Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2018 Dec 28.
Published in final edited form as: Biochim Biophys Acta. 2012 Apr 9;1819(7):836–845. doi: 10.1016/j.bbagrm.2012.03.009

Targeting epigenetic mediators of gene expression in thoracic malignancies

David S Schrump 1,*
PMCID: PMC6310137  NIHMSID: NIHMS999481  PMID: 22507242

Abstract

Lung and esophageal cancers and malignant pleural mesotheliomas are highly lethal neoplasms that are leading causes of cancer-related deaths worldwide. Presently, limited information is available pertaining to epi-genetic mechanisms mediating initiation and progression of these neoplasms. The following presentation will focus on the potential clinical relevance of epigenomic alterations in thoracic malignancies mediated by DNA methylation, perturbations in the histone code, and polycomb group proteins, as well as ongoing translational efforts to target epigenetic regulators of gene expression for treatment of these neoplasms.

Keywords: Lung cancer, Esophageal cancer, Mesothelioma, Epigenetics, Prognosis, Clinical trials

1. Introduction

Lung and esophageal cancers and malignant pleural mesotheliomas (MPM) rank among the most lethal neoplasms worldwide [74]. In 2011, these malignancies will account for approximately 1.8 million deaths globally; in the United States, nearly 160,000 deaths will be attributed to lung cancer, 15,000 will be due to esophageal cancer, and approximately 2500 will be attributed to MPM [149,185]. A variety of genetic and environmental factors contribute to these neoplasms; of particular concern, 80% of lung cancers and 50% of esophageal carcinomas are directly attributable to tobacco smoke [1,186]. Interestingly, although not typically considered to be a tobacco-related disease, MPM occurs much more frequently in smokers [48]. These clinical observations are consistent with experimental studies demonstrating that cigarette smoke enhances asbestos-mediated development of MPM in rodents [180].

Relatively limited information is available pertaining to the frequency, mechanisms and clinical relevance of potentially reversible, epigenomic alterations that perturb gene expression in thoracic malignancies [11,14,144]. Similar to other neoplasms [9], thoracic malignancies exhibit genomic hypomethylation, primarily involving repetitive DNA sequences [45,155], with paradoxical site-specific DNA hypermethylation, which silences numerous homeobox and tumor suppressor genes [145]. Alterations in DNA methylation coincide with reversible modifications of core histones reflecting activated, repressed or bivalent chromatin structure within the respective genomic loci, and appear to recapitulate stem cell development paradigms [8,72,102].

Although recent studies suggest that global DNA demethylation may be linked to aberrant DNA repair [55,139], decreased DNMT1 expression [100,173], replication independent DNA demethylases [45,127], or formation of 5-hydroxymethylcytosine [37,75], the mechanisms mediating genomic demethylation in thoracic malignancies remain elusive. DNA hypomethylation appears to occur early during aerodigestive tract carcinogenesis. Liu et al. [100] exposed cdk-4/h-tert immortalized human bronchial epithelial cells to cigarette smoke condensate, and observed that hypomethylation of DNA repeats and upregulation of imprinted alleles preceded epi-genetic silencing of tumor suppressors. In an additional recent study, Alvarez et al. [3] observed that DNA hypomethylation is an early epigenetic event, which synergizes with gene amplification during esophageal carcinogenesis.

Irrespective of the mechanisms, global DNA demethylation facilitates de-repression of endogenous retroviruses [178], pseudogenes, and imprinted alleles [45,64,66,68,8890,163,192], thereby enhancing genomic instability (Table 1). Global DNA demethylation also results in de-repression of a variety of genes that are silent in normal somatic cells, yet exhibit stage-specific expression during germ cell development in testes or ovary (Table 1). To date, more than 100 of these “cancer-testis (CT)” genes have been identified, approximately half of which map to the X chromosome [152]. Relative to autosomal CT genes, CT-X chromosome (CT-X) genes are more frequently activated in cancer cells, and particular gene families appear to be coordinately de-repressed in a tumor-specific manner.

Table 1.

Genes frequently de-repressed in thoracic malignancies.

Chromosome
Imprinted genes
H19 [88] 11p15.5
IGF2 [90] 11p15.5
MEST [88] 7q32
Cancer testis genes
BORIS [67] 20q13.2
MAGE A3/6 [87,146] Xq28
NY-ESO-1 [84,87] Xq28
LAGE-1 [87] Xq28
GAGE [2,54,108] Xp11.4
XAGE [115] Xp11.22
TRAG-3 [27] Xq28
OY-TES-1 [121] 12p13
CT-45 [26] Xq26–3
CTSP-1 [125] 21q11.2

Although several studies have suggested that de-repression of CT genes enhances the malignant phenotype of cancer cells by up-regulation of additional CT genes [67,161], activation of telomerase [130], induction of cell cycle progression and inhibition of p53 function [86,106,113,126,176], or increasing resistance to death receptor ligands, radiation or chemotherapeutic agents [4,34], limited evidence indicates that CT gene activation directly impacts prognosis of patients with thoracic malignancies. Melloni et al. [108] observed that GAGE expression correlated with diminished survival of lung cancer patients. Gure et al.[54] examined CT-X gene expression in tumors from over 500 lung cancer patients undergoing potentially curative resections. De-repression of CT-X genes coincided with tobacco use and advanced stage of disease; expression of NY-ESO-1 and/or MAGE-A3 correlated with poor survival. Consistent with these findings, Yanagawa et al. [174] observed that MAGE-A1 or A3 expression in lung cancers correlated with adverse patient outcome. In contrast, Akcakanat et al. [2] detected expression of GAGE, NY-ESO-1 and MAGE-A1 protein expression in 20%, 21% and 52% of 213 esophageal cancer specimens, respectively. De-repression of these CT genes did not correlate with tumor stage, disease progression, or patient survival. To date the clinical relevance of CT genes activation in MPM has not been established.

Unlike the effects of DNA hypomethylation, the clinical ramifications of aberrant silencing of tumor suppressor genes in thoracic malignancies are more evident (Table 2). The remaining presentation will focus on the potential clinical relevance of epigenomic alterations in thoracic malignancies mediated by DNA methylation, perturbations in the histone code, and polycomb group proteins (Table 3), as well as translational efforts to target epigenetic regulators of gene expression for treatment of these neoplasms.

Table 2.

Tumor suppressor genes hypermethylated with prognostic significance in thoracic malignancies.

DAPK [19,47,103] CDH-13 [16]
FHIT [83,98,117] TFPI-2 [169]
HIC-1 [58] TIMP-3 [39]
APC [15,16] RUNX3 [175]
WIF-1 [181] DLEC1 [148,191]
Cdh1 [93,117] RAR-β [47,98]
P16/pl4ARF [16,83,98,181] RASSF1A [16,19,47,175]

Table 3.

Epigenetic regulators of gene expression implicated in thoracic malignancies.

DNMT1 [82,96,98,150] SUV39H1 [165]
DNMT3b [82,98,150,159] SUV39H2 [179]
MBD2 [170] G9a [25,165]
HDAC1 [134] KDM1 [56]
HDAC3 [110,111] JARID1B [57]
HDAC5 [122] BMI1 [61,101,162,190]
HDAC8 [116] HDAC10 [122]
EZH2 [59,60,80,81,172]

2. DNA methyltransferases and methylated DNA binding proteins

DNA methylation is the major epigenetic mechanism mediating dynamic patterns of gene expression during differentiation and normal cellular homeostasis [63], as well as long-term repression of gene expression associated with imprinting and X chromosome inactivation [9,40,65]. To date, three major DNMTs (DNMT1, 3a, and 3b), with numerous isoforms have been characterized in somatic cells [29], all of which mediate transfer of a methyl group from S-adenosyl-methionine to the 5′ position of cytosine in the context of CpG. Whereas the specificities of various isoforms have not been fully elucidated, and considerable overlap exists, DNMT1 binds preferentially to hemimethylated DNA, and functions primarily as a maintenance methyltransferase; DNMT3a and 3b, interact primarily with unmethylated or hemimethylated DNA to mediate de-novo DNA methylation [168]. An additional isoform-designated DNMT3L, which is normally expressed only in germ cells[51], lacks methyltransferase activity, and functions to enhance targeting of DNMT3a and 3b to DNA [168].

Several laboratory studies indicate that aberrant expression of DNMTs contributes to the pathogenesis of thoracic malignancies [38,100]. Furthermore, increasing evidence indicates that altered expression of DNMTs may be of prognostic significance in these neoplasms. For example, Kim et al. [82] observed over-expression of DNMT1 and DNMT3b in approximately 50% of lung cancer specimens; over-expression of DNMT1 correlated with p16 promoter hyper-methylation, and diminished patient survival. Lin et al. [98] reported that coordinate expression of DNMT1, 3a, and 3b coincided with increased methylation of a variety of tumor suppressor genes including p16, FHIT and RAR-β in lung cancers, and diminished survival of patients with these neoplasms; this phenomenon was especially pronounced in smokers with squamous cell carcinomas. In a subsequent study, Lin et al. [96] examined DNMT1 expression in 124 lung cancers, and observed that high-level intranuclear DNMT1 levels correlated significantly with smoking status, and diminished patient survival. Vallbohmer et al. [159] examined DNMT1, 3a and 3b expression levels relative to methylation status of APC, DAPK, GSTP1 and MGMT in 91 lung cancer specimens and adjacent normal lung samples. Expression of the three DNMTs was higher in tumors compared to normal lung. Whereas no correlation was observed between DNMT expression and DNA methylation, hypermethylation status was an independent adverse prognostic factor. Consistent with these findings, Brock et al.[16] observed that methylation of cdk2a, p16, CDH13, RASSF1A, and APC in resected early stage (node negative) lung cancers was associated with tumor recurrence, independent of histology or stage, gender, or smoking history.

Xing et al. [170] used quantitative RT-PCR techniques to examine DMNT1 and DNMT3b, as well as methylated DNA-binding protein 2 (MBD2) expression in 148 resected NSCLCs. High-level DNMT1 expression correlated significantly with increased risk of cancer-related death in all patients, whereas increased DNMT3b expression was associated with poor outcome in patients less than 65 years of age. High-level expression of MBD2 correlated with poor survival in male patients and those with squamous cell carcinomas. Combinatorial effects of DNMT1 and 3b, DMNT1 and MBD2, and DNMT3b and MBD2 were evident in male patients, and those with squamous cell carcinomas. Collectively, aforementioned clinical data as well as laboratory experiments indicating that knock-down of DNMT1 and/or 3b induces genotoxic stress and apoptosis [79], whereas knock-down of MBD2 inhibits growth and tumorigenicity of cultured lung cancer cells [21], attest to the relevance of aberrant expression of these modulators of DNA methylation during pulmonary carcinogenesis.

Several studies have been performed to examine DNMT expression, promoter methylation, and outcome in patients with esophageal cancers. Simao et al. [150] observed over-expression of DNMT1, DNMT3a, and DNMT3b in 35%, 41%, and 65% of esophageal squamous cell carcinomas (ESCC), relative to paired normal esophageal mucosae; over-expression of DNMT3b correlated with down-regulation of p16/p14. Li et al. [95] examined promoter methylation status of 12 tumor suppressor genes (TSG) using MSP in ESCC and paired normal esophageal tissues from 47 patients. Forty-six of 47 ESCC exhibited methylation of at least one of these TSG, with methylation being most frequently observed for RAR-β, DAPK, p16 and CDH1. Over-expression of DNMT1, DMNT3a and DNMT3b was observed in 48%, 48%, and 72% of tumors, respectively. A significant increase in DNMT3b levels was observed, which correlated with hypermethylation of TSGs. No significant correlation was observed between promoter hypermethylation, nodal status, or smoking/ethanol history.

Ling et al. [99] examined promoter methylation status of 9 TSGs in 50 ESCC, 50 dysplastic lesions, and 50 normal esophageal samples. CpG island methylation phenotype (CIMP), defined as 5 or more methylated genes, was identified in 54% of ESCC compared to 8% dys-plastic tissues, and 0% of normal esophageal samples. CIMP positivity correlated with more advanced TNM stage and metastases. Furthermore, patients with CIMP-positive tumors had significantly shorter survivals relative to patients with CIMP-negative tumors following esophagectomy. These findings are consistent with data reported by Brock et al. [15] who observed that patients with esophageal adenocarcinomas exhibiting methylation of >50% of seven TSGs had significantly poorer survival and earlier tumor recurrence than patients without such methylation. Interestingly, CIMP status was a more powerful predictor of survival than tumor stage.

Limited information is available regarding DNMT expression levels and aberrant DNA methylation in MPM. Christensen et al. [31] examined methylation status of six cell cycle regulatory genes in 70 MPM specimens, and observed significant correlation between methylation status and asbestos body burden, which remained significant irrespective of gender, age or tumor histology. In a subsequent study, Christensen et al. [32] observed that extent of gene copy number alterations correlated with DNA methylation profile, an association that appeared to be due, in part, to allelic loss of DNMT1. Fischer et al.[47] examined methylation status of nine genes in serum DNA, and observed that methylation status of any single gene had no association with survival. However, combining RAR-β with either RASSFIA or DAPK correlated significantly with decreased survival. Furthermore, patients with tumors exhibiting methylation of any two or all three of these TSGs did significantly worse than those with no methylation of these genes, suggesting that the extent of DNA methylation coincided with aggressive tumor phenotype. Destro et al. [41] evaluated methylation status of p15, p16, RASSFIA and NORE1A in 79 MPM patients. Methylation of at least one of these genes was observed in 30% of the cases (the frequency of methylation ranged from 5 to 20%). Patients undergoing extra-pleural pneumonectomy for MPM with DNA methylation had shorter median overall survivals compared to patients with tumors lacking methylation of these genes (16 vs. 35 months; p=0.06). Collectively, these data suggest that extent of DNA methylation coincides with more aggressive tumor histology and decreased survival of patients with MPM.

3. Histone acetylases/deacetylases and histone methyltransferases/demethylases

Lysine rich tails of core histone proteins (H2A, H2B, H3, and H4), protrude from the nucleosome, providing sites for highly diverse, reversible, covalent modifications such as acetylation, methylation, phosphorylation, sumoylation, and ubiquitination that alter chromatin structure and modulate gene expression [7,94,140]. To date, alterations in histone acetylation/deacetylation and methylation/demethylation, have been the most extensively characterized histone modifications in cancer cells [10,137,141,182]. Histone acetylation/deacetylation is mediated by a variety of histone acetyltransferases (HATs) including GNAT, MYST, and p300 families [10,42], whereas histone deacetylation is mediated by histone deacetytlases (HDACs), which are divided into four classes based on phylogenetic and functional criteria [13,141]. Histone acetylation increases net negative charge leading to repulsion of DNA, relaxation of chromatin, and activation of gene expression [5]. Many non-histone proteins including p53, Hsp90, Sp1, and HDAC1 are also targets for HATS/HDACs [30,141,151].

Histone lysine methylation is mediated by a variety of histone methyltransferases (KMTs) including SET, MLL, SUV39H, and EZH1/2 families as well as, RIZI, G9a, ASH1, and DOT1L that mediate mono-, di- and trimethylation of specific lysine residues [50,62]; histone demethylation is mediated by histone demethyltransferases (KDMTs) comprising two groups (LSD1) and jumonji-domain containing (JHDM) proteins [35,50,119,182]. In contrast to histone acetylation, histone ly-sine methylation does not alter charge of core histones. Depending on the site, histone lysine methylation may either facilitate or inhibit gene expression. For example, methylation of histone H3K9 and H3K27 coincides with transcriptional repression; in contrast, methylation at H3K4, H3K36 or H3K79 tends to be associated with gene activation (reviewed in ref. [50,119]). A variety of non-histone proteins including p53, E2F1, and NFB are targets of HMTs and HDMTs [62,189]. Revised nomenclature and substrates for histone lysine demethylases have been reviewed recently [35].

Relatively limited information is available concerning the direct clinical implications of aberrant expression of HATs and HDACs, as well as HMTs and HDMTs in thoracic malignancies. Increased HDAC1 mRNA levels appear to coincide with advanced stage of disease in lung cancer patients [134]. Interestingly, HDAC1 deacetylates DNMT1, thereby protecting it from proteosomal degradation providing a potential mechanistic link between HDAC1 over-expression and aberrant DNA methylation during malignant transformation [43]. In a recent study, lung cancer patients with high intratumor levels of HDAC3 had significantly shorter disease-free survivals than patients whose tumors exhibited low HDAC3 expression [110]. Multivariate analysis revealed that HDAC3 over-expression was an independent prognosticator of poor survival in patients with adenocarcinomas, but not those with squamous cell carcinomas. In an additional study, Osada et al. [122] observed that reduced expression of several class II HDACs (particularly HDACs 5 and 10) correlated with adverse outcome in lung cancer patients.

Toh et al. [156] utilized immunohistochemistry techniques to examine levels of HDAC1 and histone H4 acetylation in ESCC, and observed H4 hyperacetylation in early lesions, with progressive H4 hypoacetylation in invasive lesions. HDAC1 levels also seemed to decrease with tumor invasion. In a subsequent study, Langer et al. [92] examined HDAC1 and HDAC2 levels in 180 esophageal carcinomas. Approximately 50% of tumors had no detectable or very low HDAC1 expression, whereas 30% of the tumors had no detectable HDAC2 expression. High HDAC2 expression correlated with poor differentiation and lymph node metastases. Interestingly no correlation was observed between HDAC1 or HDAC2 expression and response to therapy or survival.

Less information is available regarding roles of specific KMTs or KDMTs in the pathogenesis of thoracic malignancies. Kim et al. [85] observed up-regulation of DOT1L, which mediates methylation of H3K79 in cultured lung cancer cells and resected lung cancer specimens. Knock-down of DOT1L resulted in decreased proliferation of lung cancer cells, a phenomenon that coincided with aberrant mitotic spindles, chromosomal missegregation, and senescence; these findings suggest that increased expression/activity of this KMT enhances the malignant phenotype of lung cancer cells. Using immunohistochemistry techniques, Hudlebush et al. [69] observed over-expression of multiple myeloma SET (MMSET) histone lysine methyltransferase, which mediates di and tri methylation of H3K36 in a variety of human malignancies including lung and esophageal cancers, and pleural mesotheliomas. This study extends earlier microarray studies reported by Kassamabara et al. [78] demonstrating that numerous human malignancies including lung and esophageal cancers over-express MMSET, and that MMSET up-regulation correlates with advanced stage of disease, and in some tumor histologies, patient survival as well. Hayami et al. [57] observed over-expression of JARID1B, which demethylates H3K4Me3/Me2, in a large panel of small cell as well as non-small cell lung cancers. JARID1B over-expression correlated significantly with increased expression of E2F1 and E2F2. In subsequent studies, Hayami et al. [56] observed up-regulation of LSD1, which catalyzes demethylation of H3K4Me2/Me1 and possibly H3K9Me2/Me1, in small cell lung cancers relative to normal lung tissues; knock-down of LSD1 induced G1 arrest without apotosis in cultured lung cancer cells. Consistent with these findings, Watanabe et al. [165] observed that knock-down of EZH2, which mediates trimethylation of H3K27,or knockdown of SETDB1, G9a, and SUV39H1, which methylate H3K9, inhibited proliferation and soft agar colony formation of immortalized or fully transformed respiratory epithelial cells, implicating these modifiers of histone methylation in the pathogenesis of lung cancer. Chen et al. [25] observed that G9a expression correlates with aggressive phenotype of lung cancer cells, and poor prognosis in lung cancer patients; over-expression of G9a coincided with decreased H3K9 methylation and repression of the cell adhesion molecule Ep-Cam.

Several recent studies suggest that the summation of perturbed acetylation and methylation of core histones observed in lung cancers may be clinically relevant. Seligson et al. [147] reported that decreased global levels of H3K4Me2 and H3K18Ac correlated significantly with reduced survival of patients with early stage lung cancers undergoing potentially curative resections. Van Den Broeck [160] observed hyperacetylation of H4K5 and H4K8, hypoacetylation of H4K12/H4K16, and decreased H4K20Me3 levels in lung cancer cells relative to adjacent normal respiratory epithelia. Whereas loss of H4K20Me3 was more common in squamous cell carcinomas, this his-tone alteration, which coincided with diminished expression of SUV4–20h2, was not associated with patient outcome. In contrast, loss of H4K20Me3 in early stage adenocarcinomas correlated significantly with decreased patient survival.

Barlési et al. [6] classified 138 lung cancer patients into seven groups based on histology, stage, and global expression levels of H3K4Me2, H2AK5Ac, and H3K9Ac; the groups exhibited significant differences in disease free, as well as overall survivals. Interestingly, the four groups comprising Stage I patients displayed dramatic differences, with a median survival of ten months for adenocarcinoma patients with high-level intratumoral H3K9Ac, compared to 147 months for non-adenocarcinoma patients with high-level intratumoral H3K4Me2 expression.

Several studies have been performed to examine histone alterations in esophageal cancers. For example, Chen et al. [24] used immunohisto-chemistry techniques to examine expression of H3/H4 acetylation and H3K4/H3K27 methylation levels in 30 ESCC patients; none had undergone chemo-or radiation therapy. In addition, qRT-PCR techniques were used to examine expression levels of the respective HATs, HDACs, KMTs, and KDMTs. H3 and H4 acetylation levels were significantly reduced, whereas H3K4 and K3K27 methylation levels were higher in tumor samples compared to adjacent normal esophageal mucosa. The extent of H3 hypoacetylation and H3K27Me3 levels correlated with advanced tumor stage. H4 hypoacetylation coincided with alcohol consumption, whereas H3 hypoacetylation as well as HDAC2 levels correlated with lymph node metastases. Levels of HDAC1, HDAC2, SIRT1 and SUV39H1 were significantly higher in more advanced tumors.

In an additional study, I et al. [71] examined the effects of histone modifications (H3K9Ac, H3K18Ac, H4K12Ac, and H4R3Me2) on recurrence free survival in 237 patients undergoing esophagectomy for esophageal squamous cell carcinomas. Patients whose tumors expressed high global levels of H3K18Ac and H4R3Me2 had significantly reduced recurrence free survivals relative to comparably staged patients with tumors exhibiting low global levels of these his-tone marks. Tzao et al. [158] examined expression status of H3K18Ac, H4K12Ac, H4R3Me2, H3K4Me2, and H3K27Me3 in 97 ESCC. A positive correlation was observed for H3K18Ac, H4R3Me2, and K3K27Me3 and poor tumor differentiation. Low levels of H3K18Ac and H3K27Me3 were associated with improved prognosis in early stage tumors. Collectively, these studies attest to the relevance of perturbed histone marks in thoracic malignancies.

4. Polycomb group proteins

Polycomb group proteins (PcG) have emerged as critical epigenetic mediators of pluripotency and differentiation of stem cells [33,138], as well as aberrant gene repression during malignant transformation [109,136]. Two major polycomb repressor complexes, each of which contains a variety of core subunits have been identified in mammals. The initiation complex, PRC-2, containing EZH2, EED, PCL, and SUZ12 subunits, mediates trimethylation of H3K27. PRC-2 recruits the maintenance complex PRC-1, containing core subunits of PCAF, PHC, RING1, CBX, and SML, as well as SFMBT and L3MBTL proteins that mediate ubiquitination of H2AK119 [136]. These histone marks coincide with formation of heterochromatin, and repression of gene expression by mechanisms, which to date, have not been fully elucidated [46,114]. Of particular relevance regarding the role of PcG proteins in tumorigenesis are recent observations that EZH2 is increased and essential for maintenance of cancer stem cells [132,154].

Several recent studies highlight the potential relevance of aberrant Polycomb expression/activity in thoracic malignancies. For example, stem cell polycomb group targets are significantly more likely to have cancer-specific promoter hypermethylation than non-polycomb targets [76,167]. Consistent with these findings, Liu et al. [100] observed that polycomb target genes were seven-fold more likely to undergo DNA hypermethylation in cultured human bronchial epithelial cells exposed to cigarette smoke condensate.

Hussain et al. [70] observed polycomb-mediated repression of Dickkopf-1 (Dkk-1), which encodes a secreted antagonist of Wnt signaling in normal human small airway epithelial and lung cancer cells exposed to cigarette smoke. Knock-down of EZH2 abrogated CSC-mediated inhibition of Dkk-1. Furthermore, knock-down of Dkk-1 increased non-canonical Wnt signaling and increased tumorigenicity of lung cancer cells. Collectively, these data provided the first direct evidence that cigarette smoke engages polycomb machinery to silence tumor suppressor genes during pulmonary carcinogenesis.

In additional experiments, Liu et al. (unpublished) observed over-expression of EZH2 in a panel of lung cancer lines relative to cultured normal respiratory epithelial cells. Knock-down of KMT6 inhibited proliferation and tumorigenicity of lung cancer cells. Growth inhibition coincided with cell cycle arrest and senescence. Microarray analysis revealed that knock-down of EZH2 resulted in up-regulation of numerous gene regulatory networks mediating cell cycle control, differentiation and senescence; many of the genes activated by knockdown of EZH2 were stem cell polycomb targets.

Several potential mechanisms contribute to aberrant expression of PcG genes in thoracic malignancies. For example, the vast majority of lung and esophageal cancers and MPM exhibit disruption of Rb-mediated regulation of E2F; EZH2, EED, and SUZ12 are potential E2F targets [12,36]. Inactivation of p16, which is observed in 30–40% of lung and esophageal cancers, and the majority of MPM [14,73,135], results in up-regulation of KMT6 and SUZ12 [131]. Furthermore, several micro-RNAs such as miR-101 and miR-26a, and miR98 which target the 3′ UTR of EZH2 are down-regulated in aerodigestive tract cancers [49,80,104,187].

Several recent studies have examined the prognostic significance of aberrant PcG protein expression in thoracic malignancies. Vrzalikova et al. [162] used immunohistochemistry (IHC) techniques to examine expression of BMI1 (a component of PRC-1) in 179 NSCLCs including 106 squamous cell, 58 adeno-, and 15 large cell carcinomas. Nearly 70% of samples exhibited moderate or strong nuclear immunoreactivity for BMI1. Subsequent analysis revealed that BMI1 expression correlated significantly with advanced stage of disease (stages III or IV), as well as decreased disease free survival for patients with stage I or II neoplasms. An additional study pertaining to IHC analysis of 134 resected pulmonary adenocarcinomas, demonstrated that BMI1 expression was higher in stage III tumors relative to stage I or II cancers; multivariate analysis revealed that BMI1 expression was an independent prognosticator of poor survival of adenocarcinoma patients [190].

Kikuchi et al. [81] used IHC techniques to examine BMI1 and EZH2 expression in 157 resected NSCLC specimens, including 65 squamous cell carcinomas, 82 adenocarcinomas, 7 adenosquamous carcinomas, and 2 large cell carcinomas. In contrast to aforementioned results, BMI1 expression did not appear to be associated with any clinicopatho-logic features. However, high intratumoral EZH2 expression correlated significantly with non-adenocarcinoma histology, increased proliferation index, moderate to poor differentiation, and significantly decreased patient survival in all stages; the adverse impact of EZH2 expression was particularly apparent in stage I neoplasms.

Several studies have been undertaken to examine the clinical significance of aberrant PcG protein expression in esophageal cancers. He et al. [61] observed that expression of BMI1 mRNA was significantly higher in ESCC compared to adjacent normal tissues. High level BMI1 expression correlated with lymph node metastases, poor differentiation, and advanced pathologic stage. Liu et al. [101] examined BMI1 protein expression in 171 ESCC. BMI1 was detected in 64% of these specimens. BMI1 expression was higher in more advanced tumors. Multivariate analysis suggested that BMI1 expression was an independent adverse prognostic factor. In additional studies, He et al. [59] examined EZH2 expression in approximately 100 pretreatment biopsies of patients with ESCC who received definitive chemo-radiation therapy. High level expression of EZH2 was seen in 54% of cases, and correlated with increased proliferation, high tumor grade, lymph node metastases, and lack of complete response. Multivariate analysis revealed that EZH2 expression was an independent risk factor for poor progressive free survival, and disease specific survival. Consistent with these findings He et al. [60] observed a positive correlation between EZH2 and H3K27Me3 levels, advanced pathologic stage, poor loco-regional progression free survival, and incomplete response to chemo-radiation therapy in the same patients used in the previous study. More recently, Yamada et al. [172] evaluated BMI1 and EZH2 expression in 136 ESCC specimens, and adjacent normal tissues. Approximately 15% of the specimens exhibited over-expression of BMI1 or EZH2. Aberrant expression of EZH2 correlated with larger, more invasive tumors, distant metastases and shorter disease free survival. In contrast BMI1 expression did not appear to coincide with survival.

In a recent study, Kemp et al. [80] observed over-expression of EZH2 in cultured MPM cells relative to normal mesothelia. Subsequent qRT-PCR, immunoblot and immunohistochemistry experiments demonstrated over-expression of EZH2 in 85% of MPM; in contrast, EZH2 expression was not detected in normal pleura or peritoneum. EZH2 over-expression coincided with increased global H3K27Me3 levels, and correlated significantly with diminished survival of MPM patients undergoing potentially curative resections. Knock-down of EZH2 decreased proliferation, migration, clonogenicity and tumorigenicity of mesothelioma cells. Interestingly, although EED was not over-expressed in MPM, the anti-tumor effects of knock-down of EED were more profound than EZH2 depletion, possibly due to relative knockdown efficiencies, compensation of EZH2 knock-down by EZH1, or more profound destabilization of PRC-2 by depletion of EED. The effects of PRC-2 knock-down were recapitulated by the nucleoside analog, 3-deazaneplanocin A (DZNep), which has been shown recently to deplete PRC-2 components in cancer cells [129]. Collectively, these experiments highlight the clinical relevance of EZH2 over-expression in MPM, and suggest that targeting PRC-2 expression/activity may be a novel strategy for mesothelioma therapy.

5. Translational implications

Alterations in chromatin structure secondary to aberrant expression/function of epigenetic regulators of gene expression in thoracic malignancies result in silencing of numerous tumor suppressor genes via polycomb and DNA hypermethylation mechanisms with paradoxical de-repression of CT genes in thoracic malignancies [8,145]. Because CT genes are typically expressed only in immune-privileged sites, proteins encoded by these genes induce humoral as well as cell-mediated immune responses when aberrantly expressed in somatic cells; as such, cancer-testis antigens (CTA) have emerged as highly attractive targets for cancer immunotherapy [20]. Vaccines targeting CTAs such as NY-ESO-1, MAGE-A1, and MAGE-A3 induce anti-tumor immunity, and T cells expressing native or genetically-engineered receptors recognizing these antigens mediate tumor regression in some cancer patients [22,77,133].

Whereas NY-ESO-1, MAGE-A1, and MAGE-A3 are expressed in 25–40% of lung and esophageal cancers, and MPM, [145], immune responses to these CTAs are uncommon in patients with these neoplasms [52,153], due in part to levels of antigen expression, which are below the threshold for immune recognition. Conceivably, up-regulation of CTAs expression by chromatin remodeling agents can enhance immunogenicity of lung and esophageal cancer and MPM cells, facilitating their eradication by endogenous immune mechanisms, or adoptively transferred T cells. In preclinical studies, we have demonstrated that under exposure conditions that reactivate aberrantly silenced tumor suppressor genes, DAC and the HDAC inhibitor depsipeptide (romidepsin; DP), or DAC and DZNep mediate synergistic induction of CT gene expression in cultured lung cancer cells, but not normal respiratory epithelia or lymphoid cells [67,129]. In addition, we have reported that following DAC, sequential DAC/DP, or concurrent DAC-DZNep exposure, lung and esophageal cancer and MPM cells can be recognized by cytolytic T lymphocytes (CTL) expressing receptors specific for NY-ESO-1 or MAGE-A3 [28,129,164,166]. Furthermore, we have demonstrated that a CTA induced in tumor cells in vivo by systemic DAC administration can be effectively targeted by adoptively-transferred CTL in immunocompetent mice [53]. Collectively, these observations provide strong rationale for the use of chromatin remodeling agents as a means to simultaneously inhibit growth and enhance immunogenicity of thoracic malignancies in clinical settings.

To date, most epigenetic therapies for thoracic malignancies have focused on the evaluation of pharmacologic inhibitors of DNMT and HDAC activity [17,141]. Specifically these trials have evaluated the nucleoside analogs, 5-azacytidine (5-AC) and 5-aza-2′ deoxycytidine (DAC) as well as HDAC inhibitors (HDACi) such as depsipeptide (DP; Romidepsin), sodium butyrate, and SAHA (Vorinostat) either alone or in combination with other chromatin remodeling agents or conventional chemotherapy regimens. When evaluating results of trials utilizing chromatin remodeling agents in cancer patients, it is important to remember that the anti-tumor effects of DNA demethylating agents and HDACi are not solely attributable to re-activation of tumor suppressor genes aberrantly silenced by epigenetic mechanisms [171]. For instance, cytotoxicity mediated by DAC is attributable in part to formation of DNMT-DNA adducts (particularly those involving DNMT 3a and 3b) [120] as well as DNA damage [124]. HDACi may facilitate acetylation of a variety of non-histone proteins such as p53 and Hsp90, thereby modulating cell cycle progression and apoptosis in cancer cells [30,97]. Furthermore, DNA demethylating agents in combination with HDACi induce DNA damage [105], and inhibit removal of incorporated abases [23] in lung cancer cells. Similar issues must be kept in mind when examining mechanisms by which KMT/KDMT inhibitors mediate cytotoxicity in cancer cells.

In a phase I/II study, Momparler et al. [112] treated 15 lung cancer patients with DAC administered at various doses over 8 h, and observed prolonged survival (>5 yr) in one of nine evaluable patients. In a more recent phase I trial, Schrump et al. [143] treated 35 patients (including 22 with lung cancer, 4 with esophageal cancer and 6 with MPM) with escalating doses of DAC administered via continuous 72 h infusions. The maximum tolerated dose of DAC was 75 mg/m2; myelosuppression was dose limiting. Steady state plasma DAC concentrations ranged from 25 to 50 ng/ml, which closely approximated threshold exposures for gene activation in cultured cancer cells. Although no objective responses were observed, two lung cancer patients exhibited prolonged stabilization of disease (>1 yr). Nearly one quarter of all patients exhibited expression of p16, MAGE-3, or NY-ESO-1 in post-treatment tumor biopsies. Serologic responses to NY-ESO-1 were observed in several patients receiving DAC infusions for >6 months.

In a phase II trial, Schrump et al. [142] treated 18 lung cancer patients with Romidepsin (~18 mg/m2) administered as a 4 h infusion on days 1 and 7 of a 21 day cycle. Transient stabilization of disease (2–6 months) was observed in 9 patients. Steady state Romidepsin levels during infusion ranged from 384 to 1114 ng/ml (median: 667 ng/ml), decreasing to ~10 ng/ml within 4 h following the infusion. Intratumoral levels of H3Ac and p21 (a major target of HDAC inhibitors) were increased in approximately 50% of patients following Romidepsin therapy. In addition, several patients exhibited enhanced expression of NY-ESO-1 and MAGE-A3 in tumor biopsies following Romidepsin infusions.

In a more recent trial, Otterson et al. [123] treated 16 SCLC patients with Romidepsin (13 mg/m2) administered as 4 h infusions on days 1, 8 and 16 of a 29 day cycle. No objective responses were observed; transient stabilization of disease was observed in 3 patients. Median progression free survival was less than two months, and median overall survival approximated six months. In an additional phase II trial, Traynor et al. [157] treated 16 patients with relapsed NSCLC with Vorinostat 400 mg PO daily. No objective responses were observed in 14 evaluable patients. Median time to progression was 2.3 months, which was similar to that observed for lung cancer patients receiving other targeted agents. Collectively, results of these lung cancer trials are consistent with broader experience demonstrating minimal activity of HDAC inhibitors as single agents in solid tumors [141].

Numerous pre-clinical studies have demonstrated either additive or synergistic pro-apoptotic effects of HDAC inhibitors in combination with standard cytotoxic chemotherapeutics or targeted agents in lung cancer cells [18,177,184,188]. The mechanisms underlying this phenomenon appear to be related to HDAC inhibitor-mediated induction of reactive oxygen species, depletion of oncoprotein expression, and inhibition of survival signaling [141,183,184]. Ramalingam et al. [128] randomized 94 chemo-naive patients with stage IV NSCLC to receive carboplation and paclitaxel with either Vorinostat (400 mg) on days 1–14 of a 21 day cycle, or placebo. The median number of treatment cycles was four in both arms. Median response rate was significantly improved in patients receiving Vorinostat vs placebo (34% vs12.5%). Whereas progression free and overall survivals in patients receiving Vorinostat increased 2–3 months, this survival benefit was not statistically significant.

In a recent phase I trial, Schrump et al. (manuscript in preparation) treated 31 patients with thoracic neoplasms (including 20 lung cancer, 4 esophageal cancer and 3 MPM patients) with sequential DAC/Romidepsin infusions. DAC was administered as a continuous 72 h infusion commencing on day 1 of a 35 day treatment cycle; Romidepsin was administered as a 4 h infusion immediately following DAC, and on day10. Median steady state DAC and Romidepsin levels were 13.4 ng/ml (range: 4.7–78 ng/ml), and 323 ng/ml (range: 136–923 ng/ml), respectively. Whereas no objective tumor regressions were observed, several patients exhibited stabilization of disease lasting 4–6 months. Correlative gene expression profiling experiments demonstrated that sequential DAC/Romidepsin regimens partially reversed aberrant lung cancer gene expression in vivo. The fact that these molecular responses did not translate into more prolonged disease stabilization or tumor regressions strongly suggests suboptimal drug concentrations and exposure durations.

In an ongoing phase I dose-escalation trial at the NCI, 22 patients have received sequential Romidepsin/Flavopiridol infusions, based on preclinical studies demonstrating that abrogation of p21 induction by Flavopiridol (a synthetic cdk inhibitor that also destabilizes RNA [91]) markedly enhances DP-mediated apoptosis in cultured lung and esophageal cancer and MPM cells [118]. To date, 7 patients with lung cancer, 4 with esophageal cancer, 4 with MPM, and 3 with thymoma have received 4 h DP infusions followed by 72 h Flavopiridol infusions under exposure conditions mimicking those used in preclinical experiments. Whereas no objective responses have been observed, seven patients with refractory cancers have exhibited disease stabilization lasting 4 to greater than 12 months.

6. Conclusions

Considerable data suggest that epigenetic regulators of gene expression contribute to initiation and progression of thoracic malignancies. As such, these mediators or their co-factors are attractive targets for intervention in these malignancies. Of particular concern regarding the use of chromatin remodeling agents for these neoplasms are observations that genes aberrantly hypermethylated in cancer cells do not revert to a fully euchromatin state [107], and that genes, which have been de-repressed by DNA demethylating agents cannot be maintained in an active state by HDAC inhibitors [44]. These findings, together with results of recent clinical trials suggest that epigenetic therapies may need to be administered for prolonged periods to be ef-ficacious. Novel agents such as DZNep, which depletes PRC-2 components, as well as other histone KMT/KDMT inhibitors are currently in preclinical development. One intriguing issue regarding the development of inhibitors of KMTs and KDMTs pertains to the apparent simultaneous targeting of activation and repressive methylation marks; a critical aspect of development of these compounds will be delineation of the hierarchy of methylation marks, which coincide with anti-tumor activity, in order to improve the specificity and reduce potential toxicities of these agents.

Moving forward, it appears that epigenetic strategies will need to be combined with conventional chemotherapeutics, molecularly-targeted agents, or immunotherapy regimens to mediate regression of thoracic neoplasms in clinical settings. Further analysis of epigenetic mechanisms associated with malignant transformation, as well as the identification of novel compounds that specifically target components of the epigenetic machinery that are dysregulated in thoracic malignancies may hasten the development of more efficacious epigenetic regimens for the treatment and possible prevention of these neoplasms.

Acknowledgments

The author expresses his appreciation to Ms Tricia F. Kunst, RN for assistance with patient recruitment/care, and to Ms. Jan Pappas for assistance regarding manuscript preparation.

Footnotes

This article is part of a Special Issue entitled: Chromatin in time and space.

References

  • [1].Smoking-attributable mortality, years of potential life lost, and productivity losses — United States, 2000–2004, MMWR Morb. Mortal. Wkly. Rep 57 (2008) 1226. [PubMed] [Google Scholar]
  • [2].Akcakanat A, Kanda T, Tanabe T, Komukai S, Yajima K, Nakagawa S, Ohashi M,Hatakeyama K, Heterogeneous expression of GAGE, NY-ESO-1, MAGE-A and SSX proteins in esophageal cancer: Implications for immunotherapy, Int. J. Cancer 118 (2006) 123. [DOI] [PubMed] [Google Scholar]
  • [3].Alvarez H, Opalinska J, Zhou L, Sohal D, Fazzari MJ, Yu Y, Montagna C, Montgomery EA, Canto M, Dunbar KB, Wang J, Roa JC, Mo Y, Bhagat T, Ramesh KH, Cannizzaro L, Mollenhauer J, Thompson RF, Suzuki M, Meltzer SJ,Melnick A, Greally JM, Maitra A, Verma A, Widespread hypomethylation occurs early and synergizes with gene amplification during esophageal carcino-genesis, PLoS Genet. 7 (2011) e1001356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [4].Atanackovic D, Hildebrandt Y, Jadczak A, Cao Y, Luetkens T, Meyer S, Kobold S, Bartels K, Pabst C, Lajmi N, Gordic M, Stahl T, Zander AR, Bokemeyer C, Kroger N, Cancer-testis antigens MAGE-C1/CT7 and MAGE-A3 promote the survival of multiple myeloma cells, Haematologica 95 (2010) 785. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [5].Bannister AJ, Kouzarides T, Regulation of chromatin by histone modifications, Cell Res. 21 (2011) 381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [6].Barlesi F, Giaccone G, Gallegos-Ruiz MI, Loundou A, Span SW, Lefesvre P, Kruyt FA, Rodriguez JA, Global histone modifications predict prognosis of resected non small-cell lung cancer, J. Clin. Oncol 25 (2007) 4358. [DOI] [PubMed] [Google Scholar]
  • [7].Barth TK, Imhof A, Fast signals and slow marks: the dynamics of histone modifications, Trends Biochem. Sci 35 (2010) 618. [DOI] [PubMed] [Google Scholar]
  • [8].Baylin SB, Jones PA, A decade of exploring the cancer epigenome — biological and translational implications, Nat. Rev. Cancer 11 (2011) 726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [9].Berdasco M, Esteller M, Aberrant epigenetic landscape in cancer: how cellular identity goes awry, Dev. Cell 19 (2010) 698. [DOI] [PubMed] [Google Scholar]
  • [10].Berndsen CE, Denu JM, Catalysis and substrate selection by histone/protein ly-sine acetyltransferases, Curr. Opin. Struct. Biol 18 (2008) 682. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [11].Bowman RV, Wright CM, Davidson MR, Francis SM, Yang IA, Fong KM, Epigenomic targets for the treatment of respiratory disease, Expert Opin. Ther. Targets 13 (2009) 625. [DOI] [PubMed] [Google Scholar]
  • [12].Bracken AP, Pasini D, Capra M, Prosperini E, Colli E, Helin K, EZH2 is downstream of the pRB-E2F pathway, essential for proliferation and amplified in cancer, EMBO J. 22 (2003) 5323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [13].Bradner JE, West N, Grachan ML, Greenberg EF, Haggarty SJ, Warnow T, Mazitschek R, Chemical phylogenetics of histone deacetylases, Nat. Chem. Biol 6 (2010) 238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [14].Brambilla E, Gazdar A, Pathogenesis of lung cancer signalling pathways: road-map for therapies, Eur. Respir. J 33 (2009) 1485. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [15].Brock MV, Gou M, Akiyama Y, Muller A, Wu TT, Montgomery E, Deasel M, Germonpre P, Rubinson L, Heitmiller RF, Yang SC, Forastiere AA, Baylin SB, Herman JG, Prognostic importance of promoter hypermethylation of multiple genes in esophageal adenocarcinoma, Clin. Cancer Res 9 (2003) 2912. [PubMed] [Google Scholar]
  • [16].Brock MV, Hooker CM, Ota-Machida E, Han Y, Guo M, Ames S, Glockner S, Piantadosi S, Gabrielson E, Pridham G, Pelosky K, Belinsky SA, Yang SC, Baylin SB, Herman JG, DNA methylation markers N. Engl. J. Med 358 (2008) 1118. [DOI] [PubMed] [Google Scholar]
  • [17].Brueckner B, Kuck D, Lyko F, DNA methyltransferase inhibitors for cancer therapy, Cancer J. 13 (2007) 17. [DOI] [PubMed] [Google Scholar]
  • [18].Bruzzese F, Rocco M, Castelli S, Di GE, Desideri A, Budillon A, Synergistic anti-tumor effect between vorinostat and topotecan in small cell lung cancer cells is mediated by generation of reactive oxygen species and DNA damage-induced apoptosis, Mol. Cancer Ther 8 (2009) 3075. [DOI] [PubMed] [Google Scholar]
  • [19].Buckingham L, Penfield FL, Kim A, Liptay M, Barger C, Basu S, Fidler M, Walters K, Bonomi P, Coon J, PTEN, RASSF1 and DAPK site-specific hypermethylation and outcome in surgically treated stage I and II nonsmall cell lung cancer patients, Int. J. Cancer 126 (2010) 1630. [DOI] [PubMed] [Google Scholar]
  • [20].Caballero OL, Chen YT, Cancer/testis (CT) antigens: potential targets for immunotherapy, Cancer Sci. 100 (2009) 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [21].Campbell PM, Bovenzi V, Szyf M, Methylated DNA-binding protein 2 antisense inhibitors suppress tumourigenesis of human cancer cell lines in vitro and in vivo, Carcinogenesis 25 (2004) 499. [DOI] [PubMed] [Google Scholar]
  • [22].Carrasco J, Van PA, Neyns B, Lethe B, Brasseur F, Renkvist N, van der Bruggen P, van BN, Paulus R, Thielemans K, Boon T, Godelaine D, Vaccination of a melanoma patient with mature dendritic cells pulsed with MAGE-3 peptides triggers the activity of nonvaccine anti-tumor cells, J. Immunol 180 (2008) 3585. [DOI] [PubMed] [Google Scholar]
  • [23].Chai G, Li L, Zhou W, Wu L, Zhao Y, Wang D, Lu S, Yu Y, Wang H, McNutt MA, Hu YG, Chen Y, Yang Y, Wu X, Otterson GA, Zhu WG, HDAC inhibitors act with 5-aza-2′-deoxycytidine to inhibit cell proliferation by suppressing removal of incorporated abases in lung cancer cells, PLoS One 3 (2008) e2445. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [24].Chen C, Zhao M, Yin N, He B, Wang B, Yuan Y, Yu F, Hu J, Yin B, Lu Q, Abnormal histone acetylation and methylation levels in esophageal squamous cell carcinomas, Cancer Investig. 29 (2011) 548. [DOI] [PubMed] [Google Scholar]
  • [25].Chen MW, Hua KT, Kao HJ, Chi CC, Wei LH, Johansson G, Shiah SG, Chen PS, Jeng YM, Cheng TY, Lai TC, Chang JS, Jan YH, Chien MH, Yang CJ, Huang MS, Hsiao M, Kuo ML, H3K9 histone methyltransferase G9a promotes lung cancer invasion and metastasis by silencing the cell adhesion molecule Ep-CAM, Cancer Res. 70 (2010) 7830. [DOI] [PubMed] [Google Scholar]
  • [26].Chen YT, Hsu M, Lee P, Shin SJ, Mhawech-Fauceglia P, Odunsi K, Altorki NK, Song CJ, Jin BQ, Simpson AJ, Old LJ, Cancer/testis antigen CT45: analysis of mRNA and protein expression in human cancer, Int. J. Cancer 124 (2009) 2893. [DOI] [PubMed] [Google Scholar]
  • [27].Chen Z, Zhu B, Wu Y, Expression of TRAG-3 antigen in non-small-cell lung carcinomas, Lung Cancer 38 (2002) 101. [DOI] [PubMed] [Google Scholar]
  • [28].Chinnasamy N, Wargo JA, Yu Z, Rao M, Frankel TL, Riley JP, Hong JJ, Parkhurst MR, Feldman SA, Schrump DS, Restifo NP, Robbins PF, Rosenberg SA, Morgan RA, A TCR targeting the HLA-A*0201-restricted epitope of MAGE-A3 recognizes multiple epitopes of the MAGE-A antigen superfamily in several types of cancer, J. Immunol 186 (2011) 685. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [29].Choi SH, Heo K, Byun HM, An W, Lu W, Yang AS, Identification of preferential target sites for human DNA methyltransferases, Nucleic Acids Res. 39 (2011) 104–118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [30].Choudhary C, Kumar C, Gnad F, Nielsen ML, Rehman M, Walther TC, Olsen JV, Mann M, Lysine acetylation targets protein complexes and co-regulates major cellular functions, Science 325 (2009) 834. [DOI] [PubMed] [Google Scholar]
  • [31].Christensen BC, Godleski JJ, Marsit CJ, Houseman EA, Lopez-Fagundo CY, Longacker JL, Bueno R, Sugarbaker DJ, Nelson HH, Kelsey KT, Asbestos exposure predicts cell cycle control gene promoter methylation in pleural mesothelioma, Carcinogenesis 29 (2008) 1555. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [32].Christensen BC, Houseman EA, Poage GM, Godleski JJ, Bueno R, Sugarbaker DJ, Wiencke JK, Nelson HH, Marsit CJ, Kelsey KT, Integrated profiling reveals a global correlation between epigenetic and genetic alterations in mesothelioma, Cancer Res. 70 (2010) 5686. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [33].Christophersen NS, Helin K, Epigenetic control of embryonic stem cell fate,J. Exp. Med 207 (2010) 2287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [34].Cilensek ZM, Yehiely F, Kular RK, Deiss LP, A member of the GAGE family of tumor antigens is an anti-apoptotic gene that confers resistance to Fas/CD95/APO-1, interferon-gamma, taxol and gamma-irradiation, Cancer Biol. Ther 1 (2002) 380. [PubMed] [Google Scholar]
  • [35].Cloos PA, Christensen J, Agger K, Helin K, Erasing the methyl mark: histone demethylases at the center of cellular differentiation and disease, Genes Dev. 22 (2008) 1115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [36].Coe BP, Lockwood WW, Girard L, Chari R, Macaulay C, Lam S, Gazdar AF, Minna JD, Lam WL, Differential disruption of cell cycle pathways in small cell and non-small cell lung cancer, Br. J. Cancer 94 (2006) 1927. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [37].Dahl C, Gronbaek K, Guldberg P, Advances in DNA methylation: 5-hydroxymethylcytosine revisited, Clin. Chim. Acta 412 (2011) 831. [DOI] [PubMed] [Google Scholar]
  • [38].Damiani LA, Yingling CM, Leng S, Romo PE, Nakamura J, Belinsky SA, Carcinogen-induced gene promoter hypermethylation is mediated by DNMT1 and causal for transformation of immortalized bronchial epithelial cells, Cancer Res. 68 (2008) 9005. [DOI] [PubMed] [Google Scholar]
  • [39].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 115 (2005) 351. [DOI] [PubMed] [Google Scholar]
  • [40].De Carvalho DD, You JS, Jones PA, DNA methylation and cellular reprogramming, Trends Cell Biol. 20 (2010) 609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [41].Destro A, Ceresoli GL, Baryshnikova E, Garassino I, Zucali PA, De VF, Bianchi P, Morenghi E, Testori A, Alloisio M, Santoro A, Roncalli M, Gene methylation in pleural mesothelioma: correlations with clinico-pathological features and patient’s follow-up, Lung Cancer 59 (2008) 369. [DOI] [PubMed] [Google Scholar]
  • [42].Dose A, Liokatis S, Theillet FX, Selenko P, Schwarzer D, NMR profiling of his-tone deacetylase and acetyl-transferase activities in real time, ACS Chem. Biol 6 (2011) 419. [DOI] [PubMed] [Google Scholar]
  • [43].Du Z, Song J, Wang Y, Zhao Y, Guda K, Yang S, Kao HY, Xu Y, Willis J, Markowitz SD, Sedwick D, Ewing RM, Wang Z, DNMT1 stability is regulated by proteins coordinating deubiquitination and acetylation-driven ubiquitination, Sci. Signal 3 (2010) ra80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [44].Egger G, Aparicio AM, Escobar SG, Jones PA, Inhibition of histone deacetylation does not block resilencing of p16 after 5-aza-2′-deoxycytidine treatment, Cancer Res. 67 (2007) 346. [DOI] [PubMed] [Google Scholar]
  • [45].Ehrlich M, DNA hypomethylation in cancer cells, Epigenomics 1 (2009) 239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [46].Eskeland R, Leeb M, Grimes GR, Kress C, Boyle S, Sproul D, Gilbert N, Fan Y, Skoultchi AI, Wutz A, Bickmore WA, Ring1B compacts chromatin structure and represses gene expression independent of histone ubiquitination, Mol. Cell 38 (2010) 452. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [47].Fischer JR, Ohnmacht U, Rieger N, Zemaitis M, Stoffregen C, Kostrzewa M, Buchholz E, Manegold C, Lahm H, Promoter methylation of RASSF1A, RARbeta and DAPK predict poor prognosis of patients with malignant mesothelioma, Lung Cancer 54 (2006) 109. [DOI] [PubMed] [Google Scholar]
  • [48].Flores RM, Zakowski M, Venkatraman E, Krug L, Rosenzweig K, Dycoco J, Lee C, Yeoh C, Bains M, Rusch V, Prognostic factors in the treatment of malignant pleural mesothelioma at a large tertiary referral center, J. Thorac. Oncol 2 (2007) 957. [DOI] [PubMed] [Google Scholar]
  • [49].Friedman JM, Liang G, Liu CC, Wolff EM, Tsai YC, Ye W, Zhou X, Jones PA, The putative tumor suppressor microRNA-101 modulates the cancer epigenome by repressing the polycomb group protein EZH2, Cancer Res. 69 (6) (2009) 2623. [DOI] [PubMed] [Google Scholar]
  • [50].Ge K, Epigenetic regulation of adipogenesis by histone methylation, Biochim. Biophys. Acta (January 4 2012) [Electronic publication ahead of print]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [51].Goll MG, Bestor TH, Eukaryotic cytosine methyltransferases, Annu. Rev. Biochem 74 (2005) 481. [DOI] [PubMed] [Google Scholar]
  • [52].Groeper C, Gambazzi F, Zajac P, Bubendorf L, Adamina M, Rosenthal R, Zerkowski HR, Heberer M, Spagnoli GC, Cancer/testis antigen expression and specific cytotoxic T lymphocyte responses in non small cell lung cancer, Int. J. Cancer 120 (2007) 337. [DOI] [PubMed] [Google Scholar]
  • [53].Guo ZS, Hong JA, Irvine KR, Chen GA, Spiess PJ, Liu Y, Zeng G, Wunderlich JR, Nguyen DM, Restifo NP, Schrump DS, De novo induction of a cancer/testis antigen by 5-aza-2′-deoxycytidine augments adoptive immunotherapy in a murine tumor model, Cancer Res. 66 (2006) 1105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [54].Gure AO, Chua R, Williamson B, Gonen M, Ferrera CA, Gnjatic S, Ritter G, Simpson AJ, Chen YT, Old LJ, Altorki NK, Cancer-testis genes are coordinately expressed and are markers of poor outcome in non-small cell lung cancer, Clin. Cancer Res 11 (2005) 8055. [DOI] [PubMed] [Google Scholar]
  • [55].Ha K, Lee GE, Palii SS, Brown KD, Takeda Y, Liu K, Bhalla KN, Robertson KD, Rapid and transient recruitment of DNMT1 to DNA double-strand breaks is mediated by its interaction with multiple components of the DNA damage response machinery, Hum. Mol. Genet 20 (2011) 126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [56].Hayami S, Kelly JD, Cho HS, Yoshimatsu M, Unoki M, Tsunoda T, Field HI, Neal DE, Yamaue H, Ponder BA, Nakamura Y, Hamamoto R, Overexpression of LSD1 contributes to human carcinogenesis through chromatin regulation in various cancers, Int. J. Cancer 128 (2011) 574. [DOI] [PubMed] [Google Scholar]
  • [57].Hayami S, Yoshimatsu M, Veerakumarasivam A, Unoki M, Iwai Y, Tsunoda T, Field HI, Kelly JD, Neal DE, Yamaue H, Ponder BA, Nakamura Y, Hamamoto R, Overexpression of the JmjC histone demethylase KDM5B in human carcinogenesis: involvement in the proliferation of cancer cells through the E2F/RB pathway, Mol. Cancer 9 (2010) 59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [58].Hayashi M, Tokuchi Y, Hashimoto T, Hayashi S, Nishida K, Ishikawa Y, Nakagawa K, Tsuchiya S, Okumura S, Tsuchiya E, Reduced HIC-1 gene expression in non-small cell lung cancer and its clinical significance, Anticancer. Res 21 (2001) 535. [PubMed] [Google Scholar]
  • [59].He LR, Liu MZ, Li BK, Jia WH, Zhang Y, Liao YJ, Chen YC, Zhang LJ, Guan XY, Zeng YX, Kung HF, Xie D, High expression of EZH2 is associated with tumor aggressiveness and poor prognosis in patients with esophageal squamous cell carcinoma treated with definitive chemoradiotherapy, Int. J. Cancer 127 (1) (2010) 138. [DOI] [PubMed] [Google Scholar]
  • [60].He LR, Liu MZ, Li BK, Rao HL, Liao YJ, Guan XY, Zeng YX, Xie D, Prognostic impact of H3K27me3 expression on locoregional progression after chemoradiotherapy in esophageal squamous cell carcinoma, BMC Cancer 9 (2009) 461. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [61].He XT, Cao XF, Ji L, Zhu B, Lv J, Wang DD, Lu PH, Cui HG, Association between Bmi1 and clinicopathological status of esophageal squamous cell carcinoma, World J. Gastroenterol 15 (2009) 2389. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [62].He Y, Korboukh I, Jin J, Huang J, Targeting protein lysine methylation and demethylation in cancers, Acta Biochim. Biophys. Sin. (Shanghai) 44 (2012) 70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [63].Hernandez DG, Nalls MA, Gibbs JR, Arepalli S, van der Brug M, Chong S, Moore M, Longo DL, Cookson M, Traynor BJ, Singleton A, Distinct DNA methylation changes highly correlated with chronological age in the human brain, Hum. Mol. Genet (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [64].Hibi K, Nakamura H, Hirai A, Fujikake Y, Kasai Y, Akiyama S, Ito K, Takagi H, Loss of H19 imprinting in esophageal cancer, Cancer Res. 56 (1996) 480. [PubMed] [Google Scholar]
  • [65].Hirasawa R, Feil R, Genomic imprinting and human disease, Essays Biochem. 48 (2010) 187. [DOI] [PubMed] [Google Scholar]
  • [66].Holm TM, Jackson-Grusby L, Brambrink T, Yamada Y, Rideout III WM, Jaenisch R, Global loss of imprinting leads to widespread tumorigenesis in adult mice, Cancer Cell 8 (2005) 275. [DOI] [PubMed] [Google Scholar]
  • [67].Hong JA, Kang Y, Abdullaev Z, Flanagan PT, Pack SD, Fischette MR, Adnani MT, Loukinov DI, Vatolin S, Risinger JI, Custer M, Chen GA, Zhao M, Nguyen DM, Barrett JC, Lobanenkov VV, Schrump DS, Reciprocal binding of CTCF and BORIS to the NY-ESO-1 promoter coincides with derepression of this cancer-testis gene in lung cancer cells, Cancer Res. 65 (2005) 7763. [DOI] [PubMed] [Google Scholar]
  • [68].Howard G, Eiges R, Gaudet F, Jaenisch R, Eden A, Activation and transposition of endogenous retroviral elements in hypomethylation induced tumors in mice, Oncogene 27 (2008) 404. [DOI] [PubMed] [Google Scholar]
  • [69].Hudlebusch HR, Santoni-Rugiu E, Simon R, Ralfkiaer E, Rossing HH, Johansen JV, Jorgensen M, Sauter G, Helin K, The histone methyltransferase and putative oncoprotein MMSET is overexpressed in a large variety of human tumors, Clin. Cancer Res 17 (2011) 2919. [DOI] [PubMed] [Google Scholar]
  • [70].Hussain M, Rao M, Humphries AE, Hong JA, Liu F, Yang M, Caragacianu D, Schrump DS, Tobacco smoke induces polycomb-mediated repression of Dickkopf-1 in lung cancer cells, Cancer Res. 69 (2009) 3570. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [71].I K, Ko E, Kim Y, Cho EY, Han J, Park J, Kim K, Kim DH, Shim YM, Association of global levels of histone modifications with recurrence-free survival in stage IIB and III esophageal squamous cell carcinomas, Cancer Epidemiol. Biomarkers Prev. 19 (2010) 566. [DOI] [PubMed] [Google Scholar]
  • [72].Iliou MS, Lujambio A, Portela A, Brustle O, Koch P, Andersson-Vincent PH, Sundstrom E, Hovatta O, Esteller M, Bivalent histone modifications in stem cells poise miRNA loci for CpG island hypermethylation in human cancer, Epigenetics 6 (2011). [DOI] [PubMed] [Google Scholar]
  • [73].Ivanov SV, Miller J, Lucito R, Tang C, Ivanova AV, Pei J, Carbone M, Cruz C, Beck A, Webb C, Nonaka D, Testa JR, Pass HI, Genomic events associated with progression of pleural malignant mesothelioma, Int. J. Cancer 124 (2009) 589. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [74].Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D, Global cancer statistics CA Cancer J. Clin 61 (2011) 69. [DOI] [PubMed] [Google Scholar]
  • [75].Jin SG, Jiang Y, Qiu R, Rauch TA, Wang Y, Schackert G, Krex D, Lu Q, Pfeifer GP, 5-Hydroxymethylcytosine Is strongly depleted in human cancers but its levels do not correlate with IDH1 mutations, Cancer Res. 71 (2011) 7360–7365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [76].Jones PA, Baylin SB, The epigenomics of cancer, Cell 128 (2007) 683. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [77].Karbach J, Gnjatic S, Bender A, Neumann A, Weidmann E, Yuan J, Ferrara CA,Hoffmann E, Old LJ, Altorki NK, Jager E, Tumor-reactive CD8+ T-cell responses after vaccination with NY-ESO-1 peptide, CpG 7909 and Montanide ISA-51: association with survival, Int. J. Cancer 126 (2010) 909. [DOI] [PubMed] [Google Scholar]
  • [78].Kassambara A, Klein B, Moreaux J, MMSET is overexpressed in cancers: link with tumor aggressiveness, Biochem. Biophys. Res. Commun 379 (2009) 840. [DOI] [PubMed] [Google Scholar]
  • [79].Kassis ES, Zhao M, Hong JA, Chen GA, Nguyen DM, Schrump DS, Depletion of DNA methyltransferase 1 and/or DNA methyltransferase 3b mediates growth arrest and apoptosis in lung and esophageal cancer and malignant pleural mesothelioma cells, J. Thorac. Cardiovasc. Surg 131 (2006) 298. [DOI] [PubMed] [Google Scholar]
  • [80].Kemp CD, Rao M, Xi S, Inchauste S, Mani H, Fetsch P, Filie AC, Zhang M, Hong JA, Walker R, Zhu YJ, Ripley RT, Mathur A, Liu F, Yang M, Meltzer PS, Marquez VE, De RA, Bueno R, Schrump DS, Polycomb repressor complex-2 is a novel target for mesothelioma therapy, Clin. Cancer Res 18 (2012) 77–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [81].Kikuchi J, Kinoshita I, Shimizu Y, Kikuchi E, Konishi J, Oizumi S, Kaga K, Matsuno Y, Nishimura M, Dosaka-Akita H, Distinctive expression of the poly-comb group proteins Bmi1 polycomb ring finger oncogene and enhancer of zeste homolog 2 in nonsmall cell lung cancers and their clinical and clinicopathologic significance, Cancer 116 (2010) 3015. [DOI] [PubMed] [Google Scholar]
  • [82].Kim H, Kwon YM, Kim JS, Han J, Shim YM, Park J, Kim DH, Elevated mRNA levels of DNA methyltransferase-1 as an independent prognostic factor in primary nonsmall cell lung cancer, Cancer 107 (2006) 1042. [DOI] [PubMed] [Google Scholar]
  • [83].Kim JS, Kim JW, Han J, Shim YM, Park J, Kim DH, Cohypermethylation of p16 and FHIT promoters as a prognostic factor of recurrence in surgically resected stage I non-small cell lung cancer, Cancer Res. 66 (2006) 4049. [DOI] [PubMed] [Google Scholar]
  • [84].Kim SH, Lee S, Lee CH, Lee MK, Kim YD, Shin DH, Choi KU, Kim JY, Park d.Y., Sol MY, Expression of cancer-testis antigens MAGE-A3/6 and NY-ESO-1 in non-small-cell lung carcinomas and their relationship with immune cell infiltration, Lung 187 (2009) 401. [DOI] [PubMed] [Google Scholar]
  • [85].Kim W, Kim R, Park G, Park JW, Kim JE, Deficiency of H3K79 histone methyltransferase Dot1-like protein (DOT1L) inhibits cell proliferation, J. Biol. Chem 287 (2012) 5588. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [86].Kim Y, Park H, Park D, Lee YS, Choe J, Hahn JH, Lee H, Kim YM, Jeoung D, Cancer/testis antigen CAGE exerts negative regulation on p53 expression through HDAC2 and confers resistance to anti-cancer drugs, J. Biol. Chem 285 (2010) 25957. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [87].Kim YD, Park HR, Song MH, Shin DH, Lee CH, Lee MK, Lee SY, Pattern of cancer/testis antigen expression in lung cancer patients, Int. J. Mol. Med 29 (2012) 656. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [88].Kohda M, Hoshiya H, Katoh M, Tanaka I, Masuda R, Takemura T, Fujiwara M,Oshimura M, Frequent loss of imprinting of IGF2 and MEST in lung adenocarcinoma, Mol. Carcinog 31 (2001) 184. [DOI] [PubMed] [Google Scholar]
  • [89].Kondo M, Matsuoka S, Uchida K, Osada H, Nagatake M, Takagi K, Harper JW,Takahashi T, Elledge SJ, Takahashi T, Selective maternal-allele loss in human lung cancers of the maternally expressed p57KIP2 gene at 11p15.5, Oncogene 12 (1996) 1365. [PubMed] [Google Scholar]
  • [90].Kondo M, Suzuki H, Ueda R, Osada H, Takagi K, Takahashi T, Takahashi T, Frequent loss of imprinting of the H19 gene is often associated with its overexpression in human lung cancers, Oncogene 10 (1995) 1193. [PubMed] [Google Scholar]
  • [91].Lam LT, Pickeral OK, Peng AC, Rosenwald A, Hurt EM, Giltnane JM, Averett LM, Zhao H, Davis RE, Sathyamoorthy M, Wahl LM, Harris ED, Mikovits JA, Monks AP, Hollingshead MG, Sausville EA, Staudt LM, Genomic-scale measurement of mRNA turnover and the mechanisms of action of the anti-cancer drug flavopiridol, Genome Biol. 2 (2001) RESEARCH0041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [92].Langer R, Mutze K, Becker K, Feith M, Ott K, Hofler H, Keller G, Expression of class I histone deacetylases (HDAC1 and HDAC2) in oesophageal adenocarcinomas: an immunohistochemical study, J. Clin. Pathol 63 (2010) 994. [DOI] [PubMed] [Google Scholar]
  • [93].Lee EJ, Lee BB, Han J, Cho EY, Shim YM, Park J, Kim DH, CpG island hyper-methylation of E-cadherin (CDH1) and integrin alpha4 is associated with recurrence of early stage esophageal squamous cell carcinoma, Int. J. Cancer 123 (2008) 2073. [DOI] [PubMed] [Google Scholar]
  • [94].Lee JS, Smith E, Shilatifard A, The language of histone crosstalk, Cell 142 (2010) 682. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [95].Li B, Wang B, Niu LJ, Jiang L, Qiu CC, Hypermethylation of multiple tumor-related genes associated with DNMT3b up-regulation served as a biomarker for early diagnosis of esophageal squamous cell carcinoma, Epigenetics 6 (2011) 307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [96].Lin RK, Hsieh YS, Lin P, Hsu HS, Chen CY, Tang YA, Lee CF, Wang YC, The tobacco-specific carcinogen NNK induces DNA methyltransferase 1 accumulation and tumor suppressor gene hypermethylation in mice and lung cancer patients, J. Clin. Invest 120 (2010) 521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [97].Lin RK, Hsu CH, Wang YC, Mithramycin A inhibits DNA methyltransferase and metastasis potential of lung cancer cells, Anticancer Drugs 18 (2007) 1157. [DOI] [PubMed] [Google Scholar]
  • [98].Lin RK, Hsu HS, Chang JW, Chen CY, Chen JT, Wang YC, Alteration of DNA methyltransferases contributes to 5′CpG methylation and poor prognosis in lung cancer, Lung Cancer 55 (2007) 205. [DOI] [PubMed] [Google Scholar]
  • [99].Ling Y, Huang G, Fan L, Wei L, Zhu J, Liu Y, Zhu C, Zhang C, CpG island methylator phenotype of cell-cycle regulators associated with TNM stage and poor prognosis in patients with oesophageal squamous cell carcinoma, J. Clin. Pathol 64 (2011) 246. [DOI] [PubMed] [Google Scholar]
  • [100].Liu F, Killian JK, Yang M, Walker RL, Hong JA, Zhang M, Davis S, Zhang Y, Hussain M, Xi S, Rao M, Meltzer PA, Schrump DS, Epigenomic alterations and gene expression profiles in respiratory epithelia exposed to cigarette smoke condensate, Oncogene 29 (2010) 3650. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [101].Liu WL, Guo XZ, Zhang LJ, Wang JY, Zhang G, Guan S, Chen YM, Kong QL,Xu LH, Li MZ, Song LB, Zeng MS, Prognostic relevance of Bmi-1 expression and autoantibodies in esophageal squamous cell carcinoma, BMC Cancer 10 (2010) 467. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [102].Loriot A, Parvizi GK, Reister S, De SC, Silencing of cancer-germline genes in human preimplantation embryos: evidence for active de novo DNA methylation in stem cells, Biochem. Biophys. Res. Commun 417 (2012) 187–191. [DOI] [PubMed] [Google Scholar]
  • [103].Lu C, Soria JC, Tang X, Xu XC, Wang L, Mao L, Lotan R, Kemp B, Bekele BN, Feng L, Hong WK, Khuri FR, Prognostic factors in resected stage I non-small-cell lung cancer: a multivariate analysis of six molecular markers, J. Clin. Oncol 22 (2004) 4575. [DOI] [PubMed] [Google Scholar]
  • [104].Lu J, He ML, Wang L, Chen Y, Liu X, Dong Q, Chen YC, Peng Y, Yao KT, Kung HF, Li XP, MiR-26a inhibits cell growth and tumorigenesis of nasopharyngeal carcinoma through repression of EZH2, Cancer Res. 71 (1) (2011) 225. [DOI] [PubMed] [Google Scholar]
  • [105].Luszczek W, Cheriyath V, Mekhail TM, Borden EC, Combinations of DNA methyltransferase and histone deacetylase inhibitors induce DNA damage in small cell lung cancer cells: correlation of resistance with IFN-stimulated gene expression, Mol. Cancer Ther 9 (2010) 2309. [DOI] [PubMed] [Google Scholar]
  • [106].Marcar L, Maclaine NJ, Hupp TR, Meek DW, Mage-A cancer/testis antigens inhibit p53 function by blocking its interaction with chromatin, Cancer Res. 70 (2010) 10362. [DOI] [PubMed] [Google Scholar]
  • [107].McGarvey KM, Fahrner JA, Greene E, Martens J, Jenuwein T, Baylin SB, Silenced tumor suppressor genes reactivated by DNA demethylation do not return to a fully euchromatic chromatin state, Cancer Res. 66 (2006) 3541. [DOI] [PubMed] [Google Scholar]
  • [108].Melloni G, Ferreri AJ, Russo V, Gattinoni L, Arrigoni G, Ceresoli GL, Zannini P,Traversari C, Prognostic significance of cancer-testis gene expression in resected non-small cell lung cancer patients, Oncol. Rep 12 (2004) 145. [PubMed] [Google Scholar]
  • [109].Mills AA, Throwing the cancer switch: reciprocal roles of polycomb and trithorax proteins, Nat. Rev. Cancer 10 (2010) 669. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [110].Minamiya Y, Ono T, Saito H, Takahashi N, Ito M, Motoyama S, Ogawa J, Strong expression of HDAC3 correlates with a poor prognosis in patients with adeno-carcinoma of the lung, Tumour Biol. 31 (2010) 533. [DOI] [PubMed] [Google Scholar]
  • [111].Minamiya Y, Ono T, Saito H, Takahashi N, Ito M, Motoyama S, Ogawa J, Strong expression of HDAC3 correlates with a poor prognosis in patients with adeno-carcinoma of the lung, Tumour Biol. 31 (2010) 533. [DOI] [PubMed] [Google Scholar]
  • [112].Momparler RL, Bouffard DY, Momparler LF, Dionne J, Belanger K, Ayoub J, Pilot phase I-II study on 5-aza-2′-deoxycytidine (Decitabine) in patients with metastatic lung cancer, Anticancer Drugs 8 (1997) 358. [DOI] [PubMed] [Google Scholar]
  • [113].Monte M, Simonatto M, Peche LY, Bublik DR, Gobessi S, Pierotti MA, Rodolfo M, Schneider C, MAGE-A tumor antigens target p53 transactivation function through histone deacetylase recruitment and confer resistance to chemo-therapeutic agents, Proc. Natl. Acad. Sci. U. S. A 103 (2006) 11160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [114].Morey L, Helin K, Polycomb group protein-mediated repression of transcription, Trends Biochem. Sci 35 (2010) 323. [DOI] [PubMed] [Google Scholar]
  • [115].Nakagawa K, Noguchi Y, Uenaka A, Sato S, Okumura H, Tanaka M, Shimono M, Ali Eldib AM, Ono T, Ohara N, Yoshino T, Yamashita K, Tsunoda T,Aoe M, Shimizu N, Nakayama E, XAGE-1 expression in non-small cell lung cancer and antibody response in patients, Clin. Cancer Res 11 (2005) 5496. [DOI] [PubMed] [Google Scholar]
  • [116].Nakagawa M, Oda Y, Eguchi T, Aishima S, Yao T, Hosoi F, Basaki Y, Ono M, Kuwano M, Tanaka M, Tsuneyoshi M, Expression profile of class I histone deacetylases in human cancer tissues, Oncol. Rep 18 (2007) 769. [PubMed] [Google Scholar]
  • [117].Nakata S, Sugio K, Uramoto H, Oyama T, Hanagiri T, Morita M, Yasumoto K, The methylation status and protein expression of CDH1, p16(INK4A), and fragile histidine triad in nonsmall cell lung carcinoma: epigenetic silencing, clinical features, and prognostic significance, Cancer 106 (2006) 2190. [DOI] [PubMed] [Google Scholar]
  • [118].Nguyen DM, Schrump WD, Chen GA, Tsai W, Nguyen P, Trepel JB, Schrump DS, Abrogation of p21 expression by flavopiridol enhances depsipeptide-mediated apoptosis in malignant pleural mesothelioma cells, Clin. Cancer Res 10 (2004) 1813. [DOI] [PubMed] [Google Scholar]
  • [119].Nottke A, Colaiacovo MP, Shi Y, Developmental roles of the histone lysine demethylases, Development 136 (2009) 879. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [120].Oka M, Meacham AM, Hamazaki T, Rodic N, Chang LJ, Terada N, De novo DNA methyltransferases Dnmt3a and Dnmt3b primarily mediate the cytotoxic effect of 5-aza-2′-deoxycytidine, Oncogene 24 (2005) 3091. [DOI] [PubMed] [Google Scholar]
  • [121].Ono T, Kurashige T, Harada N, Noguchi Y, Saika T, Niikawa N, Aoe M, Nakamura S, Higashi T, Hiraki A, Wada H, Kumon H, Old LJ, Nakayama E, Identification of proacrosin binding protein sp32 precursor as a human cancer/testis antigen, Proc. Natl. Acad. Sci. U. S. A 98 (2001) 3282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [122].Osada H, Tatematsu Y, Saito H, Yatabe Y, Mitsudomi T, Takahashi T, Reduced expression of class II histone deacetylase genes is associated with poor prognosis in lung cancer patients, Int. J. Cancer 112 (2004) 26. [DOI] [PubMed] [Google Scholar]
  • [123].Otterson GA, Hodgson L, Pang H, Vokes EE, Phase II study of the histone deacetylase inhibitor Romidepsin in relapsed small cell lung cancer (Cancer and Leukemia Group B 30304), J. Thorac. Oncol 5 (2010) 1644. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [124].Palii SS, Van Emburgh BO, Sankpal UT, Brown KD, Robertson KD, DNA methylation inhibitor 5-aza-2′-deoxycytidine induces reversible genome-wide DNA damage that is distinctly influenced by DNA methyltransferases 1 and 3B, Mol. Cell. Biol 28 (2008) 752. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [125].Parmigiani RB, Bettoni F, Vibranovski MD, Lopes MH, Martins WK, Cunha IW, Soares FA, Simpson AJ, de Souza SJ, Camargo AA, Characterization of a cancer/testis (CT) antigen gene family capable of eliciting humoral response in cancer patients, Proc. Natl. Acad. Sci. U. S. A 103 (2006) 18066. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [126].Por E, Byun HJ, Lee EJ, Lim JH, Jung SY, Park I, Kim YM, Jeoung DI, Lee H, The cancer/testis antigen CAGE with oncogenic potential stimulates cell proliferation by up-regulating cyclins D1 and E in an AP-1- and E2F-dependent manner, J. Biol. Chem 285 (2010) 14475. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [127].Rai K, Sarkar S, Broadbent TJ, Voas M, Grossmann KF, Nadauld LD, Dehghanizadeh S, Hagos FT, Li Y, Toth RK, Chidester S, Bahr TM, Johnson WE, Sklow B, Burt R, Cairns BR, Jones DA, DNA demethylase activity maintains intestinal cells in an undifferentiated state following loss of APC, Cell 142 (2010) 930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [128].Ramalingam SS, Maitland ML, Frankel P, Argiris AE, Koczywas M, Gitlitz B,Thomas S, Espinoza-Delgado I, Vokes EE, Gandara DR, Belani CP, Carboplatin and paclitaxel in combination with either vorinostat or placebo for first-line therapy of advanced non-small-cell lung cancer, J. Clin. Oncol 28 (2010)56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [129].Rao M, Chinnasamy N, Hong JA, Zhang Y, Zhang M, Xi S, Liu F, Marquez VE, Morgan RA, Schrump DS, Inhibition of histone lysine methylation enhances cancer-testis antigen expression in lung cancer cells: implications for adoptive immunotherapy of cancer, Cancer Res. 71 (2011) 4192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [130].Renaud S, Loukinov D, Alberti L, Vostrov A, Kwon YW, Bosman FT, Lobanenkov V, Benhattar J, BORIS/CTCFL-mediated transcriptional regulation of the hTERT telomerase gene in testicular and ovarian tumor cells, Nucleic Acids Res. 39 (2011) 862–873. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [131].Reynolds PA, Sigaroudinia M, Zardo G, Wilson MB, Benton GM, Miller CJ, Hong C, Fridlyand J, Costello JF, Tlsty TD, Tumor suppressor p16INK4A regulates polycomb-mediated DNA hypermethylation in human mammary epithelial cells, J. Biol. Chem 281 (2006) 24790. [DOI] [PubMed] [Google Scholar]
  • [132].Rizzo S, Hersey JM, Mellor P, Dai W, Santos-Silva A, Liber D, Luk L, Titley I, Carden CP, Box G, Hudson DL, Kaye SB, Brown R, Ovarian cancer stem cell-like side populations are enriched following chemotherapy and overexpress EZH2, Mol. Cancer Ther 10 (2011) 325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [133].Robbins PF, Morgan RA, Feldman SA, Yang JC, Sherry RM, Dudley ME, Wunderlich JR, Nahvi AV, Helman LJ, Mackall CL, Kammula US, Hughes MS, Restifo NP, Raffeld M, Lee CC, Levy CL, Li YF, El-Gamil M, Schwarz SL, Laurencot C, Rosenberg SA, Tumor regression in patients with metastatic synovial cell sarcoma and melanoma using genetically engineered lymphocytes reactive with NY-ESO-1, J. Clin. Oncol 29 (2011) 917. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [134].Sasaki H, Moriyama S, Nakashima Y, Kobayashi Y, Kiriyama M, Fukai I, Yamakawa Y, Fujii Y, Histone deacetylase 1 mRNA expression in lung cancer, Lung Cancer 46 (2004) 171. [DOI] [PubMed] [Google Scholar]
  • [135].Sato M, Shames DS, Gazdar AF, Minna JD, A translational view of the molecular pathogenesis of lung cancer, J. Thorac. Oncol 2 (2007) 327. [DOI] [PubMed] [Google Scholar]
  • [136].Sauvageau M, Sauvageau G, Polycomb group proteins: multi-faceted regulators of somatic stem cells and cancer, Cell Stem Cell 7 (2010) 299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [137].Sawan C, Herceg Z, Histone modifications and cancer, Adv. Genet 70 (2010) 57. [DOI] [PubMed] [Google Scholar]
  • [138].Sawarkar R, Paro R, Interpretation of developmental signaling at chromatin: the Polycomb perspective, Dev. Cell 19 (2010) 651. [DOI] [PubMed] [Google Scholar]
  • [139].Schar P, Fritsch O, DNA repair and the control of DNA methylation, Prog. Drug Res 67 (2011) 51. [DOI] [PubMed] [Google Scholar]
  • [140].Scharf AN, Imhof A, Every methyl counts — epigenetic calculus, FEBS Lett. 585 (2011) 2001–2007. [DOI] [PubMed] [Google Scholar]
  • [141].Schrump DS, Cytotoxicity mediated by histone deacetylase inhibitors in cancer cells: mechanisms and potential clinical implications, Clin. Cancer Res 15 (2009) 3947. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [142].Schrump DS, Fischette MR, Nguyen DM, Zhao M, Li X, Kunst TF, Hancox A, Hong JA, Chen GA, Kruchin E, Wright JJ, Rosing DR, Sparreboom A, Figg WD, Steinberg SM, Clinical and molecular responses in lung cancer patients receiving Romidepsin, Clin. Cancer Res 14 (2008) 188. [DOI] [PubMed] [Google Scholar]
  • [143].Schrump DS, Fischette MR, Nguyen DM, Zhao M, Li X, Kunst TF, Hancox A, Hong JA, Chen GA, Pishchik V, Figg WD, Murgo AJ, Steinberg SM, Phase I study of decitabine-mediated gene expression in patients with cancers involving the lungs, esophagus, or pleura, Clin. Cancer Res 12 (2006) 5777. [DOI] [PubMed] [Google Scholar]
  • [144].Schrump DS, Hong JA, Nguyen DM, Utilization of chromatin remodeling agents for lung cancer therapy, Cancer J. 13 (2007) 56. [DOI] [PubMed] [Google Scholar]
  • [145].Schrump DS, Nguyen DM, Targeting the epigenome for the treatment and prevention of lung cancer, Semin. Oncol 32 (2005) 488. [DOI] [PubMed] [Google Scholar]
  • [146].Schultz-Thater E, Piscuoglio S, Iezzi G, Le MC, Zajac P, Carafa V, Terracciano L,Tornillo L, Spagnoli GC, MAGE-A10 is a nuclear protein frequently expressed in high percentages of tumor cells in lung, skin and urothelial malignancies, Int. J. Cancer 129 (2011) 1137. [DOI] [PubMed] [Google Scholar]
  • [147].Seligson DB, Horvath S, McBrian MA, Mah V, Yu H, Tze S, Wang Q, Chia D, Goodglick L, Kurdistani SK, Global levels of histone modifications predict prognosis in different cancers, Am. J. Pathol 174 (2009) 1619. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [148].Seng TJ, Currey N, Cooper WA, Lee CS, Chan C, Horvath L, Sutherland RL, Kennedy C, McCaughan B, Kohonen-Corish MR, DLEC1 and MLH1 promoter methylation are associated with poor prognosis in non-small cell lung carcinoma, Br. J. Cancer 99 (2008) 375. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [149].Siegel R, Ward E, Brawley O, Jemal A, Cancer statistics, 2011: the impact of eliminating socioeconomic and racial disparities on premature cancer deaths, CA Cancer J. Clin 61 (2011) 212. [DOI] [PubMed] [Google Scholar]
  • [150].Simao TA, Simoes GL, Ribeiro FS, Cidade DA, Andreollo NA, Lopes LR, Macedo JM, Acatauassu R, Teixeira AM, Felzenszwalb I, Pinto LF, Albano RM, Lower expression of p14ARF and p16INK4a correlates with higher DNMT3B expression in human oesophageal squamous cell carcinomas, Hum. Exp. Toxicol 25 (2006) 515. [DOI] [PubMed] [Google Scholar]
  • [151].Spange S, Wagner T, Heinzel T, Kramer OH, Acetylation of non-histone proteins modulates cellular signalling at multiple levels, Int. J. Biochem. Cell Biol 41 (2009) 185. [DOI] [PubMed] [Google Scholar]
  • [152].Stevenson BJ, Iseli C, Panji S, Zahn-Zabal M, Hide W, Old LJ, Simpson AJ, Jongeneel CV, Rapid evolution of cancer/testis genes on the X chromosome, BMC Genomics 8 (2007) 129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [153].Stockert E, Jager E, Chen YT, Scanlan MJ, Gout I, Karbach J, Arand M, Knuth A, Old LJ, A survey of the humoral immune response of cancer patients to a panel of human tumor antigens, J. Exp. Med 187 (1998) 1349. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [154].Suva ML, Riggi N, Janiszewska M, Radovanovic I, Provero P, Stehle JC, Baumer K, Le Bitoux MA, Marino D, Cironi L, Marquez VE, Clement V, Stamenkovic I, EZH2 is essential for glioblastoma cancer stem cell maintenance, Cancer Res. 69 (2009) 9211. [DOI] [PubMed] [Google Scholar]
  • [155].Ting DT, Lipson D, Paul S, Brannigan BW, Akhavanfard S, Coffman EJ, Contino G, Deshpande V, Iafrate AJ, Letovsky S, Rivera MN, Bardeesy N, Maheswaran S, Haber DA, Aberrant overexpression of satellite repeats in pancreatic and other epithelial cancers, Science 331 (2011) 593–596. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [156].Toh Y, Yamamoto M, Endo K, Ikeda Y, Baba H, Kohnoe S, Yonemasu H, Hachitanda Y, Okamura T, Sugimachi K, Histone H4 acetylation and histone deacetylase 1 expression in esophageal squamous cell carcinoma, Oncol. Rep 10 (2003) 333. [PubMed] [Google Scholar]
  • [157].Traynor AM, Dubey S, Eickhoff JC, Kolesar JM, Schell K, Huie MS, Groteluschen DL, Marcotte SM, Hallahan CM, Weeks HR, Wilding G, Espinoza-Delgado I, Schiller JH, Vorinostat (NSC# 701852) in patients with relapsed non-small cell lung cancer: a Wisconsin Oncology Network phase II study, J. Thorac. Oncol 4 (2009) 522. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [158].Tzao C, Tung HJ, Jin JS, Sun GH, Hsu HS, Chen BH, Yu CP, Lee SC, Prognostic significance of global histone modifications in resected squamous cell carcinoma of the esophagus, Mod. Pathol 22 (2009) 252. [DOI] [PubMed] [Google Scholar]
  • [159].Vallbohmer D, Brabender J, Yang D, Schneider PM, Metzger R, Danenberg KD, Holscher AH, Danenberg PV, DNA methyltransferases messenger RNA expression and aberrant methylation of CpG islands in non-small-cell lung cancer: association and prognostic value, Clin. Lung Cancer 8 (2006) 39. [DOI] [PubMed] [Google Scholar]
  • [160].Van Den Broeck A, Brambilla E, Moro-Sibilot D, Lantuejoul S, Brambilla C, Eymin B, Khochbin S, Gazzeri S, Loss of histone H4K20 trimethylation occurs in preneoplasia and influences prognosis of non-small cell lung cancer, Clin. Cancer Res 14 (2008) 7237. [DOI] [PubMed] [Google Scholar]
  • [161].Vatolin S, Abdullaev Z, Pack SD, Flanagan PT, Custer M, Loukinov DI, Pugacheva E, Hong JA, Morse III H, Schrump DS, Risinger JI, Barrett JC, Lobanenkov VV, Conditional expression of the CTCF-paralogous transcriptional factor BORIS in normal cells results in demethylation and derepression of MAGE-A1 and reactivation of other cancer-testis genes, Cancer Res. 65 (2005) 7751. [DOI] [PubMed] [Google Scholar]
  • [162].Vrzalikova K, Skarda J, Ehrmann J, Murray PG, Fridman E, Kopolovic J, Knizetova P, Hajduch M, Klein J, Kolek V, Radova L, Kolar Z, Prognostic value of Bmi-1 oncoprotein expression in NSCLC patients: a tissue microarray study,J. Cancer Res. Clin. Oncol 134 (2008) 1037. [DOI] [PubMed] [Google Scholar]
  • [163].Vu TH, Nguyen AH, Hoffman AR, Loss of IGF2 imprinting is associated with abrogation of long-range intrachromosomal interactions in human cancer cells, Hum. Mol. Genet 19 (2010) 901. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [164].Wargo JA, Robbins PF, Li Y, Zhao Y, El-Gamil M, Caragacianu D, Zheng Z, Hong JA, Downey S, Schrump DS, Rosenberg SA, Morgan RA, Recognition of NYESO-1+ tumor cells by engineered lymphocytes is enhanced by improved vector design and epigenetic modulation of tumor antigen expression, Cancer Immunol. Immunother 58 (2009) 383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [165].Watanabe H, Soejima K, Yasuda H, Kawada I, Nakachi I, Yoda S, Naoki K, Ishizaka A, Deregulation of histone lysine methyltransferases contributes to oncogenic transformation of human bronchoepithelial cells, Cancer Cell Int. 8 (2008) 15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [166].Weiser TS, Guo ZS, Ohnmacht GA, Parkhurst ML, Tong-On P, Marincola FM, Fischette MR, Yu X, Chen GA, Hong JA, Stewart JH, Nguyen DM, Rosenberg SA, Schrump DS, Sequential 5-Aza-2 deoxycytidine-depsipeptide FR901228 treatment induces apoptosis preferentially in cancer cells and facilitates their recognition by cytolytic T lymphocytes specific for NY-ESO-1, J. Immunother 24 (2001) 151. [DOI] [PubMed] [Google Scholar]
  • [167].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 39 (2007) 157. [DOI] [PubMed] [Google Scholar]
  • [168].Wienholz BL, Kareta MS, Moarefi AH, Gordon CA, Ginno PA, Chedin F, DNMT3L modulates significant and distinct flanking sequence preference for DNA methylation by DNMT3A and DNMT3B in vivo, PLoS Genet. 6 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [169].Wu D, Xiong L, Wu S, Jiang M, Lian G, Wang M, TFPI-2 methylation predicts poor prognosis in non-small cell lung cancer, Lung Cancer 76 (2012) 106. [DOI] [PubMed] [Google Scholar]
  • [170].Xing J, Stewart DJ, Gu J, Lu C, Spitz MR, Wu X, Expression of methylation-related genes is associated with overall survival in patients with non-small cell lung cancer, Br. J. Cancer 98 (2008) 1716. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [171].Xiong J, Epstein RJ, Growth inhibition of human cancer cells by 5-aza-2′-deoxycytidine does not correlate with its effects on INK4a/ARF expression or initial promoter methylation status, Mol. Cancer Ther 8 (2009) 779. [DOI] [PubMed] [Google Scholar]
  • [172].Yamada A, Fujii S, Daiko H, Nishimura M, Chiba T, Ochiai A, Aberrant expression of EZH2 is associated with a poor outcome and P53 alteration in squamous cell carcinoma of the esophagus, Int. J. Oncol 38 (2011) 345. [DOI] [PubMed] [Google Scholar]
  • [173].Yamada Y, Jackson-Grusby L, Linhart H, Meissner A, Eden A, Lin H, Jaenisch R, Opposing effects of DNA hypomethylation on intestinal and liver carcinogenesis, Proc. Natl. Acad. Sci. U. S. A 102 (2005) 13580. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [174].Yanagawa N, Tamura G, Oizumi H, Endoh M, Motoyama T, MAGE expressions mediated by demethylation of MAGE promoters induce progression of non-small cell lung cancer, Anticancer. Res 31 (2011) 171. [PubMed] [Google Scholar]
  • [175].Yanagawa N, Tamura G, Oizumi H, Kanauchi N, Endoh M, Sadahiro M, Motoyama T, Promoter hypermethylation of RASSF1A and RUNX3 genes as an independent prognostic prediction marker in surgically resected non-small cell lung cancers, Lung Cancer 58 (2007) 131. [DOI] [PubMed] [Google Scholar]
  • [176].Yang B, O’Herrin SM, Wu J, Reagan-Shaw S, Ma Y, Bhat KM, Gravekamp C, Setaluri V, Peters N, Hoffmann FM, Peng H, Ivanov AV, Simpson AJ, Longley BJ, MAGE-A, mMage-b, and MAGE-C proteins form complexes with KAP1 and suppress p53-dependent apoptosis in MAGE-positive cell lines, Cancer Res. 67 (2007) 9954. [DOI] [PubMed] [Google Scholar]
  • [177].Yeow WS, Ziauddin MF, Maxhimer JB, Shamimi-Noori S, Baras A, Chua A, Schrump DS, Nguyen DM, Potentiation of the anticancer effect of valproic acid, an antiepileptic agent with histone deacetylase inhibitory activity, by the kinase inhibitor Staurosporine or its clinically relevant analogue UCN-01, Br. J. Cancer 94 (2006) 1436. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [178].Yi JM, Kim HS, Molecular phylogenetic analysis of the human endogenous retrovirus E (HERV-E) family in human tissues and human cancers, Genes Genet. Syst 82 (2007) 89. [DOI] [PubMed] [Google Scholar]
  • [179].Yoon KA, Hwangbo B, Kim IJ, Park S, Kim HS, Kee HJ, Lee JE, Jang YK, Park JG, Lee JS, Novel polymorphisms in the SUV39H2 histone methyltransferase and the risk of lung cancer, Carcinogenesis 27 (2006) 2217. [DOI] [PubMed] [Google Scholar]
  • [180].Yoshimura H, Takemoto K, Effect of cigarette smoking and/or N-bis(2-hydroxypropyl)nitrosamine (DHPN) on the development of lung and pleural tumors in rats induced by administration of asbestos, Sangyo Igaku 33 (1991) 81. [DOI] [PubMed] [Google Scholar]
  • [181].Yoshino M, Suzuki M, Tian L, Moriya Y, Hoshino H, Okamoto T, Yoshida S, Shibuya K, Yoshino I, Promoter hypermethylation of the p16 and Wif-1 genes as an independent prognostic marker in stage IA non-small cell lung cancers, Int. J. Oncol 35 (2009) 1201. [DOI] [PubMed] [Google Scholar]
  • [182].Yost JM, Korboukh I, Liu F, Gao C, Jin J, Targets in epigenetics: inhibiting the methyl writers of the histone code, Curr. Chem. Genomics 5 (2011) 72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [183].Yu X, Guo ZS, Marcu MG, Neckers L, Nguyen DM, Chen GA, Schrump DS, Modulation of p53, ErbB1, ErbB2, and Raf-1 expression in lung cancer cells by depsipeptide FR901228, J. Natl. Cancer Inst 94 (2002) 504. [DOI] [PubMed] [Google Scholar]
  • [184].Yu XD, Wang SY, Chen GA, Hou CM, Zhao M, Hong JA, Nguyen DM, Schrump DS, Apoptosis induced by depsipeptide FK228 coincides with inhibition of survival signaling in lung cancer cells, Cancer J. 13 (2007) 105. [DOI] [PubMed] [Google Scholar]
  • [185].Zauderer MG, Krug LM, The evolution of multimodality therapy for malignant pleural mesothelioma, Curr. Treat. Options Oncol (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [186].Zhai R, Chen F, Liu G, Su L, Kulke MH, Asomaning K, Lin X, Heist RS, Nishioka NS, Sheu CC, Wain JC, Christiani DC, Interactions among genetic variants in apoptosis pathway genes, reflux symptoms, body mass index, and smoking indicate two distinct etiologic patterns of esophageal adenocarcinoma,J. Clin. Oncol 28 (2010) 2445. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [187].Zhang JG, Guo JF, Liu DL, Liu Q, Wang JJ, MicroRNA-101 exerts tumor-suppressive functions in non-small cell lung cancer through directly targeting enhancer of zeste homolog 2, J. Thorac. Oncol 4 (2009) 161–166. [DOI] [PubMed] [Google Scholar]
  • [188].Zhang W, Peyton M, Xie Y, Soh J, Minna JD, Gazdar AF, Frenkel EP, Histone deacetylase inhibitor romidepsin enhances anti-tumor effect of erlotinib in non-small cell lung cancer (NSCLC) cell lines, J. Thorac. Oncol 4 (2009) 161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [189].Zhang X, Wen H, Shi X, Lysine methylation: beyond histones, Acta Biochim. Biophys. Sin. (Shanghai) 44 (2012) 14. [DOI] [PubMed] [Google Scholar]
  • [190].Zhang XY, Dong QG, Huang JS, Huang AM, Shi CL, Jin B, Sha HF, Feng JX,Geng Q, Zhou J, Xu HL, Han BH, The expression of stem cell-related indicators as a prognostic factor in human lung adenocarcinoma, J. Surg. Oncol 102 (2010) 856. [DOI] [PubMed] [Google Scholar]
  • [191].Zhang Y, Miao Y, Yi J, Wang R, Chen L, Frequent epigenetic inactivation of deleted in lung and esophageal cancer 1 gene by promoter methylation in non-small-cell lung cancer, Clin. Lung Cancer 11 (2010) 264. [DOI] [PubMed] [Google Scholar]
  • [192].Zhao R, DeCoteau JF, Geyer CR, Gao M, Cui H, Casson AG, Loss of imprinting of the insulin-like growth factor II (IGF2) gene in esophageal normal and adeno-carcinoma tissues, Carcinogenesis 30 (2009) 2117. [DOI] [PubMed] [Google Scholar]

RESOURCES