer Res. 2007;13(8):2309–12. doi: 10.1158/1078-0432.CCR-07-0086. [DOI] [PubMed] [Google Scholar]
  • 53.Kiyohara C, Horiuchi T, Takayama K, Nakanishi Y. Methylenetetrahydrofolate reductase polymorphisms and interaction with smoking and alcohol consumption in lung cancer risk: a case-control study in a Japanese population. BMC Cancer. 2011;11:459. doi: 10.1186/1471-2407-11-459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Kawakita D, Matsuo K, Sato F, Oze I, Hosono S, Ito H, et al. Association between dietary folate intake and clinical outcome in head and neck squamous cell carcinoma. Ann Oncol. 2012;23(1):186–92. doi: 10.1093/annonc/mdr057. [DOI] [PubMed] [Google Scholar]
  • 55.Poage GM, Butler RA, Houseman EA, McClean MD, Nelson HH, Christensen BC, et al. Identification of an Epigenetic Profile Classifier That Is Associated with Survival in Head and Neck Cancer. Cancer Res. 2012;72(11):2728–2737. doi: 10.1158/0008-5472.CAN-11-4121-T. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Verma M. Epigenetic biomarkers in cancer epidemiology. Methods Mol Biol. 2012;863:467–80. doi: 10.1007/978-1-61779-612-8_28. [DOI] [PubMed] [Google Scholar]
  • 57.Khare S, Verma M. Epigenetics of colon cancer. Methods Mol Biol. 2012;863:177–85. doi: 10.1007/978-1-61779-612-8_10. [DOI] [PubMed] [Google Scholar]
  • 58.Mishra A, Verma M. Epigenetics of solid cancer stem cells. Methods Mol Biol. 2012;863:15–31. doi: 10.1007/978-1-61779-612-8_2. [DOI] [PubMed] [Google Scholar]
  • 59.Verma M. Cancer control and prevention by nutrition and epigenetic approaches. Antioxid Redox Signal. 2012;17(2):355–64. doi: 10.1089/ars.2011.4388. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Kumar D, Verma M. Methods in cancer epigenetics and epidemiology. Methods Mol Biol. 2009;471:273–88. doi: 10.1007/978-1-59745-416-2_14. [DOI] [PubMed] [Google Scholar]
  • 61.Verma M, Maruvada P, Srivastava S. Epigenetics and cancer. Crit Rev Clin Lab Sci. 2004;41(5–6):585–607. doi: 10.1080/10408360490516922. [DOI] [PubMed] [Google Scholar]
  • 62.Verma M, Dunn BK, Ross S, Jain P, Wang W, Hayes R, et al. Early detection and risk assessment: proceedings and recommendations from the Workshop on Epigenetics in Cancer Prevention. Ann N Y Acad Sci. 2003;983:298–319. doi: 10.1111/j.1749-6632.2003.tb05984.x. [DOI] [PubMed] [Google Scholar]
  • 63.Verma M. Viral genes and methylation. Ann N Y Acad Sci. 2003;983:170–80. doi: 10.1111/j.1749-6632.2003.tb05972.x. [DOI] [PubMed] [Google Scholar]
  • 64.Verma M, Srivastava S. Epigenetics in cancer: implications for early detection and prevention. Lancet Oncol. 2002;3(12):755–63. doi: 10.1016/s1470-2045(02)00932-4. [DOI] [PubMed] [Google Scholar]
  • 65.Holmes RS, Zheng Y, Baron JA, Li L, McKeown-Eyssen G, Newcomb PA, et al. Use of folic acid-containing supplements after a diagnosis of colorectal cancer in the Colon Cancer Family Registry. Cancer Epidemiol Biomarkers Prev. 2010;19(8):2023–34. doi: 10.1158/1055-9965.EPI-09-1097. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Duthie SJ. Epigenetic modifications and human pathologies: cancer and CVD. Proc Nutr Soc. 2011;70(1):47–56. doi: 10.1017/S0029665110003952. [DOI] [PubMed] [Google Scholar]
  • 67.Park Y, Spiegelman D, Hunter DJ, Albanes D, Bergkvist L, Buring JE, et al. Intakes of vitamins A, C, and E and use of multiple vitamin supplements and risk of colon cancer: a pooled analysis of prospective cohort studies. Cancer Causes Control. 2010;21(11):1745–57. doi: 10.1007/s10552-010-9549-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Nystrom M, Mutanen M. Diet and epigenetics in colon cancer. World J Gastroenterol. 2009;15(3):257–63. doi: 10.3748/wjg.15.257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Ben Gacem R, Hachana M, Ziadi S, Amara K, Ksia F, Mokni M, et al. Contribution of epigenetic alteration of BRCA1 and BRCA2 genes in breast carcinomas in Tunisian patients. Cancer Epidemiol. 2012;36(2):190–7. doi: 10.1016/j.canep.2011.09.001. [DOI] [PubMed] [Google Scholar]
  • 70.Fuhrman BJ, Schairer C, Gail MH, Boyd-Morin J, Xu X, Sue LY, et al. Estrogen metabolism and risk of breast cancer in postmenopausal women. J Natl Cancer Inst. 2012;104(4):326–39. doi: 10.1093/jnci/djr531. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Mirabello L, Sun C, Ghosh A, Rodriguez AC, Schiffman M, Wentzensen N, et al. Methylation of human papillomavirus type 16 genome and risk of cervical precancer in a Costa Rican population. J Natl Cancer Inst. 2012;104(7):556–65. doi: 10.1093/jnci/djs135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Han SS, Sue LY, Berndt SI, Selhub J, Burdette LA, Rosenberg PS, et al. Associations between genes in the one-carbon metabolism pathway and advanced colorectal adenoma risk in individuals with low folate intake. Cancer Epidemiol Biomarkers Prev. 2012;21(3):417–27. doi: 10.1158/1055-9965.EPI-11-0782. [DOI] [PubMed] [Google Scholar]
  • 73.Balassiano K, Lima S, Jenab M, Overvad K, Tjonneland A, Boutron-Ruault MC, et al. Aberrant DNA methylation of cancer-associated genes in gastric cancer in the European Prospective Investigation into Cancer and Nutrition (EPIC-EURGAST) Cancer Lett. 2011;311(1):85–95. doi: 10.1016/j.canlet.2011.06.038. [DOI] [PubMed] [Google Scholar]
  • 74.Gao Y, Baccarelli A, Shu XO, Ji BT, Yu K, Tarantini L, et al. Blood leukocyte Alu and LINE-1 methylation and gastric cancer risk in the Shanghai Women’s Health Study. Br J Cancer. 2012;106(3):585–91. doi: 10.1038/bjc.2011.562. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Langevin SM, Koestler DC, Christensen BC, Butler RA, Wiencke JK, Nelson HH, et al. Peripheral blood DNA methylation profiles are indicative of head and neck squamous cell carcinoma: an epigenome-wide association study. Epigenetics. 2012;7(3):291–9. doi: 10.4161/epi.7.3.19134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Cheng Y, Zhang C, Zhao J, Wang C, Xu Y, Han Z, et al. Correlation of CpG island methylator phenotype with poor prognosis in hepatocellular carcinoma. Exp Mol Pathol. 2010;88(1):112–7. doi: 10.1016/j.yexmp.2009.10.008. [DOI] [PubMed] [Google Scholar]
  • 77.Liao LM, Brennan P, van Bemmel DM, Zaridze D, Matveev V, Janout V, et al. LINE-1 methylation levels in leukocyte DNA and risk of renal cell cancer. PLoS One. 2011;6(11):e27361. doi: 10.1371/journal.pone.0027361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Gibson TM, Brennan P, Han S, Karami S, Zaridze D, Janout V, et al. Comprehensive evaluation of one-carbon metabolism pathway gene variants and renal cell cancer risk. PLoS One. 2011;6(10):e26165. doi: 10.1371/journal.pone.0026165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Ahmad ST, Arjumand W, Seth A, Saini AK, Sultana S. Methylation of the APAF-1 and DAPK-1 promoter region correlates with progression of renal cell carcinoma in North Indian population. Tumour Biol. 2012;33(2):395–402. doi: 10.1007/s13277-011-0235-9. [DOI] [PubMed] [Google Scholar]
  • 80.Ho PA, Kutny MA, Alonzo TA, Gerbing RB, Joaquin J, Raimondi SC, et al. Leukemic mutations in the methylation-associated genes DNMT3A and IDH2 are rare events in pediatric AML: a report from the Children’s Oncology Group. Pediatr Blood Cancer. 2011;57(2):204–9. doi: 10.1002/pbc.23179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Songserm N, Promthet S, Sithithaworn P, Pientong C, Ekalaksananan T, Chopjitt P, et al. Risk factors for cholangiocarcinoma in high-risk area of Thailand: role of lifestyle, diet and methylenetetrahydrofolate reductase polymorphisms. Cancer Epidemiol. 2012;36(2):e89–94. doi: 10.1016/j.canep.2011.11.007. [DOI] [PubMed] [Google Scholar]
  • 82.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(8):E32. doi: 10.1093/nar/28.8.e32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Fazzari MJ, Greally JM. Introduction to epigenomics and epigenome-wide analysis. Methods Mol Biol. 2010;620:243–65. doi: 10.1007/978-1-60761-580-4_7. [DOI] [PubMed] [Google Scholar]
  • 84.Weidlich S, Walsh K, Crowther D, Burczynski ME, Feuerstein G, Carey FA, et al. Pyrosequencing-based methods reveal marked inter-individual differences in oncogene mutation burden in human colorectal tumours. Br J Cancer. 2011;105(2):246–54. doi: 10.1038/bjc.2011.197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Tetzner R, Dietrich D, Distler J. Control of carry-over contamination for PCR-based DNA methylation quantification using bisulfite treated DNA. Nucleic Acids Res. 2007;35(1):e4. doi: 10.1093/nar/gkl955. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Eads CA, Lord RV, Wickramasinghe K, Long TI, Kurumboor SK, Bernstein L, et al. Epigenetic patterns in the progression of esophageal adenocarcinoma. Cancer Res. 2001;61(8):3410–8. [PubMed] [Google Scholar]
  • 87.Eads CA, Lord RV, Kurumboor SK, Wickramasinghe K, Skinner ML, Long TI, et al. Fields of aberrant CpG island hypermethylation in Barrett’s esophagus and associated adenocarcinoma. Cancer Res. 2000;60(18):5021–6. [PubMed] [Google Scholar]
  • 88.Zhou J, Cao J, Lu Z, Liu H, Deng D. A 115-bp MethyLight assay for detection of p16 (CDKN2A) methylation as a diagnostic biomarker in human tissues. BMC Med Genet. 2011;12:67. doi: 10.1186/1471-2350-12-67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Cantone L, Nordio F, Hou L, Apostoli P, Bonzini M, Tarantini L, et al. Inhalable metal-rich air particles and histone H3K4 dimethylation and H3K9 acetylation in a cross-sectional study of steel workers. Environ Health Perspect. 2011;119(7):964–9. doi: 10.1289/ehp.1002955. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Enroth S, Rada-Iglesisas A, Andersson R, Wallerman O, Wanders A, Pahlman L, et al. Cancer associated epigenetic transitions identified by genome-wide histone methylation binding profiles in human colorectal cancer samples and paired normal mucosa. BMC Cancer. 2011;11:450. doi: 10.1186/1471-2407-11-450. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Mo Q. A fully Bayesian hidden Ising model for ChIP-seq data analysis. Biostatistics. 2012;13(1):113–28. doi: 10.1093/biostatistics/kxr029. [DOI] [PubMed] [Google Scholar]
  • 92.Wang C, Caron M, Burdick D, Kang Z, Auld D, Hill WA, et al. A sensitive, homogeneous, and high-throughput assay for lysine-specific histone demethylases at the H3K4 site. Assay Drug Dev Technol. 2012;10(2):179–86. doi: 10.1089/adt.2011.0395. [DOI] [PubMed] [Google Scholar]
  • 93.Pellegrini M, Ferrari R. Epigenetic analysis: ChIP-chip and ChIP-seq. Methods Mol Biol. 2012;802:377–87. doi: 10.1007/978-1-61779-400-1_25. [DOI] [PubMed] [Google Scholar]
  • 94.Jayani RS, Ramanujam PL, Galande S. Studying histone modifications and their genomic functions by employing chromatin immunoprecipitation and immunoblotting. Methods Cell Biol. 2010;98:35–56. doi: 10.1016/S0091-679X(10)98002-3. [DOI] [PubMed] [Google Scholar]
  • 95.Liu BL, Cheng JX, Zhang X, Wang R, Zhang W, Lin H, et al. Global histone modification patterns as prognostic markers to classify glioma patients. Cancer Epidemiol Biomarkers Prev. 2010;19(11):2888–96. doi: 10.1158/1055-9965.EPI-10-0454. [DOI] [PubMed] [Google Scholar]
  • 96.Viertler C, Groelz D, Gundisch S, Kashofer K, Reischauer B, Riegman PH, et al. A new technology for stabilization of biomolecules in tissues for combined histological and molecular analyses. J Mol Diagn. 2012;14(5):458–66. doi: 10.1016/j.jmoldx.2012.05.002. [DOI] [PubMed] [Google Scholar]
  • 97.Gordanpour A, Nam RK, Sugar L, Bacopulos S, Seth A. MicroRNA detection in prostate tumors by quantitative real-time PCR (qPCR) J Vis Exp. 2012;(63):e3874. doi: 10.3791/3874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Borgan E, Navon R, Vollan HK, Schlichting E, Sauer T, Yakhini Z, et al. Ischemia caused by time to freezing induces systematic microRNA and mRNA responses in cancer tissue. Mol Oncol. 2011;5(6):564–76. doi: 10.1016/j.molonc.2011.08.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Li J, Smyth P, Flavin R, Cahill S, Denning K, Aherne S, et al. Comparison of miRNA expression patterns using total RNA extracted from matched samples of formalin-fixed paraffin-embedded (FFPE) cells and snap frozen cells. BMC Biotechnol. 2007;7:36. doi: 10.1186/1472-6750-7-36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Hunter MP, Ismail N, Zhang X, Aguda BD, Lee EJ, Yu L, et al. Detection of microRNA expression in human peripheral blood microvesicles. PLoS One. 2008;3(11):e3694. doi: 10.1371/journal.pone.0003694. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Barton RH. A decade of advances in metabonomics. Expert Opin Drug Metab Toxicol. 2011;7(2):129–36. doi: 10.1517/17425255.2011.547473. [DOI] [PubMed] [Google Scholar]
  • 102.Fan L, Zhang W, Yin M, Zhang T, Wu X, Zhang H, et al. Identification of metabolic biomarkers to diagnose epithelial ovarian cancer using a UPLC/QTOF/MS platform. Acta Oncol. 2012;51(4):473–9. doi: 10.3109/0284186X.2011.648338. [DOI] [PubMed] [Google Scholar]
  • 103.Zhang T, Wu X, Yin M, Fan L, Zhang H, Zhao F, et al. Discrimination between malignant and benign ovarian tumors by plasma metabolomic profiling using ultra performance liquid chromatography/mass spectrometry. Clin Chim Acta. 2012;413(9–10):861–8. doi: 10.1016/j.cca.2012.01.026. [DOI] [PubMed] [Google Scholar]
  • 104.Bictash M, Ebbels TM, Chan Q, Loo RL, Yap IK, Brown IJ, et al. Opening up the “Black Box”: metabolic phenotyping and metabolome-wide association studies in epidemiology. J Clin Epidemiol. 2010;63(9):970–9. doi: 10.1016/j.jclinepi.2009.10.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Yap IK, Brown IJ, Chan Q, Wijeyesekera A, Garcia-Perez I, Bictash M, et al. Metabolome-wide association study identifies multiple biomarkers that discriminate north and south Chinese populations at differing risks of cardiovascular disease: INTERMAP study. J Proteome Res. 2010;9(12):6647–54. doi: 10.1021/pr100798r. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Loo RL, Coen M, Ebbels T, Cloarec O, Maibaum E, Bictash M, et al. Metabolic profiling and population screening of analgesic usage in nuclear magnetic resonance spectroscopy-based large-scale epidemiologic studies. Anal Chem. 2009;81(13):5119–29. doi: 10.1021/ac900567e. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Holmes E, Loo RL, Stamler J, Bictash M, Yap IK, Chan Q, et al. Human metabolic phenotype diversity and its association with diet and blood pressure. Nature. 2008;453(7193):396–400. doi: 10.1038/nature06882. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Holmes E, Nicholson JK. Human metabolic phenotyping and metabolome wide association studies. Ernst Schering Found Symp Proc. 2007;(4):227–49. doi: 10.1007/2789_2008_096. [DOI] [PubMed] [Google Scholar]
  • 109.Xiao JF, Varghese RS, Zhou B, Nezami Ranjbar MR, Zhao Y, Tsai TH, et al. LC-MS Based Serum Metabolomics for Identification of Hepatocellular Carcinoma Biomarkers in Egyptian Cohort. J Proteome Res. 2012 doi: 10.1021/pr300673x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Nishiumi S, Kobayashi T, Ikeda A, Yoshie T, Kibi M, Izumi Y, et al. A novel serum metabolomics-based diagnostic approach for colorectal cancer. PLoS One. 2012;7(7):e40459. doi: 10.1371/journal.pone.0040459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Rojo D, Barbas C, Ruperez FJ. LC-MS metabolomics of polar compounds. Bioanalysis. 2012;4(10):1235–43. doi: 10.4155/bio.12.100. [DOI] [PubMed] [Google Scholar]
  • 112.Livengood P, Maciejewski R, Chen W, Ebert DS. OmicsVis: an interactive tool for visually analyzing metabolomics data. BMC Bioinformatics. 2012;13 (Suppl 8):S6. doi: 10.1186/1471-2105-13-S8-S6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Xia J, Mandal R, Sinelnikov IV, Broadhurst D, Wishart DS. MetaboAnalyst 2. 0--a comprehensive server for metabolomic data analysis. Nucleic Acids Res. 2012;40(Web Server issue):W127–33. doi: 10.1093/nar/gks374. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Sugimoto M, Kawakami M, Robert M, Soga T, Tomita M. Bioinformatics Tools for Mass Spectroscopy-Based Metabolomic Data Processing and Analysis. Curr Bioinform. 2012;7(1):96–108. doi: 10.2174/157489312799304431. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Hnatyshyn S, Shipkova P. Automated and unbiased analysis of LC-MS metabolomic data. Bioanalysis. 2012;4(5):541–54. doi: 10.4155/bio.12.9. [DOI] [PubMed] [Google Scholar]
  • 116.Ludwig C, Gunther UL. MetaboLab--advanced NMR data processing and analysis for metabolomics. BMC Bioinformatics. 2011;12:366. doi: 10.1186/1471-2105-12-366. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Biswas A, Mynampati KC, Umashankar S, Reuben S, Parab G, Rao R, et al. MetDAT: a modular and workflow-based free online pipeline for mass spectrometry data processing, analysis and interpretation. Bioinformatics. 2010;26(20):2639–40. doi: 10.1093/bioinformatics/btq436. [DOI] [PubMed] [Google Scholar]
  • 118.Xia J, Wishart DS. MetPA: a web-based metabolomics tool for pathway analysis and visualization. Bioinformatics. 2010;26(18):2342–4. doi: 10.1093/bioinformatics/btq418. [DOI] [PubMed] [Google Scholar]
  • 119.Carroll AJ, Badger MR, Harvey Millar A. The MetabolomeExpress Project: enabling web-based processing, analysis and transparent dissemination of GC/MS metabolomics datasets. BMC Bioinformatics. 2010;11:376. doi: 10.1186/1471-2105-11-376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Goodpaster AM, Romick-Rosendale LE, Kennedy MA. Statistical significance analysis of nuclear magnetic resonance-based metabonomics data. Anal Biochem. 2010;401(1):134–43. doi: 10.1016/j.ab.2010.02.005. [DOI] [PubMed] [Google Scholar]
  • 121.Yuan M, Breitkopf SB, Yang X, Asara JM. A positive/negative ion-switching, targeted mass spectrometry-based metabolomics platform for bodily fluids, cells, and fresh and fixed tissue. Nat Protoc. 2012;7(5):872–81. doi: 10.1038/nprot.2012.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Greider CW. Telomerase discovery: the excitement of putting together pieces of the puzzle (Nobel lecture) Angew Chem Int Ed Engl. 2010;49(41):7422–39. doi: 10.1002/anie.201002408. [DOI] [PubMed] [Google Scholar]
  • 123.Gilson E, Segal-Bendirdjian E. The telomere story or the triumph of an open-minded research. Biochimie. 2010;92(4):321–6. doi: 10.1016/j.biochi.2009.12.014. [DOI] [PubMed] [Google Scholar]
  • 124.Greider CW. Telomeres. Curr Opin Cell Biol. 1991;3(3):444–51. doi: 10.1016/0955-0674(91)90072-7. [DOI] [PubMed] [Google Scholar]
  • 125.Beattie TL, Zhou W, Robinson MO, Harrington L. Functional multimerization of the human telomerase reverse transcriptase. Mol Cell Biol. 2001;21(18):6151–60. doi: 10.1128/MCB.21.18.6151-6160.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Nan H, Qureshi AA, Prescott J, De Vivo I, Han J. Genetic variants in telomere-maintaining genes and skin cancer risk. Hum Genet. 2011;129(3):247–53. doi: 10.1007/s00439-010-0921-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Prescott J, Wentzensen IM, Savage SA, De Vivo I. Epidemiologic evidence for a role of telomere dysfunction in cancer etiology. Mutat Res. 2012;730(1–2):75–84. doi: 10.1016/j.mrfmmm.2011.06.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Hofer P, Baierl A, Feik E, Fuhrlinger G, Leeb G, Mach K, et al. MNS16A tandem repeats minisatellite of human telomerase gene: a risk factor for colorectal cancer. Carcinogenesis. 2011;32(6):866–71. doi: 10.1093/carcin/bgr053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Winnikow EP, Medeiros LR, Edelweiss MI, Rosa DD, Edelweiss M, Simoes PW, et al. Accuracy of telomerase in estimating breast cancer risk: a systematic review and meta-analysis. Breast. 2012;21(1):1–7. doi: 10.1016/j.breast.2011.08.136. [DOI] [PubMed] [Google Scholar]
  • 130.Mirabello L, Yu K, Kraft P, De Vivo I, Hunter DJ, Prescott J, et al. The association of telomere length and genetic variation in telomere biology genes. Hum Mutat. 2010;31(9):1050–8. doi: 10.1002/humu.21314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Johnatty SE, Beesley J, Chen X, Macgregor S, Duffy DL, Spurdle AB, et al. Evaluation of candidate stromal epithelial cross-talk genes identifies association between risk of serous ovarian cancer and TERT, a cancer susceptibility “hot-spot”. PLoS Genet. 2010;6(7):e1001016. doi: 10.1371/journal.pgen.1001016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Petersen GM, Amundadottir L, Fuchs CS, Kraft P, Stolzenberg-Solomon RZ, Jacobs KB, et al. A genome-wide association study identifies pancreatic cancer susceptibility loci on chromosomes 13q22.1, 1q32.1 and 5p15. 33. Nat Genet. 2010;42(3):224–8. doi: 10.1038/ng.522. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Rafnar T, Sulem P, Stacey SN, Geller F, Gudmundsson J, Sigurdsson A, et al. Sequence variants at the TERT-CLPTM1L locus associate with many cancer types. Nat Genet. 2009;41(2):221–7. doi: 10.1038/ng.296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Terry KL, Tworoger SS, Vitonis AF, Wong J, Titus-Ernstoff L, De Vivo I, et al. Telomere length and genetic variation in telomere maintenance genes in relation to ovarian cancer risk. Cancer Epidemiol Biomarkers Prev. 2012;21(3):504–12. doi: 10.1158/1055-9965.EPI-11-0867. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Meeker AK, Gage WR, Hicks JL, Simon I, Coffman JR, Platz EA, et al. Telomere length assessment in human archival tissues: combined telomere fluorescence in situ hybridization and immunostaining. Am J Pathol. 2002;160(4):1259–68. doi: 10.1016/S0002-9440(10)62553-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Zhou X, Meeker AK, Makambi KH, Kosti O, Kallakury BV, Sidawy MK, et al. Telomere length variation in normal epithelial cells adjacent to tumor: potential biomarker for breast cancer local recurrence. Carcinogenesis. 2012;33(1):113–8. doi: 10.1093/carcin/bgr248. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Barwell J, Pangon L, Georgiou A, Docherty Z, Kesterton I, Ball J, et al. Is telomere length in peripheral blood lymphocytes correlated with cancer susceptibility or radiosensitivity? Br J Cancer. 2007;97(12):1696–700. doi: 10.1038/sj.bjc.6604085. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.De Vivo I, Prescott J, Wong JY, Kraft P, Hankinson SE, Hunter DJ. A prospective study of relative telomere length and postmenopausal breast cancer risk. Cancer Epidemiol Biomarkers Prev. 2009;18(4):1152–6. doi: 10.1158/1055-9965.EPI-08-0998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Svenson U, Nordfjall K, Stegmayr B, Manjer J, Nilsson P, Tavelin B, et al. Breast cancer survival is associated with telomere length in peripheral blood cells. Cancer Res. 2008;68(10):3618–23. doi: 10.1158/0008-5472.CAN-07-6497. [DOI] [PubMed] [Google Scholar]
  • 140.Divella R, Daniele A, Gadaleta C, Tufaro A, Venneri MT, Paradiso A, et al. Circulating transforming growth factor-beta and epidermal growth factor receptor as related to virus infection in liver carcinogenesis. Anticancer Res. 2012;32(1):141–5. [PubMed] [Google Scholar]
  • 141.El-Tayeh SF, Hussein TD, El-Houseini ME, Amer MA, El-Sherbini M, Elshemey WM. Serological biomarkers of hepatocellular carcinoma in Egyptian patients. Dis Markers. 2012;32(4):255–63. doi: 10.3233/DMA-2011-0883. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Qu Z, Cui N, Qin M, Wu X. Epidemiological survey of biomarkers of hepatitis virus in patients with extrahepatic cholangiocarcinomas. Asia Pac J Clin Oncol. 2012;8(1):83–7. doi: 10.1111/j.1743-7563.2011.01466.x. [DOI] [PubMed] [Google Scholar]
  • 143.Yu X, Zhang J, Hong L, Wang J, Yuan Z, Zhang X, et al. High prevalence of human parvovirus 4 infection in HBV and HCV infected individuals in shanghai. PLoS One. 2012;7(1):e29474. doi: 10.1371/journal.pone.0029474. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Li G, Ruan X, Auerbach RK, Sandhu KS, Zheng M, Wang P, et al. Extensive promoter-centered chromatin interactions provide a topological basis for transcription regulation. Cell. 2012;148(1–2):84–98. doi: 10.1016/j.cell.2011.12.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Ioannidis JP, Khoury MJ. Improving validation practices in “omics” research. Science. 2011;334(6060):1230–2. doi: 10.1126/science.1211811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Ioannidis JP. A roadmap for successful applications of clinical proteomics. Proteomics Clin Appl. 2011;5(5–6):241–7. doi: 10.1002/prca.201000096. [DOI] [PubMed] [Google Scholar]
  • 147.Castaldi PJ, Dahabreh IJ, Ioannidis JP. An empirical assessment of validation practices for molecular classifiers. Brief Bioinform. 2011;12(3):189–202. doi: 10.1093/bib/bbq073. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Ioannidis JP. Expectations, validity, and reality in omics. J Clin Epidemiol. 2010;63(9):945–9. doi: 10.1016/j.jclinepi.2010.04.002. [DOI] [PubMed] [Google Scholar]
  • 149.Gallo V, Egger M, McCormack V, Farmer PB, Ioannidis JP, Kirsch-Volders M, et al. STrengthening the Reporting of OBservational studies in Epidemiology - Molecular Epidemiology (STROBE-ME): an extension of the STROBE statement. Eur J Clin Invest. 2012;42(1):1–16. doi: 10.1111/j.1365-2362.2011.02561.x. [DOI] [PubMed] [Google Scholar]
  • 150.von Elm E, Altman DG, Egger M, Pocock SJ, Gotzsche PC, Vandenbroucke JP. The Strengthening the Reporting of Observational Studies in Epidemiology (STROBE) statement: guidelines for reporting observational studies. PLoS Med. 2007;4(10):e296. doi: 10.1371/journal.pmed.0040296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Altman DG, McShane LM, Sauerbrei W, Taube SE. Reporting Recommendations for Tumor Marker Prognostic Studies (REMARK): explanation and elaboration. PLoS Med. 2012;9(5):e1001216. doi: 10.1371/journal.pmed.1001216. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Alsheikh-Ali AA, Qureshi W, Al-Mallah MH, Ioannidis JP. Public availability of published research data in high-impact journals. PLoS One. 2011;6(9):e24357. doi: 10.1371/journal.pone.0024357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.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(9):1133–9. doi: 10.1200/JCO.2010.31.3577. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Zhu Y, Stevens RG, Hoffman AE, Tjonneland A, Vogel UB, Zheng T, et al. Epigenetic impact of long-term shiftwork: pilot evidence from circadian genes and whole-genome methylation analysis. Chronobiol Int. 2011;28(10):852–61. doi: 10.3109/07420528.2011.618896. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Bullinger L, Ehrich M, Dohner K, Schlenk RF, Dohner H, Nelson MR, et al. Quantitative DNA methylation predicts survival in adult acute myeloid leukemia. Blood. 2010;115(3):636–42. doi: 10.1182/blood-2009-03-211003. [DOI] [PubMed] [Google Scholar]
  • 156.Hong YS, Kang HJ, Kwak JY, Park BL, You CH, Kim YM, et al. Association between microRNA196a2 rs11614913 genotypes and the risk of non-small cell lung cancer in Korean population. J Prev Med Public Health. 2011;44(3):125–30. doi: 10.3961/jpmph.2011.44.3.125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Leite KR, Tomiyama A, Reis ST, Sousa-Canavez JM, Sanudo A, Dall’Oglio MF, et al. MicroRNA-100 expression is independently related to biochemical recurrence of prostate cancer. J Urol. 2011;185(3):1118–22. doi: 10.1016/j.juro.2010.10.035. [DOI] [PubMed] [Google Scholar]
  • 158.Li J, Wang Y, Yu W, Chen J, Luo J. Expression of serum miR-221 in human hepatocellular carcinoma and its prognostic significance. Biochem Biophys Res Commun. 2011;406(1):70–3. doi: 10.1016/j.bbrc.2011.01.111. [DOI] [PubMed] [Google Scholar]
  • 159.Zhao R, Wu J, Jia W, Gong C, Yu F, Ren Z, et al. Plasma miR-221 as a predictive biomarker for chemoresistance in breast cancer patients who previously received neoadjuvant chemotherapy. Onkologie. 2011;34(12):675–80. doi: 10.1159/000334552. [DOI] [PubMed] [Google Scholar]
  • 160.Lynch SM, Weinstein SJ, Virtamo J, Lan Q, Liu CS, Cheng WL, et al. Mitochondrial DNA copy number and pancreatic cancer in the alpha-tocopherol beta-carotene cancer prevention study. Cancer Prev Res (Phila) 2011;4(11):1912–9. doi: 10.1158/1940-6207.CAPR-11-0002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Audet-Walsh E, Lepine J, Gregoire J, Plante M, Caron P, Tetu B, et al. Profiling of endogenous estrogens, their precursors, and metabolites in endometrial cancer patients: association with risk and relationship to clinical characteristics. J Clin Endocrinol Metab. 2011;96(2):E330–9. doi: 10.1210/jc.2010-2050. [DOI] [PubMed] [Google Scholar]
  • 162.Gallagher RP, Macarthur AC, Lee TK, Weber JP, Leblanc A, Mark Elwood J, et al. Plasma levels of polychlorinated biphenyls and risk of cutaneous malignant melanoma: a preliminary study. Int J Cancer. 2011;128(8):1872–80. doi: 10.1002/ijc.25503. [DOI] [PubMed] [Google Scholar]
  • 163.Bitisik O, Yavuz S, Yasasever V, Dalay N. Telomerase activity in patients with chronic myeloid leukemia and lymphoma. Res Commun Mol Pathol Pharmacol. 2000;107(1–2):3–12. [PubMed] [Google Scholar]
  • 164.Tatsuma T, Goto S, Kitano S, Lin YC, Lee CM, Chen CL. Telomerase activity in peripheral blood for diagnosis of hepatoma. J Gastroenterol Hepatol. 2000;15(9):1064–70. doi: 10.1046/j.1440-1746.2000.02293.x. [DOI] [PubMed] [Google Scholar]
  • 165.van Bemmel D, Lenz P, Liao LM, Baris D, Sternberg LR, Warner AC, et al. Correlation of LINE-1 methylation levels in patient matched buffy coat, serum, buccal cell and bladder tumor tissue DNA samples. Cancer Epidemiol Biomarkers Prev. 2012 doi: 10.1158/1055-9965.EPI-11-1030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Szaumkessel M, Richter J, Giefing M, Jarmuz M, Kiwerska K, Tonnies H, et al. Pyrosequencing-based DNA methylation profiling of Fanconi anemia/BRCA pathway genes in laryngeal squamous cell carcinoma. Int J Oncol. 2011;39(2):505–14. doi: 10.3892/ijo.2011.1039. [DOI] [PubMed] [Google Scholar]
  • 167.Tan D, Goerlitz DS, Dumitrescu RG, Han D, Seillier-Moiseiwitsch F, Spernak SM, et al. Associations between cigarette smoking and mitochondrial DNA abnormalities in buccal cells. Carcinogenesis. 2008;29(6):1170–7. doi: 10.1093/carcin/bgn034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Shen C, Hu L, Xia L, Li Y. The detection of circulating tumor cells of breast cancer patients by using multimarker (Survivin, hTERT and hMAM) quantitative real-time PCR. Clin Biochem. 2009;42(3):194–200. doi: 10.1016/j.clinbiochem.2008.10.016. [DOI] [PubMed] [Google Scholar]
  • 169.Wong CM, Anderton DL, Smith-Schneider S, Wing MA, Greven MC, Arcaro KF. Quantitative analysis of promoter methylation in exfoliated epithelial cells isolated from breast milk of healthy women. Epigenetics. 2010;5(7):645–55. doi: 10.4161/epi.5.7.12961. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Browne EP, Punska EC, Lenington S, Otis CN, Anderton DL, Arcaro KF. Increased promoter methylation in exfoliated breast epithelial cells in women with a previous breast biopsy. Epigenetics. 2011;6(12):1425–35. doi: 10.4161/epi.6.12.18280. [DOI] [PubMed] [Google Scholar]
  • 171.Masayesva BG, Mambo E, Taylor RJ, Goloubeva OG, Zhou S, Cohen Y, et al. Mitochondrial DNA content increase in response to cigarette smoking. Cancer Epidemiol Biomarkers Prev. 2006;15(1):19–24. doi: 10.1158/1055-9965.EPI-05-0210. [DOI] [PubMed] [Google Scholar]
  • 172.Dasgupta S, Shao C, Keane TE, Duberow DP, Mathies RA, Fisher PB, et al. Detection of mitochondrial deoxyribonucleic acid alterations in urine from urothelial cell carcinoma patients. Int J Cancer. 2012;131(1):158–64. doi: 10.1002/ijc.26357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Whittaker RG, Blackwood JK, Alston CL, Blakely EL, Elson JL, McFarland R, et al. Urine heteroplasmy is the best predictor of clinical outcome in the m. 3243A>G mtDNA mutation. Neurology. 2009;72(6):568–9. doi: 10.1212/01.wnl.0000342121.91336.4d. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Antill YC, Mitchell G, Johnson SA, Devereux L, Milner A, Di Iulio J, et al. Gene methylation in breast ductal fluid from BRCA1 and BRCA2 mutation carriers. Cancer Epidemiol Biomarkers Prev. 2010;19(1):265–74. doi: 10.1158/1055-9965.EPI-09-0359. [DOI] [PubMed] [Google Scholar]
  • 175.Krassenstein R, Sauter E, Dulaimi E, Battagli C, Ehya H, Klein-Szanto A, et al. Detection of breast cancer in nipple aspirate fluid by CpG island hypermethylation. Clin Cancer Res. 2004;10(1 Pt 1):28–32. doi: 10.1158/1078-0432.ccr-0410-3. [DOI] [PubMed] [Google Scholar]
  • 176.Jakupciak JP, Maggrah A, Maragh S, Maki J, Reguly B, Maki K, et al. Facile whole mitochondrial genome resequencing from nipple aspirate fluid using MitoChip v2. 0. BMC Cancer. 2008;8:95. doi: 10.1186/1471-2407-8-95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Zhu W, Qin W, Bradley P, Wessel A, Puckett CL, Sauter ER. Mitochondrial DNA mutations in breast cancer tissue and in matched nipple aspirate fluid. Carcinogenesis. 2005;26(1):145–52. doi: 10.1093/carcin/bgh282. [DOI] [PubMed] [Google Scholar]
  • 178.Isaacs C, Cavalli LR, Cohen Y, Pennanen M, Shankar LK, Freedman M, et al. Detection of LOH and mitochondrial DNA alterations in ductal lavage and nipple aspirate fluids from hngh-risk patients. Breast Cancer Res Treat. 2004;84(2):99–105. doi: 10.1023/B:BREA.0000018406.03679.2e. [DOI] [PubMed] [Google Scholar]
  • 179.Thompson PA, Hsu CH, Green S, Stopeck AT, Johnson K, Alberts DS, et al. Sulindac and sulindac metabolites in nipple aspirate fluid and effect on drug targets in a phase I trial. Cancer Prev Res (Phila) 2010;3(1):101–7. doi: 10.1158/1940-6207.CAPR-09-0120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Mannello F, Tonti GA, Pagliarani S, Benedetti S, Canestrari F, Zhu W, et al. The 8-epimer of prostaglandin F(2alpha), a marker of lipid peroxidation and oxidative stress, is decreased in the nipple aspirate fluid of women with breast cancer. Int J Cancer. 2007;120(9):1971–6. doi: 10.1002/ijc.22522. [DOI] [PubMed] [Google Scholar]
  • 181.Kim SJ, Kelly WK, Fu A, Haines K, Hoffman A, Zheng T, et al. Genome-wide methylation analysis identifies involvement of TNF-alpha mediated cancer pathways in prostate cancer. Cancer Lett. 2011;302(1):47–53. doi: 10.1016/j.canlet.2010.12.010. [DOI] [PubMed] [Google Scholar]
  • 182.Peurala H, Greco D, Heikkinen T, Kaur S, Bartkova J, Jamshidi M, et al. MiR-34a expression has an effect for lower risk of metastasis and associates with expression patterns predicting clinical outcome in breast cancer. PLoS One. 2011;6(11):e26122. doi: 10.1371/journal.pone.0026122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Song JS, Kim YS, Kim DK, Park SI, Jang SJ. Global histone modification pattern associated with recurrence and disease-free survival in non-small cell lung cancer patients. Pathol Int. 2012;62(3):182–90. doi: 10.1111/j.1440-1827.2011.02776.x. [DOI] [PubMed] [Google Scholar]
  • 184.Eggers H, Steffens S, Grosshennig A, Becker JU, Hennenlotter J, Stenzl A, et al. Prognostic and diagnostic relevance of hypermethylated in cancer 1 (HIC1) CpG island methylation in renal cell carcinoma. Int J Oncol. 2012;40(5):1650–8. doi: 10.3892/ijo.2012.1367. [DOI] [PubMed] [Google Scholar]
  • 185.Kuhn E, Meeker AK, Visvanathan K, Gross AL, Wang TL, Kurman RJ, et al. Telomere length in different histologic types of ovarian carcinoma with emphasis on clear cell carcinoma. Mod Pathol. 2011;24(8):1139–45. doi: 10.1038/modpathol.2011.67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Chen T, He J, Shen L, Fang H, Nie H, Jin T, et al. The mitochondrial DNA 4,977-bp deletion and its implication in copy number alteration in colorectal cancer. BMC Med Genet. 2011;12:8. doi: 10.1186/1471-2350-12-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Yuan JM, Gao YT, Murphy SE, Carmella SG, Wang R, Zhong Y, et al. Urinary levels of cigarette smoke constituent metabolites are prospectively associated with lung cancer development in smokers. Cancer Res. 2011;71(21):6749–57. doi: 10.1158/0008-5472.CAN-11-0209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Benowitz NL, Dains KM, Dempsey D, Wilson M, Jacob P. Racial differences in the relationship between number of cigarettes smoked and nicotine and carcinogen exposure. Nicotine Tob Res. 2011;13(9):772–83. doi: 10.1093/ntr/ntr072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Torroni A, Wallace DC. Mitochondrial DNA variation in human populations and implications for detection of mitochondrial DNA mutations of pathological significance. J Bioenerg Biomembr. 1994;26(3):261–71. doi: 10.1007/BF00763098. [DOI] [PubMed] [Google Scholar]
  • 190.Bhat A, Koul A, Sharma S, Rai E, Bukhari SI, Dhar MK, et al. The possible role of 10398A and 16189C mtDNA variants in providing susceptibility to T2DM in two North Indian populations: a replicative study. Hum Genet. 2007;120(6):821–6. doi: 10.1007/s00439-006-0272-4. [DOI] [PubMed] [Google Scholar]
  • 191.Xing J, Chen M, Wood CG, Lin J, Spitz MR, Ma J, et al. Mitochondrial DNA content: its genetic heritability and association with renal cell carcinoma. J Natl Cancer Inst. 2008;100(15):1104–12. doi: 10.1093/jnci/djn213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Ye C, Gao YT, Wen W, Breyer JP, Shu XO, Smith JR, et al. Association of mitochondrial DNA displacement loop (CA)n dinucleotide repeat polymorphism with breast cancer risk and survival among Chinese women. Cancer Epidemiol Biomarkers Prev. 2008;17(8):2117–22. doi: 10.1158/1055-9965.EPI-07-2798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Dasgupta S, Soudry E, Mukhopadhyay N, Shao C, Yee J, Lam S, et al. Mitochondrial DNA mutations in respiratory complex-I in never-smoker lung cancer patients contribute to lung cancer progression and associated with EGFR gene mutation. J Cell Physiol. 2012;227(6):2451–60. doi: 10.1002/jcp.22980. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Zheng S, Qian P, Li F, Qian G, Wang C, Wu G, et al. Association of mitochondrial DNA variations with lung cancer risk in a Han Chinese population from southwestern China. PLoS One. 2012;7(2):e31322. doi: 10.1371/journal.pone.0031322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Poage GM, Butler RA, Houseman EA, McClean MD, Nelson HH, Christensen BC, et al. Identification of an epigenetic profile classifier that is associated with survival in head and neck cancer. Cancer Res. 2012;72(11):2728–37. doi: 10.1158/0008-5472.CAN-11-4121-T. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Marsit CJ, Houseman EA, Christensen BC, Gagne L, Wrensch MR, Nelson HH, et al. Identification of methylated genes associated with aggressive bladder cancer. PLoS One. 2010;5(8):e12334. doi: 10.1371/journal.pone.0012334. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Floegel A, Drogan D, Wang-Sattler R, Prehn C, Illig T, Adamski J, et al. Reliability of serum metabolite concentrations over a 4-month period using a targeted metabolomic approach. PLoS One. 2011;6(6):e21103. doi: 10.1371/journal.pone.0021103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Chai W, Bostick RM, Ahearn TU, Franke AA, Custer LJ, Cooney RV. Effects of vitamin D3 and calcium supplementation on serum levels of tocopherols, retinol, and specific vitamin D metabolites. Nutr Cancer. 2012;64(1):57–64. doi: 10.1080/01635581.2012.630552. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Zeleniuch-Jacquotte A, Shore RE, Afanasyeva Y, Lukanova A, Sieri S, Koenig KL, et al. Postmenopausal circulating levels of 2- and 16alpha-hydroxyestrone and risk of endometrial cancer. Br J Cancer. 2011;105(9):1458–64. doi: 10.1038/bjc.2011.381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Sreekumar A, Poisson LM, Rajendiran TM, Khan AP, Cao Q, Yu J, et al. Metabolomic profiles delineate potential role for sarcosine in prostate cancer progression. Nature. 2009;457(7231):910–4. doi: 10.1038/nature07762. [DOI] [PMC free article] [PubMed] [Google Scholar] [Research Misconduct Found]
  • 201.Mocellin S, Verdi D, Pooley KA, Landi MT, Egan KM, Baird DM, et al. Telomerase reverse transcriptase locus polymorphisms and cancer risk: a field synopsis and meta-analysis. J Natl Cancer Inst. 2012;104(11):840–54. doi: 10.1093/jnci/djs222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Zienolddiny S, Skaug V, Landvik NE, Ryberg D, Phillips DH, Houlston R, et al. The TERT-CLPTM1L lung cancer susceptibility variant associates with higher DNA adduct formation in the lung. Carcinogenesis. 2009;30(8):1368–71. doi: 10.1093/carcin/bgp131. [DOI] [PubMed] [Google Scholar]
  • 203.Chang J, Dinney CP, Huang M, Wu X, Gu J. Genetic variants in telomere-maintenance genes and bladder cancer risk. PLoS One. 2012;7(2):e30665. doi: 10.1371/journal.pone.0030665. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Wauters E, Smeets D, Coolen J, Verschakelen J, De Leyn P, Decramer M, et al. The TERT-CLPTM1L locus for lung cancer predisposes to bronchial obstruction and emphysema. Eur Respir J. 2011;38(4):924–31. doi: 10.1183/09031936.00187110. [DOI] [PubMed] [Google Scholar]
  • 205.Pande M, Spitz MR, Wu X, Gorlov IP, Chen WV, Amos CI. Novel genetic variants in the chromosome 5p15. 33 region associate with lung cancer risk. Carcinogenesis. 2011;32(10):1493–9. doi: 10.1093/carcin/bgr136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Lu L, Zhang C, Zhu G, Irwin M, Risch H, Menato G, et al. Telomerase expression and telomere length in breast cancer and their associations with adjuvant treatment and disease outcome. Breast Cancer Res. 2011;13(3):R56. doi: 10.1186/bcr2893. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES