Review Open Access
Copyright ©2006 Baishideng Publishing Group Co., Limited. All rights reserved.
World J Gastroenterol. Nov 21, 2006; 12(43): 6933-6940
Published online Nov 21, 2006. doi: 10.3748/wjg.v12.i43.6933
Methylation in esophageal carcinogenesis
Da-Long Wu, Feng-Ying Sui, Xiao-Ming Jiang, Xiao-Hong Jiang, Department of Pharmacology, School of Medicine, College of Jiaxing, Jiaxing 314001, Zhejiang Province, China
Author contributions: All authors contributed equally to the work.
Correspondence to: Da-Long Wu, Department of Pharmacology, School of Medicine, College of Jiaxing, Jiaxing 314001, Zhejiang Province, China. wudalong66@sina.com
Telephone: +86-573-3643836 Fax: +86-573-3643497
Received: July 4, 2006
Revised: July 12, 2006
Accepted: July 18, 2006
Published online: November 21, 2006

Abstract

Genetic abnormalities of proto-oncogenes and tumor suppressor genes have been demonstrated to be changes that are frequently involved in esophageal cancer pathogenesis. However, hypermethylation of CpG islands, an epigenetic event, is coming more and more into focus in carcinogenesis of the esophagus. Recent studies have proved that promoter hypermethylation of tumor suppressor genes is frequently observed in esophageal carcinomas and seems to play an important role in the pathogenesis of this tumor type. In this review, we will discuss current research on genes that are hypermethylated in human esophageal cancer and precancerous lesions of the esophagus. We will also discuss the potential use of hypermethylated genes as targets for detection, prognosis and treatment of esophageal cancer.

Key Words: Methylation, Esophageal cancer, Tumor suppressor gene, Carcinogenesis



INTRODUCTION

Esophageal cancer is one of the least studied and deadliest cancers, with a remarkable geographical distribution and a low likelihood of cure[1]. Therefore, the current challenges in the management of esophageal cancer are to obtain a better understanding of the underlying molecular biological alterations to provide new treatment options. Cancer of the esophagus exists in two main forms with different etiological and pathological characteristics-esophageal squamous cell carcinoma (ESCC) and esophageal adenocarcinoma (EAC)[1]. It is well known that esophageal carcinogenesis is a multistage and progressive process which includes basal cell hyperplasia (BCH), dysplasia (DYS), carcinoma in site (CIS) and advanced esophageal carcinoma[1,2]. A variety of genetic lesions are involved in esophageal carcinogenesis, including gene amplifications, loss of heterozygosity(LOH) or homozygous deletions, mutations, and chromosomal rearrangements[1,2]. From the above mentioned genetic lesions , mutations are greatly focused on. The vast majority of esophageal cancers have mutations of the TP53 and p16 genes at an early stage followed by mutations in genes such as APC, Rb and cyclin D1 at later stages of progression[3]. Furthermore, the alteration of the gene mutations may show different types in ESCCs and EACs. For example, ESCCs and EACs show distinct patterns of TP53 mutations, namely a high prevalence of G > A transitions at CpG sites in EACs whereas in ESCCs a higher prevalence of G to T transversions and mutations at A:T base pairs is present[4].

Genetic mutation of genes that inhibit the formation of tumors has long been known to be one of the main driving forces in the development of cancer[1]. However, recent data have focused our attention to the contribution of epigenetics to tumorigenesis. In tumorigenesis of the esophagus, the epigenetic inactivation of genes is as an important a driving force as the inactivation of genes by mutation[5]. ‘Epigenetic’ events, i.e. heritable changes in gene function which cannot be explained by changes in DNA sequence, are composed of histone acetylation, the chromatin structure and DNA methylation[5]. DNA methylation seems to be the most important mechanism for “epigenetic change” at present[5,6]. Through a process of post-replicative covalent modification catalysed by DNA methyltransferases(DNMTs), the DNA of mammalian cells contains a ‘fifth base’, namely 5-methylcytosine. The most frequent target for this modification is cytosine in the context of the dinucleotide CpG[5]. Throughout the genome CpG dinucleotides are found at one-fifth of their predicted frequency[6]. In marked contrast to the genome-wide underrepresentation of CpGs, there are regions of the genome termed CpG islands which have maintained their expected frequency of the dinucleotides. And the CpG islands are often found within the promoter of genes[6,7]. It has been known for many years that, in general terms, there is an inverse relationship between the density of promoter methylation and the transcriptional activity of a gene[8,9]. The mechanism of gene silencing by promoter hypermethylation has recently been shown to be related to the recruitment of repressor protein complex, resulting in de-acetylation of the chromatin and histone, thus barring access to the active transcription complex. However, the actual mechanisms by which DNA methylation modulates gene expression have remained elusive[7,10]. The assays for detection of cytosine methylation can be divided into two groups: restriction enzyme-based and bisulfite treatment-based[11,12]. The former employs the inhibition of certain restriction enzymes by methylation of their recognition sites as an indicator for the presence of methylation. The latter translates the epigenetic information of cytosine methylation in primary sequence differences by converting unmethylated cytosine to uracil whereas methylated cytosine remains unaltered. The bisulfite-converted genomic DNA can be analyzed by a wide variety of PCR-based methods[11,12]. Methylation is needed for the normal development of cells. And genome stability and normal gene expression are largely maintained by a fixed and predetermined pattern of DNA methylation[13]. Aberrant methylation confers a selective growth advantage that results in cancerous growth[13]. From various lines of evidence, it is known that the methylation pattern of the cancerous cell is associated with a broad genomic hypomethylated state that is often accompanied by a more regional and locus-specific hypermethylated pattern[7]. The presence of alterations in the profile of DNA methylation in cancer was initially thought to be exclusively a global hypomethylation of the genome that would possibly lead to massive overexpression of oncogenes whose CpG islands were normally hypermethylated[14]. Nowadays, however, this is considered to be an unlikely or, at least, incomplete scenario. The popularity of the concept of demethylation of oncogenes leading to their activation is in clear decadency[14,15]. Hypermethylation of CpG islands located in the promoter regions of tumor suppressor genes(TSGs) is now firmly established as an important mechanism for gene inactivation[16,17,18]. The particular genes that are hypermethylated in tumor cells are strongly specific to the tissue of origin of the tumor. A profile of CpG island hypermethylation exists according to the tumor type[15]. The mechanism responsible for this type of pattern remains largely unclear. Moreover, accumulating evidence indicates that CpG island hypermethylation is an early event in cancer development and, in some cases, may precede the neoplastic process[19]. Therefore, such profiles would provide invaluable insight into mechanisms underlying the evolution of each tumor type and will provide new molecular markers. This review will focus on the current understanding of DNA methylation abnormalities in esophageal cancer and discuss how this knowledge contributes to our understanding of the pathogenesis of esophageal cancer.

STUDIES OF GENE PROMOTER HYPER-METHYLATION IN ESOPHAGEAL CANCER

Putative tumor suppressor genes, involving apoptosis, cell adherence, DNA repair, and the cell cycle, have been investigated for hypermethylation by various techniques in esophageal cancer. Below, we have chosen to describe those genes that have been most extensively studied in the past and that have been shown to undergo epigenetic changes in esophageal carcinoma.

p14ARF, p15 and p16

The 9p21 chromosomal band is one of the most frequently altered genomic regions in human cancers[20]. Within a short distance of 50 kb, a gene cluster consisting of three genes, p14ARF, p15 and p16, is harbored. All of which have putative tumor suppressor roles[20,21,22]. Inactivation of p14ARF, p15 and p16 genes has been observed in many types of human cancers including ESCC[21,22,23]. For example, the results from immunohistochemical analysis indicated that p16 expression was present in only 3 out of 22 ESCC cases[24]. Some studies showed that germline mutations in the p16 gene might be related to familial melanoma[25], but another study found the mutation of the p16 gene in esophageal cancer was rare[26]. Hemizygous and homozygous deletion at 9p21 are widely considered to be one of the primary mechanisms of p16/p15 inactivation[26]. Recently, however, aberrant methylation of the CpG islands at the promoter regions of p16 and p15 genes was reported in many cancers and was associated with loss of transcription[8,27]. Abbaszadegan et al[28] assessed a large family with clustering of ESCC in northeastern Iran and found aberrant p16 promoter methylation in 64% of ESCC family members and none in normal volunteers. By analyzing the p14ARF, p15, and p16 genes individually in 40 ESCCs, Xing et al[29] detected aberrant promoter methylation of the p16 gene in 40%, of p14ARF in 15%, and of p15 in 13% tumor samples. They further detected homozygous deletion of p16 in 18%, of p14ARF in 33%, and of p15 in 40% tumor samples, and detected no mutation in the p14ARF and p16 genes[29]. Hardie et al[30] reported that hypermethylation of the p16 promoter is detected in 85% (18/21) of EACs while p16 mutations are uncommon (1.9%; 1/54). Another report found that in 50 Barrett’s esophagus-associated EACs, hypermethylation of p16 was present frequently (54%), but hypermethylation of p14ARF was absent[27]. The above results suggest that p14ARF, together with p15, is a primary target of homozygous deletion, whereas p16 is the hypermethylation hotspot in human esophageal cancer.

The FHIT gene

The FHIT gene is located at chromosome 3p14.2 and encodes a polypeptide of 147 amino acids[31]. FHIT allelic deletions and reduced or absent FHIT protein expression have been observed in a variety of tumors suggesting a putative tumor suppressor function[31,32]. In ESCCs, the CpG island in the FHIT promoter region was hypermethylated in 25 of the 36 (69%) analyzed cases, significantly correlated with the deletion of FHIT protein expression[33]. Methylated ESCC cell lines exhibit re-expression of the FHIT gene and demethylation in the CpG islands after treatment with demethylating agent 5-aza-2’-deoxycytidine[34]. Another report showed that aberrant methylation of FHIT was found in 85 of 257 (33%) ESCCs[35]. These findings suggest that methylation of the 5’ CpG islands of the FHIT gene is closely associated with transcriptional inactivation and might be involved in tumor development of the esophagus.

The RARβ2 gene

The retinoic acid receptor-beta2 (RARβ2) gene located at 3p24 has been intensively studied in many cancers and found to have defective function, thus making it a candidate TSG[36]. RARβ2 expression was detected in 88% (14/16) of normal esophageal tissues and only 54% (84/162) of esophageal carcinomas[36]. And 14 of 20 (70%) ESCC samples had hypermethylation of the RARβ2 promoter[37]. Another group reported that 34 of 47 (73%) primary resected ESCC samples showed RARβ2 methylation[38]. After 5-aza-2’-deoxycytidine treatment the expression of RARβ2 was reversed in two RARβ2-downregulated ESCC cell lines[39]. These results identified methylation as the underlying mechanism for this frequent loss of RARβ2 in esophageal cancer.

The APC gene

The adenomatous polyposis coli (APC) gene, located on chromosome 5q21, is a TSG in the wnt signaling pathway[40]. APC shows frequent LOH in esophageal carcinomas, and the prevalence of mutations in the APC gene in esophageal carcinomas is low[41]. Hypermethylation of the promoter region of the APC gene occurred in abnormal esophageal tissue in 48 of 52 (92%) patients with EAC, in 16 of 32 (50%) patients with ESCC, but not in matching normal esophageal tissues[41]. So methylation of the promoter region of this gene constitutes an alternative mechanism of gene inactivation in esophageal carcinoma.

The ER gene

The estrogen receptor (ER) gene at chromosome 6q, which has growth and metastasis suppressor activity in many different cell types, is widely expressed in tissues other than breast, and is methylated in 51% of EAC patients[42].

The MGMT gene

The human enzyme O6-methylguanine-DNA methyltransferase (MGMT) is located on chromosome band 10q26, and protects the cell from guanine methylation by irreversibly transferring the alkyl group of the O6-methylguanine to a specific cysteine residue within the molecule[43]. Approximately 20% of tumor cell lines lack MGMT activity and are highly sensitive to alkylating agents[44]. In established cancer cell lines, MGMT expression appears to be correlated with methylation in the promoter of the gene[44]. The gene has been shown to be methylated in 46/119 (39%) ESCC, but all 21 normal esophageal tissues had unmethylated MGMT[45]. Another report studied the role of DNA hypermethylation in the loss of expression of MGMT during the development of ESCC, and found that 5 of 17 (29%) normal esophagus, 10 of 20 (50%) BCH, 8 of 12 (67%) DYS, and 13 of 18 (72%) ESCC samples had DNA hypermethylation in the MGMT promoter region, showing a gradual increase with the progression of carcinogenesis, while the frequency of the loss of MGMT mRNA and protein expression progressively decreased from normal to BCH, DYS, and ESCC, and it was highly correlated with MGMT promoter hypermethylation[46].

The E-cadherin gene

E-cadherin gene on chromosome 16q22.1 encodes a Mr 120 000 transmembrane glycoprotein expressed on the surface of epithelial cells. In epithelial tissues, E-cadherin mediates homophilic, Ca2+-dependent intercellular adhesion that is essential for the maintenance of normal tissue architecture[47]. Loss of E-cadherin expression occurs in a variety of human tumors and is correlated with invasion and metastasis, and activation of E-cadherin results in the growth inhibition of tumor cell lines[48]. E-cadherin can be targeted by both genetic and epigenetic means. Moreover, the hypermethylation of E-cadherin was seen frequently in most tumor types, but mutations only frequently in a small number of specific subtypes[48]. In esophageal carcinoma, downregulation of E-cadherin is common and is associated with an increase in invasive and metastatic potential, but mutations of the gene are rare[49]. E-cadherin was methylated in 26 of 31 (84%) EAC specimens, 16 of 20 (80%) ESCC samples and 4 of 6 ESCC cell lines[49,50,51]. And treatment of E-cadherin-negative carcinoma cells with the demethylating agent, 5-aza-2’-deoxycytidine, induced re-expression of the gene[51]. These data suggest that epigenetic silencing via aberrant methylation of the E-cadherin promoter is the critical mechanism for inactivation of this gene in esophageal cancer.

The TSLC1 gene

The TSLC1 (tumor suppressor in lung cancer) gene located on 11q23.2 was first characterized as a TSG in human non-small cell lung cancer(NSCLC) and termed TSLC1[52]. The tumor suppressor role of this gene has been demonstrated in the cell lines of NSCLC, hepatocellular carcinoma, pancreatic cancer and ESCC[52,53]. Loss of TSLC1 expression was observed in 75% of the ESCC cell lines and 50% of the primary tumors from ESCC patients[53]. Ito et al[53] examined the methylation status of six cytosine residues of CpG sites in a putative promoter sequence upstream from the TSLC1 translation initiation site by bisulfite sequencing in four cell lines, including KYSE270, which expressed TSLC1, and KYSE410, KYSE520, and KYSE960, which did not express it. They also found all of the cytosine residues in KYSE270 DNA were unmethylated, whereas all of the six cytosine residues in KYSE520 DNA and five residues in KYSE410 and KYSE960 DNA were methylated. Especially, the cytosine residues in KYSE520 DNA were all hypermethylated. However, the report about the status of the promoter methylation of TSLC1 gene in esophageal cancer tissue has not been available.

The RASSF1A gene

Many known RAS effectors are oncoproteins on their own. Less is known about Ras effectors possessing tumor suppressor properties[54]. Recently, a new family of genes encoding a putative Ras effector, the Ras-association domain family 1 (RASSF1) gene, has been identified within the critical lung and breast cancer deletion region at 3p21.3. The RASSF1 locus encodes several major transcripts by alternative promoter selection and alternative mRNA splicing: RASSF1A, RASSF1B and RASSF1C. Many studies have suggested that RASSF1A was a new putative TSG[54,55]. RASSF1A acts as a negative effector of Ras in a pro-apoptotic signaling pathway. Interestingly, mutational inactivation of this gene is very rare (< 2%), and the main mechanism of its inactivation is through promoter methylation and LOH[56]. The RASSF1A isoform is highly epigenetically inactivated in lung, breast, ovarian, kidney, prostate, thyroid, esophagus and several other carcinomas[54]. Hypermethylation of RASSF1A was detected in 73% of ESCC cell lines and 51% of primary ESCCs, whereas only 4.3% of RASSF1A hypermethylation were detected in corresponding noncancerous tissues[38]. There was a statistically significant correlation between the presence of hypermethylation and tumor stage[57]. Wong et al[58] also found that RASSF1A was partially methylated in 3/7 (43%) esophageal cancer cell lines; 22/64 (34%) ESCCs and 3/64 (4.7%) corresponding non-tumor samples; and was not methylated in 2 immortalized normal oesophageal epithelial cell lines and 6 normal esophageal epithelium samples. These findings suggest that epigenetic silencing of RASSF1A gene expression by promoter hypermethylation could play an important role in ESCC carcinogenesis.

Besides the above mentioned genes, there are hypermethylations of some other genes involving esophageal cancer, including hMLH1[59,60], VHL[38], TIMP3[42,61], DAP-kinase[42], pRb[62], ECRG4[63], Chfr[64], HLA class I[65], EYA4[66], CDH13[67], SFRP1[68] and PGP9.5[69]. Table 1 gives a summary of the profile of gene hypermethylation in human esophageal cancer.

Table 1 Compilation of genes hypermethylated in esophageal cancer n (%).
GeneEntity of pathologyIncidence of methylation
Reference
Cancer tissue(Adjacent)nonmalignant tissue
p14ARFESCC6/40 (15)ND29
EAC0/50 (0.0)ND27
p15ESCC6/34 (18)ND26
ESCC5/40 (13)ND29
p16ESCC17/34 (50)ND26
ESCC18/28 (64)0/30 (0.0)28
ESCC16/40 (40)ND29
EAC27/50 (54)ND27
EAC8/21 (38)ND27
EAC16/41 (39)10/41 (24)42
FHITESCC25/36 (69)ND33
ESCC85/257 (33)ND35
RARβ2ESCC14/20 (70)2/17 (12)37
ESCC34/47 (73)18/47 (38)38
APCEAC48/52 (92)0/52 (0.0)41
ESCC16/32 (50)0/32 (0.0)41
EAC28/41 (68)3/41 (7.3)42
EAC39/50 (78)ND27
EREAC21/41 (51)5/41 (12)42
MGMTESCC46/119 (39)0/21 (0.0)45
EAC24/41 (56)10/41 (24)42
E-CadherinEAC26/31 (84)ND49
EAC27/41 (66)5/41 (12)42
ESCC16/20 (80)ND50
TSLC1ESCC28/56 (50)ND53
RASSF1AESCC24/47 (51)2/47 (4.3)38
ESCC22/64 (34)3/64 (4.7)58
hMLH1ESCC0/30 (0.0)ND59
ESCC + EAC79/124 (64)ND60
VHLESCC6/47 (13)0/47 (0)39
TIMP3EAC8/41 (19)0/41 (0.0)42
EAC71/79 (90)0/79 (0.0)61
DAP-kinaseEAC8/41 (19)2/41 (4.9)42
pRbEAC10/30 (33)ND62
ECRG4ESCC12/15 (80 )3/20 (15)63
ChfrESCC + EAC7/43 (16)ND64
HLA class IESCC13/29 (45)ND65
EYA4EAC33/40 (83)2/58 (3.4)66
CDH13ESCC+EAC5/37 (14)ND67
SFRP1EAC37/40 (93)3/30 (10)68
PGP9.5ESCC21/50 (42)ND69

From the above mentioned reports, we find that there is obvious different methylation frequency in a gene from different authors in some cases, which may be due to different assay methods and different specimen resources. And the geographical difference may be due to variable carcinogens in the different areas. Furthermore, there seems to be obvious difference of methylation frequency of some genes between ESCCs and EACs. For example, hypermethylation of the promoter region of the APC gene occurred in 48 of 52 (92%) patients with EAC, but in 16 of 32 (50%) patients with ESCC[41], which suggests that hypermethylation of the APC gene has distinct roles in ESCC and EAC.

METHYLATION IN SERUM DNA FROM ESOPHAGEAL CANCER PATIENTS

Despite advances in diagnosis and treatment of various cancers, early detection and treatment of cancer remain a challenge. One potential early detection biomarker is DNA methylation of the promoter region of certain cancer-associated genes[70]. Genetic analysis has shown that cell-free circulating DNA in plasma or serum of cancer patients shares similar genetic alterations to those described in the corresponding tumor[70,71]. Numerous studies have demonstrated the presence of promoter hypermethylation of tumor suppressor genes in the serum DNA of patients with various cancers[72]. Hypermethylated APC DNA was observed in the plasma of 13 of 52 (25%) EACs and 2 of 32 (6.3%) ESCCs[41]. Hibi et al[73] found that aberrant promoter methylation of the p16 gene was detected in 31 of 38 (82%) ESCCs, and 7 of these 31 (23%) patients with a p16 alteration in the primary tumor had the same methylation changes in the corresponding serum DNA. This study yielded a promising result: a tumor associated DNA alteration could be detected in the serum of 18% of ESCC patients (7 of 38 patients) using p16 methylation as a target. Moreover, the clinical sensitivity of this assay can be potentially improved by incorporating other possibly methylated target genes, which have been estimated in other tumor types. For example, Esteller et al[74] analyzed primary NSCLCs and serum from 22 patients for the methylation pattern of four TSGs (DAPK, GSTP1, p16, and MGMT). Methylation of at least one of these genes was detected in 68% of NSCLCs. Comparing primary tumors with methylation and matched serum samples, 73% of the matched serum samples were found to be methylated[74]. In addition, none of the sera from patients with tumors not demonstrating methylation were positive[74]. Therefore, combined detection of aberrant promoter hypermethylation of cancer-related genes in serum may be useful for esophageal cancer diagnosis or the detection of recurrence.

PROGNOSTIC SIGNIFICANCE OF GENE HYPERMETHYLATION

In the past few years, numerous attempts have been made to establish a genetic technique for reliably predicting tumor prognosis, but these attempts have been hindered by two main problems. First, only a few genes are somatically mutated in solid tumors and, second, because cell populations of primary neoplasms are heterogeneous, no single marker can accurately predict the behavior of the tumor[13]. Fortunately, emerging evidence suggests a possible prognostic value of gene promoter hypermethylation[75]. Lee et al[35] reported that aberrant methylation of the FHIT promoter in ESCC was found to be significantly associated with a poor prognosis for stage 1-2 cases. Mandelker et al[69] reported that PGP9.5 methylation was an independent prognostic factor for ESCC survival (P = 0.03) . Kawakami et al[41] reported that high plasma levels of methylated APC DNA were statistically significantly associated with reduced EAC patient survival. Brock et al[42] analyzed the methylation status of seven genes (including APC, E-cadherin, MGMT, ER, p16, DAP-kinase and TIMP3) of 41 EAC samples and found that DNA methylation of some genes individually showed only trends toward diminished survival, whereas patients whose tumors had > 50% of their gene profile methylated had both significantly poorer survival and earlier tumor recurrence than those without positive methylation. The data suggest that combined detection of methylation status for multiple genes is an effective strategy for prediction of esophageal tumor behavior. Although some genes that are frequently inactivated by methylation and are of prognostic impact for esophageal cancer patients have already been found, additional genes need to be identified. Thus, patients with a worse prognosis could be selected. These patients might benefit from a more aggressive treatment strategy.

ABERRANT DNA METHYLATION IS AN EARLY EVENT IN ESOPHAGEAL CARCINOGENESIS

In many tumors, it has been proved that aberrant DNA methylation frequently occurs in precancerous tissue as well as cancer tissue, and both factors, genetic and epigenetic, lie at the origin of carcinogenesis[76]. The relative contribution of each varies significantly in different human tumors[76]. Nie et al[77] compared hypermethylation of p16, p15, p14, HLA-A, -B, -C, hMLH1, E-cadherin, FHIT and VHL genes in precancerous esophageal tissues and found that in 48 biopsy samples with BCH or DYS, the most frequent hypermethylated genes were p16 (19%) and p14ARF (15%), and seventeen out of these 48 samples (35%) contained hypermethylation of at least one gene. In the resected tissues, 52% of the BCH and 81% of the tumors showed hypermethylation of at least one gene. Another study reported that 2 of 17 (12%) normal esophagus, 9 of 21 (43%)BCH, 7 of 12 (58%) DYS, and 14 of 20 (70%) ESCC samples had hypermethylation of the RARβ2 promoter region[37]. As to progression of EAC, it has been reported that, methylation of the p16 promoter was detected in 18 of 22 (82%) EAC and 10 of 33 (30%) premalignant lesions, whereas no methylation of the p16 promoter was found in normal esophageal epithelia[78]. Hardie et al[30] reported hypermethylation of the p16 promoter was detected in 77% (14/18) of Barrett’s epithelia, and in 85% (18/21) of EACs. These data suggest that aberrant DNA methylation participates early in the development of esophageal cancer. Recently, the lab of professor Yang CS reported that EGCG, the major polyphenol from green tea, inhibited DNMT activity and reactivated several methylation-silenced genes, including p16, RARβ2, MGMT and hMLH1, in human esophageal cancer KYSE 510 cells, accompanied by the expression of mRNA of these genes[79]. The result suggests that methylation might be a new target of chemopreventive activity. In the last two decades, it has been proven that many drugs, such as tamoxifen, aspirin, COX-2 inhibitors, possess positive chemopreventive activity against esophageal cancer[2]. However, the exact mechanisms have not been elucidated so far. Therefore, it will be very attractive to examine the effect of these drugs on promoter methylation status of key genes in esophageal cancer cells, esophageal cancer tissue, and especially precancerous tissue of the esophagus.

HYPERMETHYLATION AS A TARGET OF THERAPEUTIC INTERVENTION

It has been reported that demethylating agents 5-azacytidine and 5-aza-2’-deoxycytidine can restore the normal demethylated state of several types of TSGs and increase their expression in various cancers, including esophageal cancer, in vitro and in vivo[13,14,15,39,54]. Since methylation and transcriptional status are inversely correlated, the use of demethylating agents appears to be a promising option for the treatment of tumors. Methylation of genes in tumor cells could provide a tumor-specific target for new therapies[80,81,82]. In fact, these demethylating agents have exhibited significant activity in the treatment of patients with myelodysplastic syndrome, chronic myeloid leukaemia and acute myeloid leukaemia[83,84]. However, preliminary experience with these agents in solid tumors has been relatively low[85]. Esophageal tumor shows a high prevalence of TSG hypermethylation, and the above studies demonstrated that gene expression could be restored after treatment of esophageal tumor cells with demethylating agents in vitro. However, up to date the clinical trial about demethylating agents in esophageal cancer is unavailable. Although it is too early to make any expectation about the effect of these drugs on esophageal cancer, this is a very promising concept and needs to be tested in clinical trials.

CONCLUSION AND PERSPECTIVES

Esophageal carcinogenesis is a stepwise process of the accumulation of genetic and epigenetic abnormalities. It has become clear that promoter hypermethylation of TSGs is as important for this multistep process as genetic changes in the progression of esophageal carcinogenesis. The steadily growing list of genes inactivated by promoter hypermethylation in esophageal carcinoma provides not only new insights into the molecular basis of the diseases but also a long list of interesting candidate genes for the development of molecular markers which might contribute to the improvement of diagnosis and also prognosis. In addition, the fact that methylation can be reversed in vitro and the effect of the demethylating agent 5-aza-2’-deoxycytidine in vitro raise hope for new treatment strategies for esophageal cancer patients. Furthermore, understanding of the significance of aberrant DNA methylation in the precancerous stage may show that a new strategy, correction of aberrant DNA methylation, can prevent esophageal cancer in people with premalignant lesions, such as Barrett’s esophagus, BCH and DYS.

ACKNOWLEDGMENTS

We acknowledge the help given by Li-Dong Wang, Professor of Pathology and Oncology, College of Medicine, Zhengzhou University, China.

Footnotes

S- Editor Liu Y L- Editor Schreyer AG E- Editor Ma WH

References
1.  Enzinger PC, Mayer RJ. Esophageal cancer. N Engl J Med. 2003;349:2241-2252.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2115]  [Cited by in F6Publishing: 2142]  [Article Influence: 102.0]  [Reference Citation Analysis (0)]
2.  Chen X, Yang CS. Esophageal adenocarcinoma: a review and perspectives on the mechanism of carcinogenesis and chemoprevention. Carcinogenesis. 2001;22:1119-1129.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 125]  [Cited by in F6Publishing: 128]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
3.  Raja S, Godfrey TE, Luketich JD. The role of tumor suppressor genes in esophageal cancer. Minerva Chir. 2002;57:767-780.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Montesano R, Hainaut P. Molecular precursor lesions in oesophageal cancer. Cancer Surv. 1998;32:53-68.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Momparler RL. Cancer epigenetics. Oncogene. 2003;22:6479-6483.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 209]  [Cited by in F6Publishing: 200]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
6.  Farrell WE, Clayton RN. Epigenetic change in pituitary tumorigenesis. Endocr Relat Cancer. 2003;10:323-330.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 35]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
7.  Chim CS, Liang R, Kwong YL. Hypermethylation of gene promoters in hematological neoplasia. Hematol Oncol. 2002;20:167-176.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 82]  [Cited by in F6Publishing: 87]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
8.  Garinis GA, Patrinos GP, Spanakis NE, Menounos PG. DNA hypermethylation: when tumour suppressor genes go silent. Hum Genet. 2002;111:115-127.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 83]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
9.  Herman JG, Baylin SB. Gene silencing in cancer in association with promoter hypermethylation. N Engl J Med. 2003;349:2042-2054.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2489]  [Cited by in F6Publishing: 2369]  [Article Influence: 112.8]  [Reference Citation Analysis (0)]
10.  Jain PK. Epigenetics: the role of methylation in the mechanism of action of tumor suppressor genes. Ann N Y Acad Sci. 2003;983:71-83.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 30]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
11.  Lehmann U, Brakensiek K, Kreipe H. Role of epigenetic changes in hematological malignancies. Ann Hematol. 2004;83:137-152.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 28]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
12.  Fraga MF, Esteller M. DNA methylation: a profile of methods and applications. Biotechniques. 2002;33:632, 634, 636-649.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Esteller M. Relevance of DNA methylation in the management of cancer. Lancet Oncol. 2003;4:351-358.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 239]  [Cited by in F6Publishing: 251]  [Article Influence: 12.0]  [Reference Citation Analysis (0)]
14.  Ehrlich M. DNA methylation in cancer: too much, but also too little. Oncogene. 2002;21:5400-5413.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1124]  [Cited by in F6Publishing: 1058]  [Article Influence: 48.1]  [Reference Citation Analysis (0)]
15.  Esteller M. CpG island hypermethylation and tumor suppressor genes: a booming present, a brighter future. Oncogene. 2002;21:5427-5440.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 862]  [Cited by in F6Publishing: 850]  [Article Influence: 38.6]  [Reference Citation Analysis (0)]
16.  Jones PA, Laird PW. Cancer epigenetics comes of age. Nat Genet. 1999;21:163-167.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1657]  [Cited by in F6Publishing: 1589]  [Article Influence: 63.6]  [Reference Citation Analysis (0)]
17.  Momparler RL, Bovenzi V. DNA methylation and cancer. J Cell Physiol. 2000;183:145-154.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
18.  Esteller M. Cancer epigenetics: DNA methylation and chromatin alterations in human cancer. Adv Exp Med Biol. 2003;532:39-49.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 94]  [Cited by in F6Publishing: 93]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
19.  Nephew KP, Huang TH. Epigenetic gene silencing in cancer initiation and progression. Cancer Lett. 2003;190:125-133.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 128]  [Cited by in F6Publishing: 132]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
20.  Carnero A, Hannon GJ. The INK4 family of CDK inhibitors. Curr Top Microbiol Immunol. 1998;227:43-55.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 47]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
21.  Kamb A. Cyclin-dependent kinase inhibitors and human cancer. Curr Top Microbiol Immunol. 1998;227:139-148.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 11]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
22.  Ortega S, Malumbres M, Barbacid M. Cyclin D-dependent kinases, INK4 inhibitors and cancer. Biochim Biophys Acta. 2002;1602:73-87.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Tokugawa T, Sugihara H, Tani T, Hattori T. Modes of silencing of p16 in development of esophageal squamous cell carcinoma. Cancer Res. 2002;62:4938-4944.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Yang G, Zhang Z, Liao J, Seril D, Wang L, Goldstein S, Yang CS. Immunohistochemical studies on Waf1p21, p16, pRb and p53 in human esophageal carcinomas and neighboring epithelia from a high-risk area in northern China. Int J Cancer. 1997;72:746-751.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 2]  [Reference Citation Analysis (0)]
25.  Platz A, Hansson J, Månsson-Brahme E, Lagerlof B, Linder S, Lundqvist E, Sevigny P, Inganäs M, Ringborg U. Screening of germline mutations in the CDKN2A and CDKN2B genes in Swedish families with hereditary cutaneous melanoma. J Natl Cancer Inst. 1997;89:697-702.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 111]  [Cited by in F6Publishing: 117]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
26.  Xing EP, Nie Y, Wang LD, Yang GY, Yang CS. Aberrant methylation of p16INK4a and deletion of p15INK4b are frequent events in human esophageal cancer in Linxian, China. Carcinogenesis. 1999;20:77-84.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 89]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
27.  Sarbia M, Geddert H, Klump B, Kiel S, Iskender E, Gabbert HE. Hypermethylation of tumor suppressor genes (p16INK4A, p14ARF and APC) in adenocarcinomas of the upper gastrointestinal tract. Int J Cancer. 2004;111:224-228.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 58]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
28.  Abbaszadegan MR, Raziee HR, Ghafarzadegan K, Shakeri MT, Afsharnezhad S, Ghavamnasiry MR. Aberrant p16 methylation, a possible epigenetic risk factor in familial esophageal squamous cell carcinoma. Int J Gastrointest Cancer. 2005;36:47-54.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 35]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
29.  Xing EP, Nie Y, Song Y, Yang GY, Cai YC, Wang LD, Yang CS. Mechanisms of inactivation of p14ARF, p15INK4b, and p16INK4a genes in human esophageal squamous cell carcinoma. Clin Cancer Res. 1999;5:2704-2713.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Hardie LJ, Darnton SJ, Wallis YL, Chauhan A, Hainaut P, Wild CP, Casson AG. p16 expression in Barrett's esophagus and esophageal adenocarcinoma: association with genetic and epigenetic alterations. Cancer Lett. 2005;217:221-230.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 46]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
31.  Pekarsky Y, Zanesi N, Palamarchuk A, Huebner K, Croce CM. FHIT: from gene discovery to cancer treatment and prevention. Lancet Oncol. 2002;3:748-754.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 88]  [Cited by in F6Publishing: 94]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
32.  Sozzi G, Huebner K, Croce CM. FHIT in human cancer. Adv Cancer Res. 1998;74:141-166.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 39]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
33.  Noguchi T, Takeno S, Kimura Y, Uchida Y, Daa T, Yokoyama S, Gabbert HE, Mueller W. FHIT expression and hypermethylation in esophageal squamous cell carcinoma. Int J Mol Med. 2003;11:441-447.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Tanaka H, Shimada Y, Harada H, Shinoda M, Hatooka S, Imamura M, Ishizaki K. Methylation of the 5' CpG island of the FHIT gene is closely associated with transcriptional inactivation in esophageal squamous cell carcinomas. Cancer Res. 1998;58:3429-3434.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Lee EJ, Lee BB, Kim JW, Shim YM, Hoseok I, Han J, Cho EY, Park J, Kim DH. Aberrant methylation of Fragile Histidine Triad gene is associated with poor prognosis in early stage esophageal squamous cell carcinoma. Eur J Cancer. 2006;42:972-980.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 44]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
36.  Qiu H, Zhang W, El-Naggar AK, Lippman SM, Lin P, Lotan R, Xu XC. Loss of retinoic acid receptor-beta expression is an early event during esophageal carcinogenesis. Am J Pathol. 1999;155:1519-1523.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 77]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
37.  Wang Y, Fang MZ, Liao J, Yang GY, Nie Y, Song Y, So C, Xu X, Wang LD, Yang CS. Hypermethylation-associated inactivation of retinoic acid receptor beta in human esophageal squamous cell carcinoma. Clin Cancer Res. 2003;9:5257-5263.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Kuroki T, Trapasso F, Yendamuri S, Matsuyama A, Alder H, Mori M, Croce CM. Allele loss and promoter hypermethylation of VHL, RAR-beta, RASSF1A, and FHIT tumor suppressor genes on chromosome 3p in esophageal squamous cell carcinoma. Cancer Res. 2003;63:3724-3728.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Liu Z, Zhang L, Ding F, Li J, Guo M, Li W, Wang Y, Yu Z, Zhan Q, Wu M. 5-Aza-2'-deoxycytidine induces retinoic acid receptor-beta(2) demethylation and growth inhibition in esophageal squamous carcinoma cells. Cancer Lett. 2005;230:271-283.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 23]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
40.  Fearnhead NS, Britton MP, Bodmer WF. The ABC of APC. Hum Mol Genet. 2001;10:721-733.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 635]  [Cited by in F6Publishing: 675]  [Article Influence: 29.3]  [Reference Citation Analysis (0)]
41.  Kawakami K, Brabender J, Lord RV, Groshen S, Greenwald BD, Krasna MJ, Yin J, Fleisher AS, Abraham JM, Beer DG. Hypermethylated APC DNA in plasma and prognosis of patients with esophageal adenocarcinoma. J Natl Cancer Inst. 2000;92:1805-1811.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 261]  [Cited by in F6Publishing: 249]  [Article Influence: 10.4]  [Reference Citation Analysis (0)]
42.  Brock MV, Gou M, Akiyama Y, Muller A, Wu TT, Montgomery E, Deasel M, Germonpré P, Rubinson L, Heitmiller RF. Prognostic importance of promoter hypermethylation of multiple genes in esophageal adenocarcinoma. Clin Cancer Res. 2003;9:2912-2919.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Margison GP, Povey AC, Kaina B, Santibáñez Koref MF. Variability and regulation of O6-alkylguanine-DNA alkyltransferase. Carcinogenesis. 2003;24:625-635.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 133]  [Cited by in F6Publishing: 138]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
44.  Herfarth KK, Brent TP, Danam RP, Remack JS, Kodner IJ, Wells SA Jr, Goodfellow PJ. A specific CpG methylation pattern of the MGMT promoter region associated with reduced MGMT expression in primary colorectal cancers. Mol Carcinog. 1999;24:90-98.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
45.  Zhang L, Lu W, Miao X, Xing D, Tan W, Lin D. Inactivation of DNA repair gene O6-methylguanine-DNA methyltransferase by promoter hypermethylation and its relation to p53 mutations in esophageal squamous cell carcinoma. Carcinogenesis. 2003;24:1039-1044.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 50]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
46.  Fang MZ, Jin Z, Wang Y, Liao J, Yang GY, Wang LD, Yang CS. Promoter hypermethylation and inactivation of O(6)-methylguanine-DNA methyltransferase in esophageal squamous cell carcinomas and its reactivation in cell lines. Int J Oncol. 2005;26:615-622.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Christofori G, Semb H. The role of the cell-adhesion molecule E-cadherin as a tumour-suppressor gene. Trends Biochem Sci. 1999;24:73-76.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 525]  [Cited by in F6Publishing: 531]  [Article Influence: 21.2]  [Reference Citation Analysis (0)]
48.  Strathdee G. Epigenetic versus genetic alterations in the inactivation of E-cadherin. Semin Cancer Biol. 2002;12:373-379.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 140]  [Cited by in F6Publishing: 143]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
49.  Corn PG, Heath EI, Heitmiller R, Fogt F, Forastiere AA, Herman JG, Wu TT. Frequent hypermethylation of the 5' CpG island of E-cadherin in esophageal adenocarcinoma. Clin Cancer Res. 2001;7:2765-2769.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Takeno S, Noguchi T, Fumoto S, Kimura Y, Shibata T, Kawahara K. E-cadherin expression in patients with esophageal squamous cell carcinoma: promoter hypermethylation, Snail overexpression, and clinicopathologic implications. Am J Clin Pathol. 2004;122:78-84.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 21]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
51.  Si HX, Tsao SW, Lam KY, Srivastava G, Liu Y, Wong YC, Shen ZY, Cheung AL. E-cadherin expression is commonly downregulated by CpG island hypermethylation in esophageal carcinoma cells. Cancer Lett. 2001;173:71-78.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 47]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
52.  Kuramochi M, Fukuhara H, Nobukuni T, Kanbe T, Maruyama T, Ghosh HP, Pletcher M, Isomura M, Onizuka M, Kitamura T. TSLC1 is a tumor-suppressor gene in human non-small-cell lung cancer. Nat Genet. 2001;27:427-430.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 339]  [Cited by in F6Publishing: 349]  [Article Influence: 15.2]  [Reference Citation Analysis (0)]
53.  Ito T, Shimada Y, Hashimoto Y, Kaganoi J, Kan T, Watanabe G, Murakami Y, Imamura M. Involvement of TSLC1 in progression of esophageal squamous cell carcinoma. Cancer Res. 2003;63:6320-6326.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  Dammann R, Schagdarsurengin U, Strunnikova M, Rastetter M, Seidel C, Liu L, Tommasi S, Pfeifer GP. Epigenetic inactivation of the Ras-association domain family 1 (RASSF1A) gene and its function in human carcinogenesis. Histol Histopathol. 2003;18:665-677.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Dammann R, Li C, Yoon JH, Chin PL, Bates S, Pfeifer GP. Epigenetic inactivation of a RAS association domain family protein from the lung tumour suppressor locus 3p21.3. Nat Genet. 2000;25:315-319.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 814]  [Cited by in F6Publishing: 847]  [Article Influence: 35.3]  [Reference Citation Analysis (0)]
56.  Schagdarsurengin U, Gimm O, Hoang-Vu C, Dralle H, Pfeifer GP, Dammann R. Frequent epigenetic silencing of the CpG island promoter of RASSF1A in thyroid carcinoma. Cancer Res. 2002;62:3698-3701.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  Kuroki T, Trapasso F, Yendamuri S, Matsuyama A, Alder H, Mori M, Croce CM. Promoter hypermethylation of RASSF1A in esophageal squamous cell carcinoma. Clin Cancer Res. 2003;9:1441-1445.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  Wong ML, Tao Q, Fu L, Wong KY, Qiu GH, Law FB, Tin PC, Cheung WL, Lee PY, Tang JC. Aberrant promoter hypermethylation and silencing of the critical 3p21 tumour suppressor gene, RASSF1A, in Chinese oesophageal squamous cell carcinoma. Int J Oncol. 2006;28:767-773.  [PubMed]  [DOI]  [Cited in This Article: ]
59.  Hayashi M, Tamura G, Jin Z, Kato I, Sato M, Shibuya Y, Yang S, Motoyama T. Microsatellite instability in esophageal squamous cell carcinoma is not associated with hMLH1 promoter hypermethylation. Pathol Int. 2003;53:270-276.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 21]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
60.  Vasavi M, Ponnala S, Gujjari K, Boddu P, Bharatula RS, Prasad R, Ahuja YR, Hasan Q. DNA methylation in esophageal diseases including cancer: special reference to hMLH1 gene promoter status. Tumori. 2006;92:155-162.  [PubMed]  [DOI]  [Cited in This Article: ]
61.  Darnton SJ, Hardie LJ, Muc RS, Wild CP, Casson AG. Tissue inhibitor of metalloproteinase-3 (TIMP-3) gene is methylated in the development of esophageal adenocarcinoma: loss of expression correlates with poor prognosis. Int J Cancer. 2005;115:351-358.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 51]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
62.  Li H, Lu S, Fong L. [Study on the status of methylation of Rb gene promoter in human esophageal cancer and effect of NMBzA on Rb gene promoter in monkey esophageal epithelium]. Zhonghua Zhongliu Zazhi. 1998;20:412-414.  [PubMed]  [DOI]  [Cited in This Article: ]
63.  Yue CM, Deng DJ, Bi MX, Guo LP, Lu SH. Expression of ECRG4, a novel esophageal cancer-related gene, downregulated by CpG island hypermethylation in human esophageal squamous cell carcinoma. World J Gastroenterol. 2003;9:1174-1178.  [PubMed]  [DOI]  [Cited in This Article: ]
64.  Shibata Y, Haruki N, Kuwabara Y, Ishiguro H, Shinoda N, Sato A, Kimura M, Koyama H, Toyama T, Nishiwaki T. Chfr expression is downregulated by CpG island hypermethylation in esophageal cancer. Carcinogenesis. 2002;23:1695-1699.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 76]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
65.  Nie Y, Yang G, Song Y, Zhao X, So C, Liao J, Wang LD, Yang CS. DNA hypermethylation is a mechanism for loss of expression of the HLA class I genes in human esophageal squamous cell carcinomas. Carcinogenesis. 2001;22:1615-1623.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 143]  [Cited by in F6Publishing: 145]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
66.  Zou H, Osborn NK, Harrington JJ, Klatt KK, Molina JR, Burgart LJ, Ahlquist DA. Frequent methylation of eyes absent 4 gene in Barrett's esophagus and esophageal adenocarcinoma. Cancer Epidemiol Biomarkers Prev. 2005;14:830-834.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 52]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
67.  Hibi K, Kodera Y, Ito K, Akiyama S, Nakao A. Methylation pattern of CDH13 gene in digestive tract cancers. Br J Cancer. 2004;91:1139-1142.  [PubMed]  [DOI]  [Cited in This Article: ]
68.  Zou H, Molina JR, Harrington JJ, Osborn NK, Klatt KK, Romero Y, Burgart LJ, Ahlquist DA. Aberrant methylation of secreted frizzled-related protein genes in esophageal adenocarcinoma and Barrett's esophagus. Int J Cancer. 2005;116:584-591.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 108]  [Cited by in F6Publishing: 119]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
69.  Mandelker DL, Yamashita K, Tokumaru Y, Mimori K, Howard DL, Tanaka Y, Carvalho AL, Jiang WW, Park HL, Kim MS. PGP9.5 promoter methylation is an independent prognostic factor for esophageal squamous cell carcinoma. Cancer Res. 2005;65:4963-4968.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 95]  [Cited by in F6Publishing: 103]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
70.  Ramirez JL, Taron M, Balaña C, Sarries C, Mendez P, de Aguirre I, Nuñez L, Roig B, Queralt C, Botia M. Serum DNA as a tool for cancer patient management. Rocz Akad Med Bialymst. 2003;48:34-41.  [PubMed]  [DOI]  [Cited in This Article: ]
71.  Ziegler A, Zangemeister-Wittke U, Stahel RA. Circulating DNA: a new diagnostic gold mine. Cancer Treat Rev. 2002;28:255-271.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 165]  [Cited by in F6Publishing: 178]  [Article Influence: 8.1]  [Reference Citation Analysis (0)]
72.  Patel A, Groopman JD, Umar A. DNA methylation as a cancer-specific biomarker: from molecules to populations. Ann N Y Acad Sci. 2003;983:286-297.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 26]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
73.  Hibi K, Taguchi M, Nakayama H, Takase T, Kasai Y, Ito K, Akiyama S, Nakao A. Molecular detection of p16 promoter methylation in the serum of patients with esophageal squamous cell carcinoma. Clin Cancer Res. 2001;7:3135-3138.  [PubMed]  [DOI]  [Cited in This Article: ]
74.  Esteller M, Sanchez-Cespedes M, Rosell R, Sidransky D, Baylin SB, Herman JG. Detection of aberrant promoter hypermethylation of tumor suppressor genes in serum DNA from non-small cell lung cancer patients. Cancer Res. 1999;59:67-70.  [PubMed]  [DOI]  [Cited in This Article: ]
75.  Frühwald MC. DNA methylation patterns in cancer: novel prognostic indicators. Am J Pharmacogenomics. 2003;3:245-260.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 16]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
76.  Lichtenstein AV, Kisseljova NP. DNA methylation and carcinogenesis. Biochemistry (Mosc). 2001;66:235-255.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 26]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
77.  Nie Y, Liao J, Zhao X, Song Y, Yang GY, Wang LD, Yang CS. Detection of multiple gene hypermethylation in the development of esophageal squamous cell carcinoma. Carcinogenesis. 2002;23:1713-1720.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 91]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
78.  Bian YS, Osterheld MC, Fontolliet C, Bosman FT, Benhattar J. p16 inactivation by methylation of the CDKN2A promoter occurs early during neoplastic progression in Barrett's esophagus. Gastroenterology. 2002;122:1113-1121.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 128]  [Cited by in F6Publishing: 138]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
79.  Fang MZ, Wang Y, Ai N, Hou Z, Sun Y, Lu H, Welsh W, Yang CS. Tea polyphenol (-)-epigallocatechin-3-gallate inhibits DNA methyltransferase and reactivates methylation-silenced genes in cancer cell lines. Cancer Res. 2003;63:7563-7570.  [PubMed]  [DOI]  [Cited in This Article: ]
80.  van Groeningen CJ, Leyva A, O'Brien AM, Gall HE, Pinedo HM. Phase I and pharmacokinetic study of 5-aza-2'-deoxycytidine (NSC 127716) in cancer patients. Cancer Res. 1986;46:4831-4836.  [PubMed]  [DOI]  [Cited in This Article: ]
81.  Aparicio A, Eads CA, Leong LA, Laird PW, Newman EM, Synold TW, Baker SD, Zhao M, Weber JS. Phase I trial of continuous infusion 5-aza-2'-deoxycytidine. Cancer Chemother Pharmacol. 2003;51:231-239.  [PubMed]  [DOI]  [Cited in This Article: ]
82.  Szyf M. Targeting DNA methylation in cancer. Ageing Res Rev. 2003;2:299-328.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 72]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
83.  Leone G, Voso MT, Teofili L, Lübbert M. Inhibitors of DNA methylation in the treatment of hematological malignancies and MDS. Clin Immunol. 2003;109:89-102.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 77]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
84.  Daskalakis M, Nguyen TT, Nguyen C, Guldberg P, Köhler G, Wijermans P, Jones PA, Lübbert M. Demethylation of a hypermethylated P15/INK4B gene in patients with myelodysplastic syndrome by 5-Aza-2'-deoxycytidine (decitabine) treatment. Blood. 2002;100:2957-2964.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 399]  [Cited by in F6Publishing: 372]  [Article Influence: 16.9]  [Reference Citation Analysis (0)]
85.  Aparicio A, Weber JS. Review of the clinical experience with 5-azacytidine and 5-aza-2'-deoxycytidine in solid tumors. Curr Opin Investig Drugs. 2002;3:627-633.  [PubMed]  [DOI]  [Cited in This Article: ]