Skip to main content
International Journal of Clinical and Experimental Pathology logoLink to International Journal of Clinical and Experimental Pathology
. 2008 Nov 15;2(4):310–326.

Epigenetics and Epigenetic Alterations in Pancreatic Cancer

Noriyuki Omura 1, Michael Goggins 1,2,3
PMCID: PMC2615589  PMID: 19158989

Abstract

Pancreatic cancer remains a major therapeutic challenge. In 2008, there will be approximately 37,680 new cases and 34,290 deaths attributable to pancreatic cancer in the United States (U.S.), making it the fourth leading cause of cancer-related death. Recent comprehensive pancreatic cancer genome project found that pancreatic adenocarcinomas harbored 63 intragenic mutations or amplifications/homozygous deletions and these alterations clustered in 12 signaling pathways. In addition to widespread genetic alterations, it is now apparent that epigenetic mechanisms are also central to the evolution and progression of human cancers. Since epigenetic silencing processes are mitotically heritable, they can drive neoplastic progression and undergo the same selective pressure as genetic alterations. This review will describe recent developments in cancer epigenetics and their importance in our understanding of pancreatic adenocarcinomas.

Keywords: Epigenetics, pancreatic cancer

Introduction and Background

Pancreatic cancer still remains a major therapeutic challenge. In 2008, there will be approximately 37,680 new cases and 34,290 deaths attributable to pancreatic cancer in the United States (U.S.), making it the fourth leading cause of cancer-related death [1]. For all stages, 1-year survival is 23% and 5-year overall survival from diagnosis is 4%. Median survival for patients with locally advanced disease is 9-12 months and, for patients with metastatic disease, the median survival is 3-6 months. The 5-year survival after curative resection is only 15-25%. Few agents have demonstrated significant benefit for patients with metastatic disease.

Numerous clinical and epidemiologic studies have demonstrated the importance of the early diagnosis of cancer and the early detection of neoplastic precursors as the most effective means of reducing cancer-related mortality. This has thus far been shown for colorectal [2], breast [3], cervical [4], and prostate cancer [5]. For example, as much as 50% of the recent decline in breast cancer mortality can be attributed to mammography and early treatment, particularly the treatment of pre-invasive lesions [6].

In the same fashion, a growing body of evidence supports the view that invasive pancreatic adenocarcinomas arise from histologically well-defined noninvasive lesions within the pancreatic ducts [7-9]. The early detection of these precursors could reduce the incidence and mortality from pancreatic adenocarcinoma. These precursor lesions include microscopic pancreatic intraepithelial neoplasias (PanINs), and macroscopic intraductal papillary mucinous neoplasms (IPMNs) [10] and mucinous cystic neoplasms (MCN) [11]. Initial efforts to identify these precursor lesions among individuals with a strong family history of pancreatic cancer have been successful in so far as these lesions can be identified with careful pancreatic imaging and surgically removed [12-15].

Most pancreatic ductal adenocarcinomas are thought to develop PanINs that progress from low-grade to high grade PanINs to invasive adenocarcinoma through a series of genetic and epigenetic alterations. The most common of these genetic abnormalities include extensive chromosomal losses and gains at selected loci [16] and mutations/deletions of oncogenes and tumor suppressor genes, including KRAS, CDKN1A/p16, TP53, MADH4/SMAD4/DPC4, and BRCA2 [17-22]. In addition, telomere shortening is a common genetic abnormality observed in all stages of PanINs including the vast majority of earliest lesions (PanIN-1A) [23].

More recently, a comprehensive pancreatic cancer genome project was undertaken to profile the genetic abnormalities of pancreatic adenocarcinomas [24]. Twenty-four pancreatic ductal adenocarcinomas were analyzed for somatic mutations in 20,661 protein-coding genes by cycle sequencing with homozygous deletions and gene amplifications detected using Illumina 1Mb SNP arrays. Pancreatic adenocarcinomas harbored 63 intragenic mutations or amplifications/homozygous deletions and these alterations clustered in 12 signaling pathways [24].

In addition to the widespread genetic alterations, it is now apparent that epigenetic mechanisms are also central to the evolution and progression of human cancers [25-29]. Since epigenetic silencing processes are mitotically heritable, they can drive neoplastic progression and undergo the same selective pressure as genetic alterations. Indeed, pancreatic cancers harbor numerous epigenetic alterations and these alterations can be observed in PanINs [30, 31] and IPMNs [31, 32] and their prevalence increases as lesions become of more advanced grade. This review will describe recent developments in cancer epigenetics and their importance in our understanding of pancreatic adenocarcinoma.

Epigenetics

Epigenetics is defined as heritable changes in gene expression that are not accompanied by changes in DNA sequence. There are two main categories of epigenetic mechanisms that affect mammalian gene expression at the chromatin level and fulfill the criterion of heritability: DNA methylation and histone modification.

DNA Methylation

DNA methylation in the mammalian genome arises due to covalent addition of a methyl group to the 5′ position of cytosine in the context of the palindromic dinucleotide, CpG. This modification is established and maintained by a family of DNA methyltransferases (DNMTs), DNMT1, DNMT3a and DNMT3b [33-35]. DNMT1 binds and methylates the daughter strands of newly replicated DNA to preserve the parental methylation pattern [36]. The other two enzymes, DNMT3a and DNMT3b, function primarily as de novo methyltransferases and establish genome methylation during pre-implantation [37, 38]. DNA methylation is crucial for mammalian development; neither DNMT1 nor DNMT3b homozygous knockout mice are viable, and DNMT3a knockout mice die 4 weeks after birth [38]. Patients with biallelic mutations in DNMT3b develop ICF syndrome (immunodeficiency, centromere instability and facial anomalies) [39]. Somatic knockout of DNMT1 and DNMT3b results in complete DNA hypomethylation but single knockouts still retain DNA methylation indicating an ability of these enzymes to compensate for each other [40]. DNMT1 overexpression has been described in certain cancers [41-44] and Li et al have found that most pancreatic cancers harbor overexpression of DNMT1 (unpublished).

CpG Islands

There are two patterns of CpG methylation in normal human somatic cells: a majority of the genome (∼98%) in which CpGs are relatively sparse (on average 1 per 100 base pairs (bp)) but highly methylated (approximately 80% of all CpG sites) [45], and a minor fraction (∼2%) that comprises short stretches of DNA (∼1,000 bp) with high CpG density (∼1 per 10 bp) and largely methylation-free [46], known as CpG islands (CGIs). CpG islands were originally defined as regions of 200 bases or more with a (G+C)-content of at least 50% and a ratio of observed CpG frequency to expected CpG frequency of at least 0.6 [47]. There are fewer CpGs in the genome than predicted by its GC content and this pattern is thought to have arisen during evolution because methylated CpGs are liable to undergo spontaneous deamination and mutation to thymine. A more strict definition of CpG islands, the Takai-Jones criteria, provides a better association of CpG islands with 5′ regions of genes and excludes most Alu repeats [48]. Unmethylated CpG islands often localize to the transcriptional start sites of genes.

Approximately 60% of human genes are associated with CpG islands, most of which were once thought to be unmethylated in all tissue types except in certain circumstances (e.g. genomic imprinting and X chromosome inactivation) [49]. However, it is clear that there are normal tissue-specific patterns of CpG island methylation and some CpG island are prone to progressive methylation during aging and during the development of certain diseases such as cancer [50-52].

Recently, a comprehensive analysis of the methylation profile of CpG islands of normal tissues has been reported. Of 16,000 human promoters examined 3% of transcription start site (TSS)-associated CpG islands were normally methylated in somatic tissues [53]. Similarly, Illingworth et al reported that 6-8% of TSS-associated CpG islands were partially methylated [54]. In addition, Esteller et al highlighted the importance of environmental influences on DNA methylation by observing that twins can have significantly different lymphocyte methylation patterns [55]. Similarly, Matsubayashi found that normal duodenal mucosa from patients with pancreatic cancer was more likely to show CpG island methylation than patients with chronic pancreatitis even after adjusting for age [56].

In addition, many promoters that lack strictly defined CpG islands have nonetheless been shown to have tissue specific methylation patterns that strongly correlate with transcriptional activity [48, 57, 58]. For example, Sato et al demonstrated that many commonly overexpressed genes in pancreatic cancers undergo hypomethylation of their normally methylated CpG poor promoters [59]. Eckhardt et al using high-resolution methylation profiles of human chromosomes 6, 20, and 22 reported that genes without a CpG island in their 5′untranslated region showed an inverse correlation between mRNA expression and methylation [60]. The functional relevance of such CpG poor promoters and their methylation awaits further study [27, 61, 62].

Some genes do not display typical patterns of CpG methylation and gene expression. For example, hTERT encodes the catalytic subunit of telomerase, is expressed in most of telomerase positive tumors and its promoter CpG island is hypermethylated in many cancers and hypomethylated in telomerase negative normal tissues [62-64]. Recently, Renaud et al concluded that hTERT expression is induced when the hTERT CpG island is sufficiently hypermethylated to avoid binding of the CTCF repressor yet hypomethylated at certain CpGs thereby enabling the transcription complex to be formed [65]. Taken together, these studies highlight the fact that the functional significance of CpG islands cannot be reliably predicted by computational predictions of CpG density but require experimental evaluation.

Alterations in CpG Island Methylation in Pancreatic Cancer

Several of the classic tumor suppressor genes show aberrant promoter CpG island hypermethylation in a subset of pancreatic cancers. In addition, an increasing number of functionally important genes have been identified that undergo promoter methylation and transcriptional silencing in pancreatic and other cancers. The first classic tumor suppressor gene shown to undergo promoter methylation and gene silencing was the p16/CDKN1A gene where epigenetic silencing is limited to genes that are not targeted for genetic inactivation [66]. Other genetically inactivated tumor suppressor genes in pancreatic cancers, such as p53, SMAD4 and STK11 have not been found to undergo silencing by DNA methylation. Similarly, few of the classic tumor suppressor genes and DNA repair genes that are mutated in other cancers have been shown to undergo epigenetic silencing in pancreatic cancers. hMLH1 has been shown to undergo methylation in a small percentage of pancreatic cancers [67-69] and is associated with microsatellite instability that is sometimes associated with a medullary histology [70, 71]. E-cadherin has been shown to be an infrequent target of DNA methylation [69] and gene silencing and E-cadherin silencing is associated with an undifferentiated phenotype and a poor prognosis [72]. But other classic tumor suppressors such as VHL, Rb, PTEN and BRCA1 have not been found to be epigenetically silenced in pancreatic cancer. More recent studies have used a variety of gene discovery approaches to systematically identify genes that undergo promoter CpG island methylation.

Ueki et al used methylated CpG island amplification (MCA) coupled with representational difference analysis (RDA) and identified the gene preproenkephalin (ppENK), which encodes a native opioid peptide with growth-suppressor properties, and is methylated in most pancreatic cancers [73]. A similar approach was later used by Hagihara [74]. Sato et al compared global gene expression profiles of pancreatic cancer cell lines before and after treatment with the DNA methyltransferase inhibitor, 5-aza-2′-deoxycytidine (5-Aza-dC) and identified 475 candidate genes induced in four pancreatic cancer cell lines, but not in a non-neoplastic pancreatic ductal epithelial cell line, and subsequent analyses confirmed aberrant hypermethylation of several genes in primary pancreatic cancers [75, 76]. Recently, we reported that an MCA approach applied to Agilent 44K promoter microarrays identified 606 genes differentially methylated in a pancreatic cancer cell line, compared to normal pancreas. This assay strategy also demonstrated high reproducibility and accuracy [77].

These approaches have led to the identification of several genes with tumor suppressor properties that are commonly inactivated in pancreatic cancers. For example, Sato et al compared the global gene expression profiles of IPMNs with that of normal pancreatic ductal epithelium samples and identified the cyclin-dependent kinase inhibitor, CDKN1C/p57KIP2 as an underexpressed gene. CDKN1C is a potent inhibitor of several G1 cyclin complexes, and is a negative regulator of cell proliferation [78, 79]. CDKN1C is an imprinted gene located on chromosome 11p15.5, and a tumor suppressor whose inactivation leads to Wilm' tumor, and Beckwith-Wiedemann syndrome. Partial methylation of the CDKN1C promoter CpG island is commonly observed in pancreatic cancer cell lines and IPMNs with reduced CDKN1C expression. In addition to methylation of the CDKN1C promoter, complete hypomethylation of LIT1, an imprinting control region important for the regulation of CDKN1C expression, was detected in some pancreatic cancers as a result of deletion of the methylated LIT1 allele at 11p15.5 rather than through loss of the epigenetic marks of imprinting [80].

SPARC (secreted protein acidic and rich in cysteine, or osteonectin/BM40) is a gene that was first identified as aberrantly methylated in pancreatic cancer by treating pancreatic cancer cells with demethylating agents [81]. SPARC is a calcium-binding protein that interacts with extracellular matrix whose expression is often lost in pancreatic cancer cells through aberrant DNA methylation [81]. SPARC knockout mice grow cancers faster than mice expressing SPARC [82, 83], highlighting its growth inhibitory functions. SPARC influences cell migration, proliferation, angiogenesis (especially during wound healing), matrix cell adhesion, and tissue remodeling [81, 84]. Interestingly, juxta-tumoral fibroblasts often express SPARC and patients whose pancreatic cancers express SPARC in their peritumoral fibroblasts have a poorer prognosis [85].

Tissue factor pathway inhibitor 2 (TFPI-2) is a Kunitz-type serine proteinase inhibitor that acts against a wide range of proteases [86], and is thought to protect the matrix from degradation thereby counteracting tumor invasion and metastasis [87-89]. Sato et al identified aberrant methylation of TFPI-2 in 73% (102/140) of pancreatic cancer xenografts and primary pancreatic adenocarcinomas. Restored expression of the TFPI-2 gene in nonexpressing pancreatic cancer cells resulted in marked suppression in their proliferation, migration, and invasive potential in vitro [90].

Several genes of the GATA family have been investigated for epigenetic alterations in pancreatic cancer as other types of cancer. Fu et al demonstrated that GATA-5 was frequently methylated in pancreatic cancers, whereas GATA-4 was infrequently methylated and often overexpressed relative to normal ductal epithelium [91].

Another gene commonly silenced epigenetically in pancreatic cancer is BNIP3. Silencing of BNIP3 was associated with CpG island methylation in the region of the transcription start site and treatment with the DNA methyltransferase inhibitor, 5-AZA-dC, restored hypoxic BNIP3 expression and reversed resistance to hypoxia-induced death [92, 93]. Suppression of BNIP3 expression in cell lines with RNAi also decreased sensitivity to 5-fluorouracil and gemcitabine, implicating BNIP3 silencing as a potential drug resistance mechanism [94].

Some other targets of aberrant methylation in pancreatic cancer include TSLC1 [95], HHIP [96], MUC2 [97], reprimo [98], CXCR4 [99] and SOCS1 [100].

DNA Hypomethylation in Cancer

Although much of the focus of cancer epigenetics is on the inactivation of tumor suppressor genes by promoter methylation, the earliest observations of aberrant methylation in human cancer identified DNA hypomethylation. The global methylated cytosine content is often reduced in cancer including pancreatic cancers [101] and this hypomethylation occurs both at normally methylated repeat sequences throughout the genome. Global DNA hypomethylation was first described in the early 1980s [102, 103], yet the cause of the global hypomethylation found in cancers is not well understood. One consequence of genome wide hypomethylation may be genomic instability, a characteristic of most pancreatic and other cancers [104, 105]. Interfering with DNA methylation functions such as by knocking out DNA methyltransferase leads to DNA hypomethylation and genetic instability [106]. Indeed, p53 deficient animals hypomorphic for DNMT1 are more likely to get sarcomas [107]. However, in some experimental settings genomic DNA hypermethylation may be more important than hypomethylation; Apc min mice hypomorphic for DNMT1 have a reduced risk of gastrointestinal neoplasia [108].

One cause of DNA hypomethylation is nutritional deficiency. Low folate status or defective folate or vitamin B12 metabolism causes methyl group deficiency which is thought to cause genetic instability by lowering S-adenosyl-methionine concentration, reducing global DNA methylation and the synthesis of thymidine from uracil [109]. Uracil misincorporation in place of thymidine leads to an imbalanced nucleotide pool and increased occurrence of DNA strand breaks [110], which increases genomic instability [111] and is thought to contribute to cancer development. Low vitamin B12 levels or low folic acid intake is a risk factor for pancreatic cancer [112]. In addition, Matsubayashi found that cancer with defective MTHFR genotypes are associated with higher levels of chromosomal losses and reduced levels of LINE1 element methylation [101]. Such observations raise the possibility that nutritional causes of methyl group deficiency could accelerate tumorigenesis in neoplasms that are genetically unstable and have already lost or inactivated genes such as MTHFR involved in methyl group metabolism.

Gene Hypomethylation and Overexpression

DNA hypomethylation also occurs at the 5′ regions of certain genes in pancreatic and other cancers and is associated with gene overexpression. For example, Rosty et al identified overexpression of S100A4 was associated with hypomethylation at specific CpG sites within the first intron [113]. In an attempt to identify additional hypomethylation targets in pancreatic cancer, Sato et al examined microarray profiles to identify genes that were overexpressed in pancreatic cancer but not normal pancreata and normal pancreatic ducts as a screen for hypomethylated genes. Among the identified transcripts, about 200 genes were found to be re-induced after combined treatment with 5Aza-dC and TSA. DNA hypomethylation of promoter CpGs were identified in over half of the genes with this expression pattern including claudin4, lipocalin2, 14-3-3sigma/ stratifin, trefoil factor 2, S100A4, mesothelin, PSCA, S100P and maspin [114].

Methyl-CpG Binding Proteins (MBDs)

The information stored by hypermethylated CpG islands is in part interpreted by methyl-CpG binding proteins (MBDs). MBDs help establish a transcriptionally inactive chromatin environment. This family of proteins consists of five well-characterized members (MeCP2, MBD1, MBD2, MBD3 and MBD4) [115]. MeCP2 was first characterized as a ‘translator’ which represses transcription of methylated DNA through the recruitment of a histone deacetylase (HDAC) complex [116, 117]. Most hypermethylated promoters are occupied by MBD proteins, whereas unmethylated promoters generally lack MBDs. Treatment of cancer cells with a demethylating agent causes CpG island hypomethylation, MBD release and gene re-expression, reinforcing the notion that association of MBDs with methylated promoters is methylation-dependent [118, 119]. It is currently thought that the specific profile of MBD occupancy for hypermethylated CpG islands of tumor-suppressor genes might be gene- and tumor- specific.

MBDs like DNMTs also recruit histone methyltransferases (HMTs) [118]. A limited number of studies have examined the significance of MBDs in pancreatic cancer [120].

Histone Modifications in Cancer

Histones are globular proteins with protruding N-terminal tails that are THE main site of biochemical modifications including acetylation, methylation, phosphorylation, and ubiquitination [118, 121]. The specificity of certain histone modifications to influence transcription led to the ‘histone code hypothesis’ which predicts that histone modifications act sequentially or in combination to alter chromatin structure and form a code that may be read by nuclear factors [122-127]. Lysine acetylation neutralizes the charge between DNA and histone tails and correlates with chromatin accessibility and transcriptional activity. Lysine methylation can have different effects depending on which residue is modified.

Hypermethylated CpG islands of silenced tumor-suppressor genes are known to display a particular histone code characterized by deacetylation of histones H3 and H4, methylation of H3 lysine 9 (H3K9), H3 lysine 27 (H3K27), and H4 lysine 20 (H4K20), and demethylation of H3 lysine 4 [128-131]. On the other hand, methylation of histone H3 lysine 4 (H3K4) and H3 lysine 36 is associated with transcribed chromatin [125, 130]. Histone modifications also function to recruit other effector proteins [132]. Acetylated lysines are recognized by bromodomains within nucleosome remodeling complexes. An interaction between methylated H3K4 and the chromodomain of the helicase Chd1 recruits activating complexes to chromatin. In contrast, methylated H3K9 and H3K27 are bound by heterochromatin protein 1 (HP 1) and Polycomb-group (PcG) proteins, respectively, which mediate chromatin compaction. A given lysine can have up to three methyl groups, and this “methyl state” can influence chromodomain binding. PcG proteins preferentially interact with trimethylated H3K27, while HP1 shows preference for both di- and trimethylated H3K9 [133].

Finally, PcG proteins function as transcriptional repressors that silence genes through chromatin modification [134]. The precise molecular mechanisms of Polycomb repressive complex (PRC)-mediated repression are still poorly understood. The PcG complex can inhibit transcription by preventing ATP-dependent nucleosome remodeling by the SWI/SNF complex as well as by directly blocking the transcription initiation machinery (See review [134]). PcG and MBD proteins collaborate in long-term gene silencing events such as X-chromosome inactivation and imprinting [135]. PcG proteins are thought to serve as recruitment platforms for DNMTs involved in the hypermethylation of tumor suppressor genes [136]. The ability of chromatin modifications to facilitate aberrant DNA methylation highlights uncertainty as to the primacy of DNA methylation or histone modifications as initiators of aberrant epigenetic gene silencing during cancer development.

Histone Modifications in Pancreatic Cancer

A global gene expression analysis of pancreatic cancers before and after treatment of histone deacetylase inhibitors [75, 76] has been described, but overall few studies have examined genes that are regulated by histone modifications in pancreatic cancers.

Genes of the mucin family have been shown to undergo histone alterations in pancreatic cancers in association with gene overexpression. Mucins are high molecular weight glycoproteins several of which are produced by pancreatic cancers. Yamada et al revealed that the 5′ region of MUC1 transcriptional start site (TSS) were enriched in tri/dimethylated H3K9 marks as well as methylated DNA in non-expressing cells [97, 137]. The TSS of MUC2 is highly enriched in di- and tri-methylated H3K4, acetylated H3K9, and acetylated H3K27 in MUC2 expressing pancreatic cancer cells. Vincent et al demonstrated that MUC4 transcription activity was directly affected by DNMT3A, DNMT3B, HDAC1 and HDAC3, DNA methylation and repressive histone code of one region in 5′UTR of MUC4, regarding one Sp1 binding site was significantly associated with MUC1 silencing [138].

Antisense and Epigenetic Regulation

Several microRNAs (miRNAs) have been identified that are regulated by DNA methylation in pancreatic cancer [77] and other cancers [139-141]. Non-coding RNA such as miRNAs and natural antisense transcripts (NATs) are known to be involved in post-transcriptional gene silencing (PTGS). For example, certain miRNAs regulate DNA methylation. miR-29 inhibits DNMT3 activity and transfection of miR-29 into miR-29 negative cell lines inhibits DNA methylation [142].

Transcriptional gene silencing (TGS) caused by small interfering RNAs (siRNA) was first observed in transformed tobacco plants and gene suppression was associated with DNA methylation [143]. Until recently, it was not known if a similar TGS mechanism existed in mammals. However in 2004, Morris et al demonstrated that siRNA targeting the EF1alpha promoter could induce transcriptional silencing and de novo methylation [144]. Recently, Tufarelli et al investigated a rare case of alpha-thalassaemia caused by a deletion that did not affect the HBA2 gene but relocated a LUC7L gene. Its expression gave rise to NATs that overlapped both coding and promoter region of HBA2 and led to aberrant methylation of the promoter CpG island [145].

Although several studies have demonstrated that RNA-dependent transcriptional silencing can occur in mammals, the mechanism for silencing identified is generally been chromatin mediated. Whether or not such silencing alters DNA methylation is controversial. For example, Ting et al has observed that small double stranded RNA targeted to CDH1 in colon cancer cells induced TGS by H3K9 methylation without DNA methylation [146]. Feinberg and Cui et al examined whether cancer cells have aberrant expression of antisense transcripts and if such transcripts cause tumor suppressor gene silencing. They found that some AML cases contained p15 NATs that were associated with chromatin silencing and demonstrated that expression of antisense RNAs from transgenes could silence tumor suppressor gene expression and lead to secondary DNA methylation and that such silencing represented a hit and run effect because it persisted after removal of the antisense RNA [147]. The mechanisms by which noncoding RNAs induce gene silencing are just beginning to be understood. The elucidation of these mechanisms may ultimately lead to therapeutic strategies to epigenetically silence oncogenes.

DNA Methylation Alterations in Pancreatic Cancer Precursors

The timing of epigenetic alterations during cancer development vs cancer progression provides evidence as to their likely significance. Support for the contribution of DNA methylation alterations to pancreatic cancer susceptibility is shown by the fact that many epigenetically silenced genes are silenced in pancreatic cancer precursors. For example, DNA methylation can be detected in PanINs and IPMNs and the prevalence of methylation increases with neoplastic grade [7, 32]. Some of the genes that have been identified as silenced include p16, RELN, TFPI-2 and ppENK [30, 148]. Methylation abnormalities have also been identified in mucinous cystic adenomas of the pancreas [149].

Epigenetic Alterations and Cancer Stem Cells

Recent reviews have emphasized that epigenetic abnormalities might play a seminal role in the earliest steps in cancer initiation [25, 150, 151]. In addition, markers of cancer stem cells are likely to be under epigenetic regulation [152]. Some studies have implicated a stem cell hierarchy to pancreatic cancer cells and to other solid cancers [153], but conclusive evidence that pancreatic cancer stem cells exist has yet to be demonstrated [154], nor have epigenetic influences on cancer stem cells been conclusively demonstrated. Normal stem cells are regulated in part by polycomb proteins that repress gene expression until cellular differentiation. For example, the PcG proteins suppressor of zeste 12 (SUZ12) and embryonic ectoderm development (EED), which form the Polycomb repressive complex 2 (PRC2) are associated with nucleosomes that are trimethylated at H3K27 [155]. Many PcG targets are methylated in adult cancer cells including pancreatic cancer cells [77, 150, 156]. There is also considerable overlap between genes that are aberrantly hypermethylated in cancer are also frequently hypermethylated in stem cells [157]. Ohm et al demonstrated that many polycomb target genes are modified by a ‘bivalent’ promoter chromatin pattern consisting of the repressive mark, methylated H3K27, and an active mark, methylated H3K4 in embryonic stem cells so that they are held in a ’transcriptional ready state [150]. Several groups hypothesized that reversible repression of these regulatory genes by histone modification in stem or progenitor cells might be replaced by permanent silencing by aberrant DNA methylation, locking the cell into a perpetual state of self renewal and thereby predisposing to tumor initiation and subsequent progression [158, 159]. It will be interesting to determine if environmental signals could drive neoplastic development by acting on epigenetic signals in stem cells.

Mechanisms of Aberrant DNA Hypermethylation in Cancer

The causes of aberrant hypermethylation in pancreatic and other cancers remain poorly understood. As indicated above, aging is associated with an increase in DNA methylation in normal tissues that is likely to predispose to the epigenetic silencing of certain tumor suppressor genes [56, 160, 161], and some studies indicate that cancers of older patients have more aberrant methylation than those in younger patients [73]. Precancerous lesions also undergo aberrant methylation that is often present in early stages of neoplasia and increases with neoplastic stage and studies indicate that epigenetic influences can alter the genetic profile of a cancer, supporting the notion that epigenetic influences can occur prior to early genetic events [162, 163]. Some studies have indicated that chronically inflamed tissues may have more aberrant methylation, though interestingly a recent report indicated that ulcerative colitis-associated cancers had low levels of aberrant methylation [164]. One interesting report found that sites of DNA repair could become foci of aberrant methylation as the DNA repair apparatus also recruits DNMTs and SIRT1 [165]. A methylator phenotype (CIMP or CpG island methylator phenotype) has been proposed as a mechanism by which cancers acquire aberrant methylation at hundreds of sites [166]. A small subset of colorectal cancers have been shown to have aberrant methylation of a unique set of genes that do not undergo frequent methylation in normal tissues or other colon cancers and these cancers have a very high prevalence of BRAF mutation, suggesting that there are mechanisms that can lead to a CIMP phenotype [167]. It is not known if dietary or other environmental factors can influence the propensity to hypermethylated CpG islands. In the same fashion that dietary deficiency of methyl groups can lead to hypomethylation, it is possible that dietary excess of methyl groups can cause hypermethylation. For example, the phenotype of the agouti mouse can be modified by its dietary level of methyl groups [168].

Aberrant DNA Methylation as a Diagnostic Marker

Methylation-specific PCR (MSP)-based assays to detect aberrant DNA hypermethylation have been evaluated in patients with pancreatic cancer. Fukushima et al first reported the utility of detecting methylation biomarker in pancreatic juice by MSP. Pancreatic juice samples were collected either intraoperatively, from 92 patients undergoing pancreaticoduodenectomy for benign (n = 20) and malignant periampullary disease (n = 72) or endoscopically (by duodenal aspiration after secretin infusion), and from 13 patients undergoing investigation for pancreatic disease. Methylated ppENK was detected in the pancreatic juice of 30 (66.7%) of 45 patients with pancreatic ductal adenocarcinoma, in 4 (44.4%) of 9 patients with intraductal papillary-mucinous adenocarcinoma, and in 7 (41.2%) of 17 patients with other periampullary carcinomas. Methylated p16 was detected in a lower percentage of these patients (11.1%, 11.1% and 23.5%, respectively). In contrast, methylated ppENK and p16 were not detected in 32 patients with non-malignant periampullary disease. However they found that methylated ppENK and p16 was present in the duodenum of 90.5% and 28.6% and furthermore 88.9% and 11.1%, respectively, of pancreatic juice samples obtained by duodenal aspiration from patients without cancer. They concluded that detection of methylated ppENK and p16 in pure pancreatic juice obtained by direct cannulation of the pancreatic duct to avoid duodenal secretions which normally have methylated DNA [169].

Matsubayashi et al quantified methylated DNA levels in the pancreatic juice of patients with benign and malignant pancreatic disease using quantitative MSP (QMSP) and demonstrated that a concentrations of >1% methylated DNA in 2 or more of 5 QMSP markers (TFPI-2, p16, ppENK, SPARC, NPTX2) was highly accurate in distinguishing patients with pancreatic cancer from those with pancreatitis or a normal pancreas [170]. Yan et al analyzed pancreatic juice for p53 mutations, KRAS mutations and p16 methylation [171]. The p53 mutations were detected with a functional yeast assay in which yeast with transfected mutant p53 PCR products are identified by a red color [171]. Other studies have used MSP to detect methylation alterations of p16, p14, ppENK, NPTX2 and TFPI-2 in pancreatic juice in preliminary studies [172-175]. Parsi et al demonstrated the diagnostic utility of quantifying aberrantly methylated DNA concentration in endoscopic brush samples of biliary strictures. Methylation analysis with QMSP was performed on endoscopically obtained brush samples from the biliary and pancreatic duct from 130 individuals with biliary tract strictures. Using with 3-gene (NPTX-2, Cyclin D2, and TFPI-2) panel, 73.2% of patients with pancreatic adenocarcinoma has positive methylation in 1 or more genes, compared to only 13.6% of individuals with non-neoplastic conditions. They defined a cut-off of at least one gene positive for methylation as >1% methylated TFPI2 and >3% methylated NPTX2 and Cyclin D2 [176].

Although plasma or serum is a very attractive sample for clinical use, and the detection of cancer-associated changes in DNA (mutations, methylation, and rearrangements) in serum, has been reported [177, 178] (for review, see [179]), results to date have been variable [180-182]. Studies evaluating mutant KRAS detection in plasma suggest that such mutant DNA is not readily detected in patients with pancreatic cancer until the disease is more advanced [183], although this may depend on the detection method. It is suspected that circulating tumor DNA is usually released into the bloodstream via necrotic or apoptotic pathways [184, 185], although in some instances it reflects the presence of circulating tumor cells. Interestingly, recent estimations of the concentrations of tumor-derived DNA in plasma, based on the detection of a confirmed tumor-specific mutations, demonstrate that concentrations of circulating mutant DNA is very low (<0.2%) in patients with early stage colon cancer and increases in concentration with tumor stage [186]. Since individual point mutations are not common to every tumor type, aberrant DNA methylation patterns may be a preferred method for identifying DNA alterations derived from cancer. Before such a goal can be realized, more information is needed regarding the methylation landscape of circulating DNA in healthy individuals.

Acknowledgments

This work was supported by the National Cancer Institute grant (CA120432, CA62924), the V foundation and the Michael Rolfe Foundation.

References

  • 1.Jemal A, Siegel R, Ward E, Hao Y, Xu J, Murray T, Thun MJ. Cancer statistics, 2008. CA Cancer J Clin. 2008;58:71–96. doi: 10.3322/CA.2007.0010. [DOI] [PubMed] [Google Scholar]
  • 2.Levin B, Lieberman DA, McFarland B, Andrews KS, Brooks D, Bond J, Dash C, Giardiello FM, Glick S, Johnson D, Johnson CD, Levin TR, Pickhardt PJ, Rex DK, Smith RA, Thorson A, Winawer SJ. Screening and surveillance for the early detection of colorectal cancer and adenomatous polyps, 2008: a joint guideline from the American Cancer Society, the US Multi-Society Task Force on Colorectal Cancer, and the American College of Radiology. Gastroenterology. 2008;134:1570–1595. doi: 10.1053/j.gastro.2008.02.002. [DOI] [PubMed] [Google Scholar]
  • 3.Smith RA, Saslow D, Sawyer KA, Burke W, Costanza ME, Evans WP, 3rd, Foster RS, Jr, Hendrick E, Eyre HJ, Sener S. American Cancer Society guidelines for breast cancer screening: update 2003. CA Cancer J Clin. 2003;53:141–169. doi: 10.3322/canjclin.53.3.141. [DOI] [PubMed] [Google Scholar]
  • 4.Saslow D, Castle PE, Cox JT, Davey DD, Einstein MH, Ferris DG, Goldie SJ, Harper DM, Kinney W, Moscicki AB, Noller KL, Wheeler CM, Ades T, Andrews KS, Doroshenk MK, Kahn KG, Schmidt C, Shafey O, Smith RA, Partridge EE, Garcia F. American Cancer Society Guideline for human papillomavirus (HPV) vaccine use to prevent cervical cancer and its precursors. CA Cancer J Clin. 2007;57:7–28. doi: 10.3322/canjclin.57.1.7. [DOI] [PubMed] [Google Scholar]
  • 5.Andriole GL, Levin DL, Crawford ED, Gelmann EP, Pinsky PF, Chia D, Kramer BS, Reding D, Church TR, Grubb RL, Izmirlian G, Ragard LR, Clapp JD, Prorok PC, Gohagan JK. Prostate Cancer Screening in the Prostate, Lung, Colorectal and Ovarian (PLCO) Cancer Screening Trial: findings from the initial screening round of a randomized trial. J Natl Cancer Inst. 2005;97:433–438. doi: 10.1093/jnci/dji065. [DOI] [PubMed] [Google Scholar]
  • 6.Berry DA, Cronin KA, Plevritis SK, Fryback DG, Clarke L, Zelen M, Mandelblatt JS, Yakovlev AY, Habbema JD, Feuer EJ. Effect of screening and adjuvant therapy on mortality from breast cancer. N Engl J Med. 2005;353:1784–1792. doi: 10.1056/NEJMoa050518. [DOI] [PubMed] [Google Scholar]
  • 7.Hruban RH, Takaori K, Canto M, Fishman EK, Campbell K, Brune K, Kern SE, Goggins M. Clinical importance of precursor lesions in the pancreas. J Hepatobiliary Pancreat Surg. 2007;14:255–263. doi: 10.1007/s00534-006-1170-9. [DOI] [PubMed] [Google Scholar]
  • 8.Singh M, Maitra A. Precursor lesions of pancreatic cancer: molecular pathology and clinical implications. Pancreatology. 2007;7:9–19. doi: 10.1159/000101873. [DOI] [PubMed] [Google Scholar]
  • 9.Hingorani SR. Location, location, location: precursors and prognoses for pancreatic cancer. Gastroenterology. 2007;133:345–350. doi: 10.1053/j.gastro.2007.05.059. [DOI] [PubMed] [Google Scholar]
  • 10.Hruban RH, Takaori K, Klimstra DS, Adsay NV, Albores-Saavedra J, Biankin AV, Biankin SA, Compton C, Fukushima N, Furukawa T, Goggins M, Kato Y, Kloppel G, Longnecker DS, Luttges J, Maitra A, Offerhaus GJ, Shimizu M, Yonezawa S. An illustrated consensus on the classification of pancreatic intraepithelial neoplasia and intraductal papillary mucinous neoplasms. Am J Surg Pathol. 2004;28:977–987. doi: 10.1097/01.pas.0000126675.59108.80. [DOI] [PubMed] [Google Scholar]
  • 11.Iacobuzio-Donahue CA, Wilentz RE, Argani P, Yeo CJ, Cameron JL, Kern SE, Hruban RH. Dpc4 protein in mucinous cystic neoplasms of the pancreas: frequent loss of expression in invasive carcinomas suggests a role in genetic progression. Am J Surg Pathol. 2000;24:1544–1548. doi: 10.1097/00000478-200011000-00011. [DOI] [PubMed] [Google Scholar]
  • 12.Rulyak SJ, Brentnall TA. Inherited pancreatic cancer: improvements in our understanding of genetics and screening. Int J Biochem Cell Biol. 2004;36:1386–1392. doi: 10.1016/j.biocel.2004.02.010. [DOI] [PubMed] [Google Scholar]
  • 13.Canto MI, Goggins M, Hruban RH, Petersen GM, Giardiello FM, Yeo C, Fishman EK, Brune K, Axilbund J, Griffin C, Ali S, Richman J, Jagannath S, Kantsevoy SV, Kalloo AN. Screening for early pancreatic neoplasia in high-risk individuals: a prospective controlled study. Clin Gastroenterol Hepatol. 2006;4:766–781. doi: 10.1016/j.cgh.2006.02.005. quiz 665. [DOI] [PubMed] [Google Scholar]
  • 14.Canto MI, Goggins M, Yeo CJ, Griffin C, Axilbund JE, Brune K, Ali SZ, Jagannath S, Petersen GM, Fishman EK, Piantadosi S, Giardiello FM, Hruban RH. Screening for pancreatic neoplasia in high-risk individuals: an EUS-based approach. Clin Gastroenterol Hepatol. 2004;2:606–621. doi: 10.1016/s1542-3565(04)00244-7. [DOI] [PubMed] [Google Scholar]
  • 15.Brentnall TA, Bronner MP, Byrd DR, Haggitt RC, Kimmey MB. Early diagnosis and treatment of pancreatic dysplasia in patients with a family history of pancreatic cancer. Ann Intern Med. 1999;131:247–255. doi: 10.7326/0003-4819-131-4-199908170-00003. [DOI] [PubMed] [Google Scholar]
  • 16.Yamano M, Fujii H, Takagaki T, Kadowaki N, Watanabe H, Shirai T. Genetic progression and divergence in pancreatic carcinoma. Am J Pathol. 2000;156:2123–2133. doi: 10.1016/S0002-9440(10)65083-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Goggins M, Hruban RH, Kern SE. BRCA2 is inactivated late in the development of pancreatic intraepithelial neoplasia: evidence and implications. Am J Pathol. 2000;156:1767–1771. doi: 10.1016/S0002-9440(10)65047-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Cancer risks in BRCA2 mutation carriers. The Breast Cancer Linkage Consortium. J Natl Cancer Inst. 1999;91:1310–1316. doi: 10.1093/jnci/91.15.1310. [DOI] [PubMed] [Google Scholar]
  • 19.Bardeesy N, Cheng KH, Berger JH, Chu GC, Pahler J, Olson P, Hezel AF, Horner J, Lauwers GY, Hanahan D, DePinho RA. Smad4 is dispensable for normal pancreas development yet critical in progression and tumor biology of pancreas cancer. Genes Dev. 2006;20:3130–3146. doi: 10.1101/gad.1478706. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Hingorani SR, Wang L, Multani AS, Combs C, Deramaudt TB, Hruban RH, Rustgi AK, Chang S, Tuveson DA. Trp53R172H and KrasG12D cooperate to promote chromosomal instability and widely metastatic pancreatic ductal adenocarcinoma in mice. Cancer Cell. 2005;7:469–483. doi: 10.1016/j.ccr.2005.04.023. [DOI] [PubMed] [Google Scholar]
  • 21.Jaffee EM, Hruban RH, Canto M, Kern SE. Focus on pancreas cancer. Cancer Cell. 2002;2:25–28. doi: 10.1016/s1535-6108(02)00093-4. [DOI] [PubMed] [Google Scholar]
  • 22.Maitra A, Hruban RH. Pancreatic cancer. Annu Rev Pathol. 2008;3:157–188. doi: 10.1146/annurev.pathmechdis.3.121806.154305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.van Heek NT, Meeker AK, Kern SE, Yeo CJ, Lillemoe KD, Cameron JL, Offerhaus GJ, Hicks JL, Wilentz RE, Goggins MG, De Marzo AM, Hruban RH, Maitra A. Telomere shortening is nearly universal in pancreatic intraepithelial neoplasia. Am J Pathol. 2002;161:1541–1547. doi: 10.1016/S0002-9440(10)64432-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Jones S, Zhang X, Parsons DW, Lin JC, Leary RJ, Angenendt P, Mankoo P, Carter H, Kamiyama H, Jimeno A, Hong SM, Fu B, Lin MT, Calhoun ES, Kamiyama M, Walter K, Nikolskaya T, Nikolsky Y, Hartigan J, Smith DR, Hidalgo M, Leach SD, Klein AP, Jaffee EM, Goggins M, Maitra A, Iacobuzio-Donahue C, Eshleman JR, Kern SE, Hruban RH, Karchin R, Papadopoulos N, Parmigiani G, Vogelstein B, Velculescu VE, Kinzler KW. Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science. 2008;321:1801–1806. doi: 10.1126/science.1164368. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Feinberg AP, Ohlsson R, Henikoff S. The epigenetic progenitor origin of human cancer. Nat Rev Genet. 2006;7:21–33. doi: 10.1038/nrg1748. [DOI] [PubMed] [Google Scholar]
  • 26.Ting AH, McGarvey KM, Baylin SB. The cancer epigenome–components and functional correlates. Genes Dev. 2006;20:3215–3231. doi: 10.1101/gad.1464906. [DOI] [PubMed] [Google Scholar]
  • 27.Jones PA, Baylin SB. The epigenomics of cancer. Cell. 2007;128:683–692. doi: 10.1016/j.cell.2007.01.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Esteller M. Cancer epigenomics: DNA methylomes and histone-modification maps. Nat Rev Genet. 2007;8:286–298. doi: 10.1038/nrg2005. [DOI] [PubMed] [Google Scholar]
  • 29.Herman JG. Epigenetic changes in cancer and preneoplasia. Cold Spring Harb Symp Quant Biol. 2005;70:329–333. doi: 10.1101/sqb.2005.70.036. [DOI] [PubMed] [Google Scholar]
  • 30.Sato N, Fukushima N, Hruban RH, Goggins M. CpG island methylation profile of pancreatic intraepithelial neoplasia. Mod Pathol. 2008;21:238–244. doi: 10.1038/modpathol.3800991. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Fukushima N, Sato N, Ueki T, Rosty C, Walter KM, Wilentz RE, Yeo CJ, Hruban RH, Goggins M. Aberrant methylation of preproenkephalin and p16 genes in pancreatic intraepithelial neoplasia and pancreatic ductal adenocarcinoma. Am J Pathol. 2002;160:1573–1581. doi: 10.1016/S0002-9440(10)61104-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Sato N, Ueki T, Fukushima N, Iacobuzio-Donahue CA, Yeo CJ, Cameron JL, Hruban RH, Goggins M. Aberrant methylation of CpG islands in intraductal papillary mucinous neoplasms of the pancreas. Gastroenterology. 2002;123:365–372. doi: 10.1053/gast.2002.34160. [DOI] [PubMed] [Google Scholar]
  • 33.Bestor T, Laudano A, Mattaliano R, Ingram V. Cloning and sequencing of a cDNA encoding DNA methyltransferase of mouse cells. The carboxyl-terminal domain of the mammalian enzymes is related to bacterial restriction methyltransferases. J Mol Biol. 1988;203:971–983. doi: 10.1016/0022-2836(88)90122-2. [DOI] [PubMed] [Google Scholar]
  • 34.Yen RW, Vertino PM, Nelkin BD, Yu JJ, el-Deiry W, Cumaraswamy A, Lennon GG, Trask BJ, Celano P, Baylin SB. Isolation and characterization of the cDNA encoding human DNA methyltransferase. Nucleic Acids Res. 1992;20:2287–2291. doi: 10.1093/nar/20.9.2287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Okano M, Xie S, Li E. Cloning and characterization of a family of novel mammalian DNA (cytosine-5) methyltransferases. Nat Genet. 1998;19:219–220. doi: 10.1038/890. [DOI] [PubMed] [Google Scholar]
  • 36.Robert MF, Morin S, Beaulieu N, Gauthier F, Chute IC, Barsalou A, MacLeod AR. DNMT1 is required to maintain CpG methylation and aberrant gene silencing in human cancer cells. Nat Genet. 2003;33:61–65. doi: 10.1038/ng1068. [DOI] [PubMed] [Google Scholar]
  • 37.Dodge JE, Ramsahoye BH, Wo ZG, Okano M, Li E. De novo methylation of MMLV provirus in embryonic stem cells: CpG versus non-CpG methylation. Gene. 2002;289:41–48. doi: 10.1016/s0378-1119(02)00469-9. [DOI] [PubMed] [Google Scholar]
  • 38.Okano M, Bell DW, Haber DA, Li E. DNA methyltransferases Dnmt3a and Dnmt3b are essential for de novo methylation and mammalian development. Cell. 1999;99:247–257. doi: 10.1016/s0092-8674(00)81656-6. [DOI] [PubMed] [Google Scholar]
  • 39.Hansen RS, Wijmenga C, Luo P, Stanek AM, Canfield TK, Weemaes CM, Gartler SM. The DNMT3B DNA methyltransferase gene is mutated in the ICF immunodeficiency syndrome. Proc Natl Acad Sci USA. 1999;96:14412–14417. doi: 10.1073/pnas.96.25.14412. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Rhee I, Bachman KE, Park BH, Jair KW, Yen RW, Schuebel KE, Cui H, Feinberg AP, Lengauer C, Kinzler KW, Baylin SB, Vogelstein B. DNMT1 and DNMT3b cooperate to silence genes in human cancer cells. Nature. 2002;416:552–556. doi: 10.1038/416552a. [DOI] [PubMed] [Google Scholar]
  • 41.Eads CA, Danenberg KD, Kawakami K, Saltz LB, Danenberg PV, Laird PW. CpG island hypermethylation in human colorectal tumors is not associated with DNA methyltransferase overexpression. Cancer Res. 1999;59:2302–2306. [PubMed] [Google Scholar]
  • 42.Patra SK, Patra A, Zhao H, Dahiya R. DNA methyltransferase and demethylase in human prostate cancer. Mol Carcinog. 2002;33:163–171. doi: 10.1002/mc.10033. [DOI] [PubMed] [Google Scholar]
  • 43.Saito Y, Kanai Y, Nakagawa T, Sakamoto M, Saito H, Ishii H, Hirohashi S. Increased protein expression of DNA methyltransferase (DNMT) 1 is significantly correlated with the malignant potential and poor prognosis of human hepatocellular carcinomas. Int J Cancer. 2003;105:527–532. doi: 10.1002/ijc.11127. [DOI] [PubMed] [Google Scholar]
  • 44.Girault I, Tozlu S, Lidereau R, Bieche I. Expression analysis of DNA methyltransferases 1, 3A, and 3B in sporadic breast carcinomas. Clin Cancer Res. 2003;9:4415–4422. [PubMed] [Google Scholar]
  • 45.Ehrlich M, Gama-Sosa MA, Huang LH, Midgett RM, Kuo KC, McCune RA, Gehrke C. Amount and distribution of 5-methylcytosine in human DNA from different types of tissues of cells. Nucleic Acids Res. 1982;10:2709–2721. doi: 10.1093/nar/10.8.2709. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Bird AP. CpG-rich islands and the function of DNA methylation. Nature. 1986;321:209–213. doi: 10.1038/321209a0. [DOI] [PubMed] [Google Scholar]
  • 47.Gardiner-Garden M, Frommer M. CpG islands in vertebrate genomes. J Mol Biol. 1987;196:261–282. doi: 10.1016/0022-2836(87)90689-9. [DOI] [PubMed] [Google Scholar]
  • 48.Takai D, Jones PA. Comprehensive analysis of CpG islands in human chromosomes 21 and 22. Proc Natl Acad Sci USA. 2002;99:3740–3745. doi: 10.1073/pnas.052410099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Antequera F, Bird A. Number of CpG islands and genes in human and mouse. Proc Natl Acad Sci USA. 1993;90:11995–11999. doi: 10.1073/pnas.90.24.11995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Matsubayashi H, Sato N, Brune K, Blackford AL, Hruban RH, Canto M, Yeo CJ, Goggins M. Age- and disease-related methylation of multiple genes in nonneoplastic duodenum and in duodenal juice. Clin Cancer Res. 2005;11:573–583. [PubMed] [Google Scholar]
  • 51.Shiota K, Yanagimachi R. Epigenetics by DNA methylation for development of normal and cloned animals. Differentiation. 2002;69:162–166. doi: 10.1046/j.1432-0436.2002.690406.x. [DOI] [PubMed] [Google Scholar]
  • 52.Bird A. DNA methylation patterns and epigenetic memory. Genes Dev. 2002;16:6–21. doi: 10.1101/gad.947102. [DOI] [PubMed] [Google Scholar]
  • 53.Weber M, Hellmann I, Stadler MB, Ramos L, Paabo S, Rebhan M, Schubeler D. Distribution, silencing potential and evolutionary impact of promoter DNA methylation in the human genome. Nat Genet. 2007;39:457–466. doi: 10.1038/ng1990. [DOI] [PubMed] [Google Scholar]
  • 54.Illingworth R, Kerr A, Desousa D, Jorgensen H, Ellis P, Stalker J, Jackson D, Clee C, Plumb R, Rogers J, Humphray S, Cox T, Langford C, Bird A. A novel CpG island set identifies tissue-specific methylation at developmental gene loci. PLoS Biol. 2008;6:e22. doi: 10.1371/journal.pbio.0060022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Fraga MF, Ballestar E, Paz MF, Ropero S, Setien F, Ballestar ML, Heine-Suner D, Cigudosa JC, Urioste M, Benitez J, Boix-Chornet M, Sanchez-Aguilera A, Ling C, Carlsson E, Poulsen P, Vaag A, Stephan Z, Spector TD, Wu YZ, Plass C, Esteller M. Epigenetic differences arise during the lifetime of monozygotic twins. Proc Natl Acad Sci USA. 2005;102:10604–10609. doi: 10.1073/pnas.0500398102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Matsubayashi H, Sato N, Brune K, Blackford AL, Hruban RH, Canto M, Yeo CJ, Goggins M. Age- and disease-related methylation of multiple genes in nonneoplastic duodenum and in duodenal juice. Clin Cancer Res. 2005;11:573–83. [PubMed] [Google Scholar]
  • 57.Bert T, Lubomierski N, Gangsauge S, Munch K, Printz H, Prasnikar N, Robbel C, Simon B. Expression spectrum and methylation-dependent regulation of melanoma antigen-encoding gene family members in pancreatic cancer cells. Pancreatology. 2002;2:146–154. doi: 10.1159/000055905. [DOI] [PubMed] [Google Scholar]
  • 58.Futscher BW, Oshiro MM, Wozniak RJ, Holtan N, Hanigan CL, Duan H, Domann FE. Role for DNA methylation in the control of cell type specific maspin expression. Nat Genet. 2002;31:175–179. doi: 10.1038/ng886. [DOI] [PubMed] [Google Scholar]
  • 59.Sato N, Maitra A, Fukushima N, van Heek NT, Matsubayashi H, Iacobuzio-Donahue CA, Rosty C, Goggins M. Frequent hypomethylation of multiple genes overexpressed in pancreatic ductal adenocarcinoma. Cancer Res. 2003;63:4158–4166. [PubMed] [Google Scholar]
  • 60.Eckhardt F, Lewin J, Cortese R, Rakyan VK, Attwood J, Burger M, Burton J, Cox TV, Davies R, Down TA, Haefliger C, Horton R, Howe K, Jackson DK, Kunde J, Koenig C, Liddle J, Niblett D, Otto T, Pettett R, Seemann S, Thompson C, West T, Rogers J, Olek A, Berlin K, Beck S. DNA methylation profiling of human chromosomes 6, 20 and 22. Nat Genet. 2006;38:1378–1385. doi: 10.1038/ng1909. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Bernstein BE, Meissner A, Lander ES. The mammalian epigenome. Cell. 2007;128:669–681. doi: 10.1016/j.cell.2007.01.033. [DOI] [PubMed] [Google Scholar]
  • 62.Renaud S, Bosman FT, Benhattar J. Implication of the exon region in the regulation of the human telomerase reverse transcriptase gene promoter. Biochem Biophys Res Commun. 2003;300:47–54. doi: 10.1016/s0006-291x(02)02775-4. [DOI] [PubMed] [Google Scholar]
  • 63.Zinn RL, Pruitt K, Eguchi S, Baylin SB, Herman JG. hTERT is expressed in cancer cell lines despite promoter DNA methylation by preservation of unmethylated DNA and active chromatin around the transcription start site. Cancer Res. 2007;67:194–201. doi: 10.1158/0008-5472.CAN-06-3396. [DOI] [PubMed] [Google Scholar]
  • 64.Dessain SK, Yu H, Reddel RR, Beijersbergen RL, Weinberg RA. Methylation of the human telomerase gene CpG island. Cancer Res. 2000;60:537–541. [PubMed] [Google Scholar]
  • 65.Renaud S, Loukinov D, Abdullaev Z, Guilleret I, Bosman FT, Lobanenkov V, Benhattar J. Dual role of DNA methylation inside and outside of CTCF-binding regions in the transcriptional regulation of the telomerase hTERT gene. Nucleic Acids Res. 2007;35:1245–1256. doi: 10.1093/nar/gkl1125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Schutte M, Hruban RH, Geradts J, Maynard R, Hilgers W, Rabindran SK, Moskaluk CA, Hahn SA, Schwarte-Waldhoff I, Schmiegel W, Baylin SB, Kern SE, Herman JG. Abrogation of the Rb/p16 tumor-suppressive pathway in virtually all pancreatic carcinomas. Cancer Res. 1997;57:3126–3130. [PubMed] [Google Scholar]
  • 67.Yamamoto H, Itoh F, Nakamura H, Fukushima H, Sasaki S, Perucho M, Imai K. Genetic and clinical features of human pancreatic ductal adenocarcinomas with widespread microsatellite instability. Cancer Res. 2001;61:3139–3144. [PubMed] [Google Scholar]
  • 68.Nakata B, Wang YQ, Yashiro M, Nishioka N, Tanaka H, Ohira M, Ishikawa T, Nishino H, Hirakawa K. Prognostic value of microsatellite instability in resectable pancreatic cancer. Clin Cancer Res. 2002;8:2536–2540. [PubMed] [Google Scholar]
  • 69.Ueki T, Toyota M, Sohn T, Yeo CJ, Issa JP, Hruban RH, Goggins M. Hypermethylation of multiple genes in pancreatic adenocarcinoma. Cancer Res. 2000;60:1835–1839. [PubMed] [Google Scholar]
  • 70.Wilentz RE, Goggins M, Redston M, Marcus VA, Adsay NV, Sohn TA, Kadkol SS, Yeo CJ, Choti M, Zahurak M, Johnson K, Tascilar M, Offerhaus GJ, Hruban RH, Kern SE. Genetic, immunohistochemical, and clinical features of medullary carcinoma of the pancreas: A newly described and characterized entity. Am J Pathol. 2000;156:1641–1651. doi: 10.1016/S0002-9440(10)65035-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Goggins M, Offerhaus GJ, Hilgers W, Griffin CA, Shekher M, Tang D, Sohn TA, Yeo CJ, Kern SE, Hruban RH. Pancreatic adenocarcinomas with DNA replication errors (RER+) are associated with wild-type K-ras and characteristic histopathology. Poor differentiation, a syncytial growth pattern, and pushing borders suggest RER+ Am J Pathol. 1998;152:1501–1507. [PMC free article] [PubMed] [Google Scholar]
  • 72.Winter JM, Ting AH, Vilardell F, Gallmeier E, Baylin SB, Hruban RH, Kern SE, Iacobuzio-Donahue CA. Absence of E-cadherin expression distinguishes noncohesive from cohesive pancreatic cancer. Clin Cancer Res. 2008;14:412–418. doi: 10.1158/1078-0432.CCR-07-0487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Ueki T, Toyota M, Skinner H, Walter KM, Yeo CJ, Issa JP, Hruban RH, Goggins M. Identification and characterization of differentially methylated CpG islands in pancreatic carcinoma. Cancer Res. 2001;61:8540–8546. [PubMed] [Google Scholar]
  • 74.Hagihara A, Miyamoto K, Furuta J, Hiraoka N, Wakazono K, Seki S, Fukushima S, Tsao MS, Sugimura T, Ushijima T. Identification of 27 5′ CpG islands aberrantly methylated and 13 genes silenced in human pancreatic cancers. Oncogene. 2004;23:8705–8710. doi: 10.1038/sj.onc.1207783. [DOI] [PubMed] [Google Scholar]
  • 75.Sato N, Fukushima N, Maitra A, Matsubayashi H, Yeo CJ, Cameron JL, Hruban RH, Goggins M. Discovery of novel targets for aberrant methylation in pancreatic carcinoma using high-throughput microarrays. Cancer Res. 2003;63:3735–3742. [PubMed] [Google Scholar]
  • 76.Sato N, Fukushima N, Chang R, Matsubayashi H, Goggins M. Differential and epigenetic gene expression profiling identifies frequent disruption of the RELN pathway in pancreatic cancers. Gastroenterology. 2006;130:548–565. doi: 10.1053/j.gastro.2005.11.008. [DOI] [PubMed] [Google Scholar]
  • 77.Omura N, Li CP, Li A, Hong SM, Walter K, Jimeno A, Hidalgo M, Goggins M. Genome-wide profiling of methylated promoters in pancreatic adenocarcinoma. Cancer Biol Ther. 2008;7:1146–1156. doi: 10.4161/cbt.7.7.6208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Matsuoka S, Edwards MC, Bai C, Parker S, Zhang P, Baldini A, Harper JW, Elledge SJ. p57KIP2, a structurally distinct member of the p21CIP1 Cdk inhibitor family, is a candidate tumor suppressor gene. Genes Dev. 1995;9:650–662. doi: 10.1101/gad.9.6.650. [DOI] [PubMed] [Google Scholar]
  • 79.Lee MH, Reynisdottir I, Massague J. Cloning of p57KIP2, a cyclin-dependent kinase inhibitor with unique domain structure and tissue distribution. Genes Dev. 1995;9:639–649. doi: 10.1101/gad.9.6.639. [DOI] [PubMed] [Google Scholar]
  • 80.Sato N, Matsubayashi H, Abe T, Fukushima N, Goggins M. Epigenetic down-regulation of CDKN1C/p57KIP2 in pancreatic ductal neoplasms identified by gene expression profiling. Clin Cancer Res. 2005;11:4681–4688. doi: 10.1158/1078-0432.CCR-04-2471. [DOI] [PubMed] [Google Scholar]
  • 81.Sato N, Fukushima N, Maehara N, Matsubayashi H, Koopmann J, Su GH, Hruban RH, Goggins M. SPARC/osteonectin is a frequent target for aberrant methylation in pancreatic adenocarcinoma and a mediator of tumor-stromal interactions. Oncogene. 2003;22:5021–5030. doi: 10.1038/sj.onc.1206807. [DOI] [PubMed] [Google Scholar]
  • 82.Mok SC, Chan WY, Wong KK, Muto MG, Berkowitz RS. SPARC, an extracellular matrix protein with tumor-suppressing activity in human ovarian epithelial cells. Oncogene. 1996;12:1895–1901. [PubMed] [Google Scholar]
  • 83.Schultz C, Lemke N, Ge S, Golembieski WA, Rempel SA. Secreted protein acidic and rich in cysteine promotes glioma invasion and delays tumor growth in vivo. Cancer Res. 2002;62:6270–6277. [PubMed] [Google Scholar]
  • 84.Lane TF, Sage EH. The biology of SPARC, a protein that modulates cell-matrix interactions. Faseb J. 1994;8:163–173. [PubMed] [Google Scholar]
  • 85.Infante JR, Matsubayashi H, Sato N, Tonascia J, Klein AP, Riall TA, Yeo C, Iacobuzio-Donahue C, Goggins M. Peritumoral fibroblast SPARC expression and patient outcome with resectable pancreatic adenocarcinoma. J Clin Oncol. 2007;25:319–325. doi: 10.1200/JCO.2006.07.8824. [DOI] [PubMed] [Google Scholar]
  • 86.Petersen LC, Sprecher CA, Foster DC, Blumberg H, Hamamoto T, Kisiel W. Inhibitory properties of a novel human Kunitz-type protease inhibitor homologous to tissue factor pathway inhibitor. Biochemistry. 1996;35:266–272. doi: 10.1021/bi951501d. [DOI] [PubMed] [Google Scholar]
  • 87.Izumi H, Takahashi C, Oh J, Noda M. Tissue factor pathway inhibitor-2 suppresses the production of active matrix metalloproteinase-2 and is down-regulated in cells harboring activated ras oncogenes. FEBS Lett. 2000;481:31–36. doi: 10.1016/s0014-5793(00)01902-5. [DOI] [PubMed] [Google Scholar]
  • 88.Jin M, Udagawa K, Miyagi E, Nakazawa T, Hirahara F, Yasumitsu H, Miyazaki K, Nagashima Y, Aoki I, Miyagi Y. Expression of serine proteinase inhibitor PP5/TFPI-2/MSPI decreases the invasive potential of human choriocarcinoma cells in vitro and in vivo. Gynecol Oncol. 2001;83:325–333. doi: 10.1006/gyno.2001.6394. [DOI] [PubMed] [Google Scholar]
  • 89.Konduri SD, Srivenugopal KS, Yanamandra N, Dinh DH, Olivero WC, Gujrati M, Foster DC, Kisiel W, Ali-Osman F, Kondraganti S, Lakka SS, Rao JS. Promoter methylation and silencing of the tissue factor pathway inhibitor-2 (TFPI-2), a gene encoding an inhibitor of matrix metalloproteinases in human glioma cells. Oncogene. 2003;22:4509–4516. doi: 10.1038/sj.onc.1206695. [DOI] [PubMed] [Google Scholar]
  • 90.Sato N, Parker AR, Fukushima N, Miyagi Y, Iacobuzio-Donahue CA, Eshleman JR, Goggins M. Epigenetic inactivation of TFPI-2 as a common mechanism associated with growth and invasion of pancreatic ductal adenocarcinoma. Oncogene. 2005;24:850–858. doi: 10.1038/sj.onc.1208050. [DOI] [PubMed] [Google Scholar]
  • 91.Fu B, Guo M, Wang S, Campagna D, Luo M, Herman JG, Iacobuzio-Donahue CA. Evaluation of GATA-4 and GATA-5 methylation profiles in human pancreatic cancers indicate promoter methylation patterns distinct from other human tumor types. Cancer Biol Ther. 2007;6:1546–1552. doi: 10.4161/cbt.6.10.4708. [DOI] [PubMed] [Google Scholar]
  • 92.Abe T, Toyota M, Suzuki H, Murai M, Akino K, Ueno M, Nojima M, Yawata A, Miyakawa H, Suga T, Ito H, Endo T, Tokino T, Hinoda Y, Imai K. Upregulation of BNIP3 by 5-aza-2′-deoxycytidine sensitizes pancreatic cancer cells to hypoxia-mediated cell death. J Gastroenterol. 2005;40:504–510. doi: 10.1007/s00535-005-1576-1. [DOI] [PubMed] [Google Scholar]
  • 93.Okami J, Simeone DM, Logsdon CD. Silencing of the hypoxia-inducible cell death protein BNIP3 in pancreatic cancer. Cancer Res. 2004;64:5338–5346. doi: 10.1158/0008-5472.CAN-04-0089. [DOI] [PubMed] [Google Scholar]
  • 94.Akada M, Crnogorac-Jurcevic T, Lattimore S, Mahon P, Lopes R, Sunamura M, Matsuno S, Lemoine NR. Intrinsic chemoresistance to gemcitabine is associated with decreased expression of BNIP3 in pancreatic cancer. Clin Cancer Res. 2005;11:3094–3101. doi: 10.1158/1078-0432.CCR-04-1785. [DOI] [PubMed] [Google Scholar]
  • 95.Jansen M, Fukushima N, Rosty C, Walter K, Altink R, Heek TV, Hruban R, Offerhaus JG, Goggins M. Aberrant methylation of the 5′ CpG island of TSLC1 is common in pancreatic ductal adenocarcinoma and is first manifest in high-grade PanlNs. Cancer Biol Ther. 2002;1:293–296. doi: 10.4161/cbt.84. [DOI] [PubMed] [Google Scholar]
  • 96.Martin ST, Sato N, Dhara S, Chang R, Hustinx SR, Abe T, Maitra A, Goggins M. Aberrant methylation of the Human Hedgehog interacting protein (HHIP) gene in pancreatic neoplasms. Cancer Biol Ther. 2005;4:728–733. doi: 10.4161/cbt.4.7.1802. [DOI] [PubMed] [Google Scholar]
  • 97.Yamada N, Hamada T, Goto M, Tsutsumida H, Higashi M, Nomoto M, Yonezawa S. MUC2 expression is regulated by histone H3 modification and DNA methylation in pancreatic cancer. Int J Cancer. 2006;119:1850–1857. doi: 10.1002/ijc.22047. [DOI] [PubMed] [Google Scholar]
  • 98.Sato N, Fukushima N, Matsubayashi H, Iacobuzio-Donahue CA, Yeo CJ, Goggins M. Aberrant methylation of Reprimo correlates with genetic instability and predicts poor prognosis in pancreatic ductal adenocarcinoma. Cancer. 2006;107:251–257. doi: 10.1002/cncr.21977. [DOI] [PubMed] [Google Scholar]
  • 99.Sato N, Matsubayashi H, Fukushima N, Goggins M. The chemokine receptor CXCR4 is regulated by DNA methylation in pancreatic cancer. Cancer Biol Ther. 2005;4:70–76. doi: 10.4161/cbt.4.1.1378. [DOI] [PubMed] [Google Scholar]
  • 100.Fukushima N, Sato N, Sahin F, Su GH, Hruban RH, Goggins M. Aberrant methylation of suppressor of cytokine signalling-1 (SOCS-1) gene in pancreatic ductal neoplasms. Br J Cancer. 2003;89:338–343. doi: 10.1038/sj.bjc.6601039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Matsubayashi H, Skinner HG, Iacobuzio-Donahue C, Abe T, Sato N, Riall TS, Yeo CJ, Kern SE, Goggins M. Pancreaticobiliary cancers with deficient methylenetetrahydrofolate reductase genotypes. Clin Gastroenterol Hepatol. 2005;3:752–760. doi: 10.1016/s1542-3565(05)00359-9. [DOI] [PubMed] [Google Scholar]
  • 102.Feinberg AP, Vogelstein B. Hypomethylation distinguishes genes of some human cancers from their normal counterparts. Nature. 1983;301:89–92. doi: 10.1038/301089a0. [DOI] [PubMed] [Google Scholar]
  • 103.Gama-Sosa MA, Slagel VA, Trewyn RW, Oxenhandler R, Kuo KC, Gehrke CW, Ehrlich M. The 5-methylcytosine content of DNA from human tumors. Nucleic Acids Res. 1983;11:6883–6894. doi: 10.1093/nar/11.19.6883. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Hansel DE, Kern SE, Hruban RH. Molecular pathogenesis of pancreatic cancer. Annu Rev Genomics Hum Genet. 2003;4:237–256. doi: 10.1146/annurev.genom.4.070802.110341. [DOI] [PubMed] [Google Scholar]
  • 105.Kimura M, Furukawa T, Abe T, Yatsuoka T, Youssef EM, Yokoyama T, Ouyang H, Ohnishi Y, Sunamura M, Kobari M, Matsuno S, Horii A. Identification of two common regions of allelic loss in chromosome arm 12q in human pancreatic cancer. Cancer Res. 1998;58:2456–2460. [PubMed] [Google Scholar]
  • 106.Gaudet F, Hodgson JG, Eden A, Jackson-Grusby L, Dausman J, Gray JW, Leonhardt H, Jaenisch R. Induction of tumors in mice by genomic hypomethylation. Science. 2003;300:489–492. doi: 10.1126/science.1083558. [DOI] [PubMed] [Google Scholar]
  • 107.Eden A, Gaudet F, Waghmare A, Jaenisch R. Chromosomal instability and tumors promoted by DNA hypomethylation. Science. 2003;300:455. doi: 10.1126/science.1083557. [DOI] [PubMed] [Google Scholar]
  • 108.Eads CA, Nickel AE, Laird PW. Complete genetic suppression of polyp formation and reduction of CpG-island hypermethylation in Apc(Min/+) Dnmt1-hypomorphic Mice. Cancer Res. 2002;62:1296–1269. [PubMed] [Google Scholar]
  • 109.van der Put NM, Gabreels F, Stevens EM, Smeitink JA, Trijbels FJ, Eskes TK, van den Heuvel LP, Blom HJ. A second common mutation in the methylenetetrahydrofolate reductase gene: an additional risk factor for neural-tube defects? Am J Hum Genet. 1998;62:1044–1051. doi: 10.1086/301825. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Blount BC, Mack MM, Wehr CM, MacGregor JT, Hiatt RA, Wang G, Wickramasinghe SN, Everson RB, Ames BN. Folate deficiency causes uracil misincorporation into human DNA and chromosome breakage: implications for cancer and neuronal damage. Proc Natl Acad Sci USA. 1997;94:3290–3295. doi: 10.1073/pnas.94.7.3290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Pogribny IP, Basnakian AG, Miller BJ, Lopatina NG, Poirier LA, James SJ. Breaks in genomic DNA and within the p53 gene are associated with hypomethylation in livers of folate/methyl-deficient rats. Cancer Res. 1995;55:1894–1901. [PubMed] [Google Scholar]
  • 112.Skinner HG, Michaud DS, Giovannucci EL, Rimm EB, Stampfer MJ, Willett WC, Colditz GA, Fuchs CS. A prospective study of folate intake and the risk of pancreatic cancer in men and women. Am J Epidemiol. 2004;160:248–258. doi: 10.1093/aje/kwh214. [DOI] [PubMed] [Google Scholar]
  • 113.Rosty C, Ueki T, Argani P, Jansen M, Yeo CJ, Cameron JL, Hruban RH, Goggins M. Overexpression of S100A4 in pancreatic ductal adenocarcinomas is associated with poor differentiation and DNA hypomethylation. Am J Pathol. 2002;160:45–50. doi: 10.1016/S0002-9440(10)64347-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Sato N, Fukushima N, Matsubayashi H, Goggins M. Identification of maspin and S100P as novel hypomethylation targets in pancreatic cancer using global gene expression profiling. Oncogene. 2004;23:1531–1538. doi: 10.1038/sj.onc.1207269. [DOI] [PubMed] [Google Scholar]
  • 115.Ballestar E, Esteller M. Methyl-CpG-binding proteins in cancer: blaming the DNA methylation messenger. Biochem Cell Biol. 2005;83:374–384. doi: 10.1139/o05-035. [DOI] [PubMed] [Google Scholar]
  • 116.Jones PL, Veenstra GJ, Wade PA, Vermaak D, Kass SU, Landsberger N, Strouboulis J, Wolffe AP. Methylated DNA and MeCP2 recruit histone deacetylase to repress transcription. Nat Genet. 1998;19:187–191. doi: 10.1038/561. [DOI] [PubMed] [Google Scholar]
  • 117.Nan X, Ng HH, Johnson CA, Laherty CD, Turner BM, Eisenman RN, Bird A. Transcriptional repression by the methyl-CpG-binding protein MeCP2 involves a histone deacetylase complex. Nature. 1998;393:386–389. doi: 10.1038/30764. [DOI] [PubMed] [Google Scholar]
  • 118.Esteller M. Epigenetic gene silencing in cancer: the DNA hypermethylome. Hum Mol Genet. 2007;16:R50–59. doi: 10.1093/hmg/ddm018. [DOI] [PubMed] [Google Scholar]
  • 119.Lopez-Serra L, Ballestar E, Fraga MF, Alaminos M, Setien F, Esteller M. A profile of methyl-CpG binding domain protein occupancy of hypermethylated promoter CpG islands of tumor suppressor genes in human cancer. Cancer Res. 2006;66:8342–8346. doi: 10.1158/0008-5472.CAN-06-1932. [DOI] [PubMed] [Google Scholar]
  • 120.Liu C, Chen Y, Yu X, Jin C, Xu J, Long J, Ni Q, Fu D, Jin H, Bai C. Proteomic analysis of differential proteins in pancreatic carcinomas: Effects of MBD1 knock-down by stable RNA interference. BMC Cancer. 2008;8:121. doi: 10.1186/1471-2407-8-121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Kouzarides T. Chromatin modifications and their function. Cell. 2007;128:693–705. doi: 10.1016/j.cell.2007.02.005. [DOI] [PubMed] [Google Scholar]
  • 122.Jenuwein T, Allis CD. Translating the histone code. Science. 2001;293:1074–1080. doi: 10.1126/science.1063127. [DOI] [PubMed] [Google Scholar]
  • 123.Strahl BD, Allis CD. The language of covalent histone modifications. Nature. 2000;403:41–45. doi: 10.1038/47412. [DOI] [PubMed] [Google Scholar]
  • 124.Kouzarides T. Histone methylation in transcriptional control. Curr Opin Genet Dev. 2002;12:198–209. doi: 10.1016/s0959-437x(02)00287-3. [DOI] [PubMed] [Google Scholar]
  • 125.Lachner M, Jenuwein T. The many faces of histone lysine methylation. Curr Opin Cell Biol. 2002;14:286–298. doi: 10.1016/s0955-0674(02)00335-6. [DOI] [PubMed] [Google Scholar]
  • 126.Berger SL. Histone modifications in transcriptional regulation. Curr Opin Genet Dev. 2002;12:142–148. doi: 10.1016/s0959-437x(02)00279-4. [DOI] [PubMed] [Google Scholar]
  • 127.Nishioka K, Reinberg D. Transcription. Switching partners in a regulatory tango. Science. 2001;294:2497–2498. doi: 10.1126/science.1067622. [DOI] [PubMed] [Google Scholar]
  • 128.Cao R, Wang L, Wang H, Xia L, Erdjument-Bromage H, Tempst P, Jones RS, Zhang Y. Role of histone H3 lysine 27 methylation in Polycomb-group silencing. Science. 2002;298:1039–1043. doi: 10.1126/science.1076997. [DOI] [PubMed] [Google Scholar]
  • 129.Rice JC, Briggs SD, Ueberheide B, Barber CM, Shabanowitz J, Hunt DF, Shinkai Y, Allis CD. Histone methyltransferases direct different degrees of methylation to define distinct chromatin domains. Mol Cell. 2003;12:1591–1598. doi: 10.1016/s1097-2765(03)00479-9. [DOI] [PubMed] [Google Scholar]
  • 130.Schotta G, Lachner M, Sarma K, Ebert A, Sengupta R, Reuter G, Reinberg D, Jenuwein T. A silencing pathway to induce H3-K9 and H4-K20 trimethylation at constitutive heterochromatin. Genes Dev. 2004;18:1251–1262. doi: 10.1101/gad.300704. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Heintzman ND, Stuart RK, Hon G, Fu Y, Ching CW, Hawkins RD, Barrera LO, Van Calcar S, Qu C, Ching KA, Wang W, Weng Z, Green RD, Crawford GE, Ren B. Distinct and predictive chromatin signatures of transcriptional promoters and enhancers in the human genome. Nat Genet. 2007;39:311–318. doi: 10.1038/ng1966. [DOI] [PubMed] [Google Scholar]
  • 132.Daniel JA, Pray-Grant MG, Grant PA. Effector proteins for methylated histones: an expanding family. Cell Cycle. 2005;4:919–926. doi: 10.4161/cc.4.7.1824. [DOI] [PubMed] [Google Scholar]
  • 133.Fischle W, Wang Y, Jacobs SA, Kim Y, Allis CD, Khorasanizadeh S. Molecular basis for the discrimination of repressive methyl-lysine marks in histone H3 by Polycomb and HP1 chromodomains. Genes Dev. 2003;17:1870–1881. doi: 10.1101/gad.1110503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Sparmann A, van Lohuizen M. Polycomb silencers control cell fate, development and cancer. Nat Rev Cancer. 2006;6:846–856. doi: 10.1038/nrc1991. [DOI] [PubMed] [Google Scholar]
  • 135.Matarazzo MR, De Bonis ML, Strazzullo M, Cerase A, Ferraro M, Vastarelli P, Ballestar E, Esteller M, Kudo S, D′Esposito M. Multiple binding of methyl-CpG and polycomb proteins in long-term gene silencing events. J Cell Physiol. 2007;210:711–719. doi: 10.1002/jcp.20879. [DOI] [PubMed] [Google Scholar]
  • 136.Vire E, Brenner C, Deplus R, Blanchon L, Fraga M, Didelot C, Morey L, Van Eynde A, Bernard D, Vanderwinden JM, Bollen M, Esteller M, Di Croce L, de Launoit Y, Fuks F. The Polycomb group protein EZH2 directly controls DNA methylation. Nature. 2006;439:871–874. doi: 10.1038/nature04431. [DOI] [PubMed] [Google Scholar]
  • 137.Yamada N, Nishida Y, Tsutsumida H, Hamada T, Goto M, Higashi M, Nomoto M, Yonezawa S. MUC1 expression is regulated by DNA methylation and histone H3 lysine 9 modification in cancer cells. Cancer Res. 2008;68:2708–2716. doi: 10.1158/0008-5472.CAN-07-6844. [DOI] [PubMed] [Google Scholar]
  • 138.Vincent A, Ducourouble MP, Van Seuningen I. Epigenetic regulation of the human mucin gene MUC4 in epithelial cancer cell lines involves both DNA methylation and histone modifications mediated by DNA methyltransferases and histone deacetylases. Faseb J. 2008;22:3035–3045. doi: 10.1096/fj.07-103390. [DOI] [PubMed] [Google Scholar]
  • 139.Lujambio A, Calin GA, Villanueva A, Ropero S, Sanchez-Cespedes M, Blanco D, Montuenga LM, Rossi S, Nicoloso MS, Faller WJ, Gallagher WM, Eccles SA, Croce CM, Esteller M. A microRNA DNA methylation signature for human cancer metastasis. Proc Natl Acad Sci USA. 2008;105:13556–13561. doi: 10.1073/pnas.0803055105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Grady WM, Parkin RK, Mitchell PS, Lee JH, Kim YH, Tsuchiya KD, Washington MK, Paraskeva C, Willson JK, Kaz AM, Kroh EM, Allen A, Fritz BR, Markowitz SD, Tewari M. Epigenetic silencing of the intronic microRNA hsa-miR-342 and its host gene EVL in colorectal cancer. Oncogene. 2008;27:3880–3888. doi: 10.1038/onc.2008.10. [DOI] [PubMed] [Google Scholar]
  • 141.Lehmann U, Hasemeier B, Christgen M, Muller M, Romermann D, Langer F, Kreipe H. Epigenetic inactivation of microRNA gene hsa-mir-9-1 in human breast cancer. J Pathol. 2008;214:17–24. doi: 10.1002/path.2251. [DOI] [PubMed] [Google Scholar]
  • 142.Fabbri M, Garzon R, Cimmino A, Liu Z, Zanesi N, Callegari E, Liu S, Alder H, Costinean S, Fernandez-Cymering C, Volinia S, Guler G, Morrison CD, Chan KK, Marcucci G, Calin GA, Huebner K, Croce CM. MicroRNA-29 family reverts aberrant methylation in lung cancer by targeting DNA methyltransferases 3A and 3B. Proc Natl Acad Sci USA. 2007;104:15805–15810. doi: 10.1073/pnas.0707628104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Matzke MA, Primig M, Trnovsky J, Matzke AJ. Reversible methylation and inactivation of marker genes in sequentially transformed tobacco plants. EMBO J. 1989;8:643–649. doi: 10.1002/j.1460-2075.1989.tb03421.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Morris KV, Chan SW, Jacobsen SE, Looney DJ. Small interfering RNA-induced transcriptional gene silencing in human cells. Science. 2004;305:1289–1292. doi: 10.1126/science.1101372. [DOI] [PubMed] [Google Scholar]
  • 145.Tufarelli C, Stanley JA, Garrick D, Sharpe JA, Ayyub H, Wood WG, Higgs DR. Transcription of antisense RNA leading to gene silencing and methylation as a novel cause of human genetic disease. Nat Genet. 2003;34:157–165. doi: 10.1038/ng1157. [DOI] [PubMed] [Google Scholar]
  • 146.Ting AH, Schuebel KE, Herman JG, Baylin SB. Short double-stranded RNA induces transcriptional gene silencing in human cancer cells in the absence of DNA methylation. Nat Genet. 2005;37:906–910. doi: 10.1038/ng1611. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Yu W, Gius D, Onyango P, Muldoon-Jacobs K, Karp J, Feinberg AP, Cui H. Epigenetic silencing of tumour suppressor gene p15 by its antisense RNA. Nature. 2008;451:202–206. doi: 10.1038/nature06468. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Hong SM, Kelly D, Griffith M, Omura N, Li A, Li CP, Hruban RH, Goggins M. Multiple genes are hypermethylated in intraductal papillary mucinous neoplasms of the pancreas. Mod Pathol. 2008 doi: 10.1038/modpathol.2008. [Epub ahead of print] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Kim SG, Wu TT, Lee JH, Yun YK, Issa JP, Hamilton SR, Rashid A. Comparison of epigenetic and genetic alterations in mucinous cystic neoplasm and serous microcystic adenoma of pancreas. Mod Pathol. 2003;16:1086–1094. doi: 10.1097/01.MP.0000094088.37888.A6. [DOI] [PubMed] [Google Scholar]
  • 150.Ohm JE, McGarvey KM, Yu X, Cheng L, Schuebel KE, Cope L, Mohammad HP, Chen W, Daniel VC, Yu W, Berman DM, Jenuwein T, Pruitt K, Sharkis SJ, Watkins DN, Herman JG, Baylin SB. A stem cell-like chromatin pattern may predispose tumor suppressor genes to DNA hypermethylation and heritable silencing. Nat Genet. 2007;39:237–242. doi: 10.1038/ng1972. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Baylin SB, Ohm JE. Epigenetic gene silencing in cancer - a mechanism for early oncogenic pathway addiction? Nat Rev Cancer. 2006;6:107–116. doi: 10.1038/nrc1799. [DOI] [PubMed] [Google Scholar]
  • 152.McGarvey KM, Van Neste L, Cope L, Ohm JE, Herman JG, Van Criekinge W, Schuebel KE, Baylin SB. Defining a chromatin pattern that characterizes DNA-hypermethylated genes in colon cancer cells. Cancer Res. 2008;68:5753–5759. doi: 10.1158/0008-5472.CAN-08-0700. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Simeone DM. Pancreatic cancer stem cells: implications for the treatment of pancreatic cancer. Clin Cancer Res. 2008;14:5646–5648. doi: 10.1158/1078-0432.CCR-08-0584. [DOI] [PubMed] [Google Scholar]
  • 154.Kern SE, Shibata D. The fuzzy math of solid tumor stem cells: a perspective. Cancer Res. 2007;67:8985–8988. doi: 10.1158/0008-5472.CAN-07-1971. [DOI] [PubMed] [Google Scholar]
  • 155.Lee TI, Jenner RG, Boyer LA, Guenther MG, Levine SS, Kumar RM, Chevalier B, Johnstone SE, Cole MF, Isono K, Koseki H, Fuchikami T, Abe K, Murray HL, Zucker JP, Yuan B, Bell GW, Herbolsheimer E, Hannett NM, Sun K, Odom DT, Otte AP, Volkert TL, Bartel DP, Melton DA, Gifford DK, Jaenisch R, Young RA. Control of developmental regulators by Polycomb in human embryonic stem cells. Cell. 2006;125:301–313. doi: 10.1016/j.cell.2006.02.043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Bibikova M, Chudin E, Wu B, Zhou L, Garcia EW, Liu Y, Shin S, Plaia TW, Auerbach JM, Arking DE, Gonzalez R, Crook J, Davidson B, Schulz TC, Robins A, Khanna A, Sartipy P, Hyllner J, Vanguri P, Savant-Bhonsale S, Smith AK, Chakravarti A, Maitra A, Rao M, Barker DL, Loring JF, Fan JB. Human embryonic stem cells have a unique epigenetic signature. Genome Res. 2006;16:1075–1083. doi: 10.1101/gr.5319906. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Calvanese V, Horrillo A, Hmadcha A, Suarez-Alvarez B, Fernandez AF, Lara E, Casado S, Menendez P, Bueno C, Garcia-Castro J, Rubio R, Lapunzina P, Alaminos M, Borghese L, Terstegge S, Harrison NJ, Moore HD, Brustle O, Lopez-Larrea C, Andrews PW, Soria B, Esteller M, Fraga MF. Cancer genes hypermethylated in human embryonic stem cells. PLoS ONE. 2008;3:e3294. doi: 10.1371/journal.pone.0003294. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Schlesinger Y, Straussman R, Keshet I, Farkash S, Hecht M, Zimmerman J, Eden E, Yakhini Z, Ben-Shushan E, Reubinoff BE, Bergman Y, Simon I, Cedar H. Polycomb-mediated methylation on Lys27 of histone H3 pre-marks genes for de novo methylation in cancer. Nat Genet. 2007;39:232–236. doi: 10.1038/ng1950. [DOI] [PubMed] [Google Scholar]
  • 159.Widschwendter M, Fiegl H, Egle D, Mueller-Holzner E, Spizzo G, Marth C, Weisenberger DJ, Campan M, Young J, Jacobs I, Laird PW. Epigenetic stem cell signature in cancer. Nat Genet. 2007;39:157–158. doi: 10.1038/ng1941. [DOI] [PubMed] [Google Scholar]
  • 160.Kim JY, Siegmund KD, Tavare S, Shibata D. Age-related human small intestine methylation: evidence for stem cell niches. BMC Med. 2005;3:10. doi: 10.1186/1741-7015-3-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Matsubayashi H, Sato N, Fukushima N, Yeo CJ, Walter KM, Brune K, Sahin F, Hruban RH, Goggins M. Methylation of cyclin D2 is observed frequently in pancreatic cancer but is also an age-related phenomenon in gastrointestinal tissues. Clin Cancer Res. 2003;9:1446–1452. [PubMed] [Google Scholar]
  • 162.Herman JG, Umar A, Polyak K, Graff JR, Ahuja N, Issa JP, Markowitz S, Willson JK, Hamilton SR, Kinzler KW, Kane MF, Kolodner RD, Vogelstein B, Kunkel TA, Baylin SB. Incidence and functional consequences of hMLH1 promoter hypermethylation in colorectal carcinoma. Proc Natl Acad Sci USA. 1998;95:6870–6875. doi: 10.1073/pnas.95.12.6870. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Esteller M, Risques RA, Toyota M, Capella G, Moreno V, Peinado MA, Baylin SB, Herman JG. Promoter hypermethylation of the DNA repair gene O(6)-methylguanine-DNA methyltransferase is associated with the presence of G:C to A:T transition mutations in p53 in human colorectal tumorigenesis. Cancer Res. 2001;61:4689–4692. [PubMed] [Google Scholar]
  • 164.Konishi K, Shen L, Wang S, Meltzer SJ, Harpaz N, Issa JP. Rare CpG island methylator phenotype in ulcerative colitis-associated neoplasias. Gastroenterology. 2007;132:1254–1260. doi: 10.1053/j.gastro.2007.01.035. [DOI] [PubMed] [Google Scholar]
  • 165.O′Hagan HM, Mohammad HP, Baylin SB. Double strand breaks can initiate gene silencing and SIRT1-dependent onset of DNA methylation in an exogenous promoter CpG island. PLoS Genet. 2008;4:e1000155. doi: 10.1371/journal.pgen.1000155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Toyota M, Ahuja N, Ohe-Toyota M, Herman JG, Baylin SB, Issa JP. CpG island methylator phenotype in colorectal cancer. Proc Natl Acad Sci USA. 1999;96:8681–8686. doi: 10.1073/pnas.96.15.8681. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Weisenberger DJ, Siegmund KD, Campan M, Young J, Long TI, Faasse MA, Kang GH, Widschwendter M, Weener D, Buchanan D, Koh H, Simms L, Barker M, Leggett B, Levine J, Kim M, French AJ, Thibodeau SN, Jass J, Haile R, Laird PW. CpG island methylator phenotype underlies sporadic microsatellite instability and is tightly associated with BRAF mutation in colorectal cancer. Nat Genet. 2006;38:787–793. doi: 10.1038/ng1834. [DOI] [PubMed] [Google Scholar]
  • 168.Morgan HD, Sutherland HG, Martin DI, Whitelaw E. Epigenetic inheritance at the agouti locus in the mouse. Nat Genet. 1999;23:314–318. doi: 10.1038/15490. [DOI] [PubMed] [Google Scholar]
  • 169.Fukushima N, Walter KM, Uek T, Sato N, Matsubayashi H, Cameron JL, Hruban RH, Canto M, Yeo CJ, Goggins M. Diagnosing pancreatic cancer using methylation specific PCR analysis of pancreatic juice. Cancer Biol Ther. 2003;2:78–83. doi: 10.4161/cbt.183. [DOI] [PubMed] [Google Scholar]
  • 170.Matsubayashi H, Canto M, Sato N, Klein A, Abe T, Yamashita K, Yeo CJ, Kalloo A, Hruban R, Goggins M. DNA methylation alterations in the pancreatic juice of patients with suspected pancreatic disease. Cancer Res. 2006;66:1208–1217. doi: 10.1158/0008-5472.CAN-05-2664. [DOI] [PubMed] [Google Scholar]
  • 171.Yan L, McFaul C, Howes N, Leslie J, Lancaster G, Wong T, Threadgold J, Evans J, Gilmore I, Smart H, Lombard M, Neoptolemos J, Greenhalf W. Molecular analysis to detect pancreatic ductal adenocarcinoma in high-risk groups. Gastroenterology. 2005;128:2124–2130. doi: 10.1053/j.gastro.2005.03.006. [DOI] [PubMed] [Google Scholar]
  • 172.Klump B, Hsieh CJ, Nehls O, Dette S, Holzmann K, Kiesslich R, Jung M, Sinn U, Ortner M, Porschen R, Gregor M. Methylation status of p14ARF and p16INK4a as detected in pancreatic secretions. Br J Cancer. 2003;88:217–222. doi: 10.1038/sj.bjc.6600734. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Jiang P, Watanabe H, Okada G, Ohtsubo K, Mouri H, Tsuchiyama T, Yao F, Sawabu N. Diagnostic utility of aberrant methylation of tissue factor pathway inhibitor 2 in pure pancreatic juice for pancreatic carcinoma. Cancer Sci. 2006;97:1267–1273. doi: 10.1111/j.1349-7006.2006.00308.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Park JK, Ryu JK, Lee KH, Lee JK, Yoon WJ, Lee SH, Yoo JW, Woo SM, Lee GY, Lee CH, Kim YT, Yoon YB. Quantitative analysis of NPTX2 hypermethylation is a promising molecular diagnostic marker for pancreatic cancer. Pancreas. 2007;35:e9–15. doi: 10.1097/MPA.0b013e318153fa42. [DOI] [PubMed] [Google Scholar]
  • 175.Ohtsubo K, Watanabe H, Yao F, Okada G, Mouri H, Yamaguchi Y, Sawabu N. Preproenkephalin hypermethylation in the pure pancreatic juice compared with p53 mutation in the diagnosis of pancreatic carcinoma. J Gastroenterol. 2006;41:791–797. doi: 10.1007/s00535-006-1857-3. [DOI] [PubMed] [Google Scholar]
  • 176.Parsi M, Li A, Li CP, Goggins M. DNA methylation alterations in ERCP brush samples of patients with suspected pancreaticobiliary disease. Clin Gastro Hepatol. 2008 doi: 10.1016/j.cgh.2008.07.007. (in press) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Tan SH, Ida H, Lau QC, Goh BC, Chieng WS, Loh M, Ito Y. Detection of promoter hypermethylation in serum samples of cancer patients by methylation-specific polymerase chain reaction for tumour suppressor genes including RUNX3. Oncol Rep. 2007;18:1225–1230. [PubMed] [Google Scholar]
  • 178.Jiao L, Zhu J, Hassan MM, Evans DB, Abbruzzese JL, Li D. K-ras mutation and p16 and preproenkephalin promoter hypermethylation in plasma DNA of pancreatic cancer patients: in relation to cigarette smoking. Pancreas. 2007;34:55–62. doi: 10.1097/01.mpa.0000246665.68869.d4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Fleischhacker M, Schmidt B. Circulating nucleic acids (CNAs) and cancer–a survey. Biochim Biophys Acta. 2007;1775:181–232. doi: 10.1016/j.bbcan.2006.10.001. [DOI] [PubMed] [Google Scholar]
  • 180.Goessl C, Muller M, Miller K. Methylation-specific PCR (MSP) for detection of tumour DNA in the blood plasma and serum of patients with prostate cancer. Prostate Cancer Prostatic Dis. 2000;3:S17. doi: 10.1038/sj.pcan.4500441. [DOI] [PubMed] [Google Scholar]
  • 181.Fujiwara K, Fujimoto N, Tabata M, Nishii K, Matsuo K, Hotta K, Kozuki T, Aoe M, Kiura K, Ueoka H, Tanimoto M. Identification of epigenetic aberrant promoter methylation in serum DNA is useful for early detection of lung cancer. Clin Cancer Res. 2005;11:1219–1225. [PubMed] [Google Scholar]
  • 182.Bastian PJ, Palapattu GS, Lin X, Yegnasubramanian S, Mangold LA, Trock B, Eisenberger MA, Partin AW, Nelson WG. Preoperative serum DNA GSTP1 CpG island hypermethylation and the risk of early prostate-specific antigen recurrence following radical prostatectomy. Clin Cancer Res. 2005;11:4037–4043. doi: 10.1158/1078-0432.CCR-04-2446. [DOI] [PubMed] [Google Scholar]
  • 183.Sorenson GD. Detection of mutated KRAS2 sequences as tumor markers in plasma/serum of patients with gastrointestinal cancer. Clin Cancer Res. 2000;6:2129–2137. [PubMed] [Google Scholar]
  • 184.Tsang JC, Lo YM. Circulating nucleic acids in plasma/serum. Pathology. 2007;39:197–207. doi: 10.1080/00313020701230831. [DOI] [PubMed] [Google Scholar]
  • 185.Diehl F, Li M, He Y, Kinzler KW, Vogelstein B, Dressman D. BEAMing: single-molecule PCR on microparticles in water-in-oil emulsions. Nat Methods. 2006;3:551–559. doi: 10.1038/nmeth898. [DOI] [PubMed] [Google Scholar]
  • 186.Diehl F, Li M, Dressman D, He Y, Shen D, Szabo S, Diaz LA, Jr, Goodman SN, David KA, Juhl H, Kinzler KW, Vogelstein B. Detection and quantification of mutations in the plasma of patients with colorectal tumors. Proc Natl Acad Sci USA. 2005;102:16368–16373. doi: 10.1073/pnas.0507904102. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from International Journal of Clinical and Experimental Pathology are provided here courtesy of e-Century Publishing Corporation

RESOURCES