Open access peer-reviewed chapter

Epigenomics in Malignant Pleural Mesothelioma

Written By

Aldo Manuel Alvarez Moran, Pablo Alejandro Ávila Sánchez, Jorge Alejandro Torres Ríos and Lorena Vega Castillo

Submitted: 29 November 2021 Reviewed: 13 May 2022 Published: 29 June 2022

DOI: 10.5772/intechopen.105408

From the Edited Volume

Mesothelioma - Diagnostics, Treatment and Basic Research

Edited by Ilze Strumfa

Chapter metrics overview

152 Chapter Downloads

View Full Metrics

Abstract

Malignant pleural mesothelioma (MPM) is a tumor with a relatively low incidence, but whose carcinogenesis, for the most part, involves epigenetic factors that keep its heterogeneity and sometimes are a therapeutic target or an obstacle to the effectiveness of the newest treatments. This chapter summarizes the principal epigenetic dysregulation mechanisms involved in the MPM pathogenesis. The most studied mechanism is hypermethylation mediated by DNA methyltransferases (DNMTs) in different tumor suppressor genes, and the relation with asbestos fiber exposure, which represents the main risk factor. Physiopathology is related to chronic inflammation mediated by free radicals that produce chromosomal alterations, genomic instability, increased angiogenesis, and tumor invasion factors like EGFR, FGFR, TGF-B, and PDGF. Additionally, independent methylation pathways that produce gene silencing such as polycomb complex and SWI/SNF mutation are reviewed. Finally, other mechanisms are described such as hypomethylation with imprint loss and pro-oncogenic gene activation that induce immunological responses, as well as acetylation, deacetylation, and demethylation in the chromatin and histone context.

Keywords

  • malignant pleural mesothelioma
  • epigenetics
  • hypermethylation
  • asbestos
  • genomic instability

1. Introduction

Human malignant pleural mesothelioma (MPM) is an invariably fatal tumor due to its heterogeneity, growing from the serous surfaces of the pleura. Many factors are involved in its occurrence, such as exposure to asbestos fibers and simian virus 40; these factors being those that are strongly associated with the tumorigenesis of this disease. The annual incidence of MPM is relatively low, estimated in a range of 0.6–30/10,00,000, but the global occurrence is expected to increase continuously in future years [1]. MPM is extremely heterogeneous in its morphology and molecular phenotypes. The latency period for MPM development is 10–50 years after asbestos exposure. The prognosis for MPM is generally poor, with a median survival time of 12 months from diagnosis [1].

Intratumor heterogeneity refers to a mixture of phenotypic, functional, and genetic differences within cancer cells with various differentiation or hierarchical statuses within the tumor. It is a common feature in most tumors. This heterogeneity has been considered the greatest obstacle to the effectiveness of most cancer therapies, manifesting itself in its sensitivity to different therapies. Several studies have been focused on genetic alterations as part of the mechanism of tumoral cells for the generation and maintenance of this heterogeneity. In addition, some other studies show the role of epigenetic modifications involved in its heterogeneity. Despite this, there is scarce information about epigenetic modifications that could explain this process [1, 2].

Epigenetic modifications are heritable and stable alterations of genes that do not change the DNA sequence, including DNA methylation, histone modification, and non-coding RNA interference modifications. DNA methylation has been extensively studied in the development of cancer. On the one hand, hypermethylation in cancer-related promoter genes induces the silencing or downregulation of tumor suppressor genes and repair genes. On the other hand, hypomethylation of DNA leads to activation of oncogenes and genomic instability. Several authors suggest that aberration in DNA methylation may play an important role on tumor cells heterogeneity [3, 4, 5].

The exact mechanisms by which asbestos fibers promote the development of cancer are unknown, however, the most accepted theory is the induction of chronic inflammation and signaling pathways in the transformation of reactive oxygen species generated by asbestos fibers. Therefore, this chapter will address an overview of the epigenetic profile of MPM and the mechanisms that promote epigenetic modifications where asbestos fibers might play an important role.

Advertisement

2. Asbestos-induced molecular alterations

As previously mentioned, asbestos exposure is a primary cause of the development of pleural mesothelioma. Molecular analyses show that asbestos-related carcinogenesis is caused by chronic inflammation that both promote the release of oxygen free radicals that alter intracellular components, and DNA mutation and its consequent transformation. Asbestos fibers also contain iron ions and can induce hemolysis by sequestering iron from hemoglobin. This is particularly important since free iron disproportionately releases H2O2, which consequently releases hydroxyl radicals (OH) that oxidize DNA, and release nucleic acids, proteins, and lipids. This process is exacerbated by the release of cytokines including tumor necrosis factor-alpha from macrophages and high mobility of box group 1 (HMGB1) proteins from necrotic cells, leading to an amplification of the inflammatory response and an increase in cells that are driven to oxidative damage. Damaged oxidized DNA, if not properly repaired, is highly mutagenic and can lead to genomic instability. A multitude of oxidative DNA lesions includes oxidation of DNA bases, baseless sites, single-strand breaking, double-strand breaking, and interchain breaking, all of which require different pathways for proper repair. These last two chain-breaking types are particularly toxic, since they cause replication collapse, as well as allow chromosome rearrangements, chromosome gains, losses, or fragmentation [2].

There are other mechanisms involved in how asbestos fibers cause MPM (Figure 1). Four proposed models related to asbestos fibers induce genetic and cellular damage to cells, in addition to the previously mentioned chronic inflammation. The different molecular models involved in asbestos exposure are explained below:

  1. Reactive oxygen species generated by asbestos fibers with their surface exposed leads to DNA damage and cell membrane rupture. Macrophages that engulf asbestos fibers but cannot digest them also produce abundant reactive oxygen species.

  2. Asbestos fibers are also engulfed by mesothelial cells. Asbestos fibers collected in cells can physically interfere with the mitotic process of the cell. The cycle is cut by the interruption of the mitotic spindles. Another important aspect is the entanglement of asbestos fibers with the chromosomes or mitotic spindles that can give rise to structurally damaged chromosomes such as aneuploidies of normal mesothelial cells.

  3. Asbestos fibers can absorb a variety of proteins and chemicals on the wide surface of asbestos, which can result in the accumulation of dangerous molecules including carcinogens. The asbestos fibers also bind to important cellular proteins and a deficiency of these proteins can also be detrimental to normal mesothelial cells.

  4. Finally, mesothelial cells and macrophages exposed to asbestos release a variety of cytokines and growth factors that induce inflammation and tumor promotion. These include tumor necrosis factor α, interleukin 1β, transforming growth factor β, and platelet-derived growth factor. Tumor necrosis factor-α has been shown to activate nuclear factor-κB, leading to mesothelial cell survival and inhibiting asbestos-induced cytotoxicity. The high mobility group protein box 1 (GAMB1) is released from mesothelial cells, which are exposed to asbestos and then undergo necrotic cell death, promoting an inflammatory response. Thus, aberrantly activated signaling between mesothelial cells, inflammatory cells, fibroblasts, and other stromal cells can create a set of mesothelial cells, which harbor aneuploidy and DNA damage, potentially developing cancer cells and together all these phenomena form a tumoral microenvironment that supports and nurtures them [6, 7, 8].

Figure 1.

Possible oncogenic mechanisms induced by asbestos. Abbreviations: HMGB1 = high-mobility group box 1. ROS = reactive oxygen species. TGF-B = transforming growth factor beta. VEGF = vascular endothelial growth factor.

2.1 DNA methylation

Methylated DNA studied through immunoprecipitation grounded on next-generation sequencing makes it possible to analyze the DNA methylome, which constitutes a useful and efficient tool in the approach of cancer epigenomics [5, 9, 10].

An important and widely described phenomenon in the development of MPM is the epigenetic dysregulation that promotes changes in gene expression [11]. DNA methylation modifications play an important role in the malignant transformation of mesothelioma. Survival in MPM has been attributed to promoter methylation and silencing of genes such as SFRP4, SFRP5, FHIT, and SLCA20.

The methylated CpG islands have been shown to affect different process, such as uncontrolled cell proliferation & differentiation and dysregulations in apoptosis, in the oncogenic process of MPM. It is important to mention that asbestos fibers have been related with increased prevalence of aberrant promoter methylation by controlling the APC and RASSF1 genes, directly affecting the cell cycle [1, 2, 3, 4].

Epigenetic modifications require active maintenance and are potentially reversible, characteristics that make them targets for therapeutic strategies. Multiple DNA methyltransferases and histone deacetylases (HDACs) participate on the regulation of some tumor suppressor genes by gene silencing and chromatin compaction. Therefore, changes in these two enzymes promote disturbances in gene expression and allow deflections in cell proliferation, differentiation, and apoptosis. When HDACs are inhibited, there is a massive production of superoxide radicals and the caspase system is activated, leading to cell death. Additionally, hyperacetylation of non-histone proteins takes place, promoting angiogenesis and tumor cells motility and invasion [12].

DNA modifications are not the only mechanisms involved in tumorogenesis. Epigenetic changes also play an important role in oncogenesis through changes in DNA-associated proteins, modifying their expression. In this regard, the most important changes are DNA methylation and histone deacetylation. These changes lead to important modifications in DNA activity and expression. As a result of this process, some proteins involved in tumorogenesis can be induced and modulated, for example, epidermal growth receptor factor, tumor necrosis factor-alpha protein fusion peptide, transforming growth factor-beta and others. As mentioned above, these changes are induced by epigenetic mechanisms that are potentially reversible [12, 13].

In recent years, inhibiting tyrosinase-like receptors (RTKs) has been used as a therapeutic target because MPM cells have been shown to express high levels of receptors that can bind to key molecules, such as epidermal growth receptor factor (EGFR) and platelet-derived growth factor (PDGF), fibroblast growth receptor factor (FGFR-1y3), transforming growth factor-beta (TGF-B), insulin-like growth factor (IGF-1R), and tumor necrosis factor-alpha protein fusion peptide (NGR-hTNF-alpha). All these molecules undergo through epigenetic changes and play a dead serious role in tumor invasion and angiogenesis [12, 13].

Numerous genes have been shown to be epigenetically downregulated, as the DNA methylation of transcriptional promoters. These changes deregulate several signaling pathways, including the WNT pathway, in which several negative regulators are hypermethylated and silenced [14, 15]. The global epigenetic profile determined by high-throughput analysis differs between MPM and normal pleura, showing that MPM has aberrant methylation in the CpG islands, as has been mentioned [16, 17]. These data support the hypothesis that a specific DNA methylation pathway is induced during mesothelial carcinogenesis.

Kim et al. [1] carried out a study in a patient with MPM, 122 differently regulated genes were found, 118 genes were down-regulated and four were up-regulated by hypomethylation. Therefore, MPM cells may be epigenetically regulated, and DNA methylation plays a main role in intratumorally heterogeneity, characteristic that boost MPM more aggressiveness.

2.2 Factors associated with methylation

There are sundry important factors that have been related with DNA methylation of gene loci in MPM such as age-related changes, ethnicity, histological subtype, and asbestos exposure. These factors could explain discrepancies between DNA methylation frequencies in published studies, as well as the experimental method used to detect it. In patients diagnosed with MPM, an increased DNA methylation associated with increased age has been reported. Some studies have shown that methylation status of the IGFBP2 (insulin growth factor binding protein) locus and GDF10 (bone morphogenetic protein) locus is significantly higher in MPM in Japanese patients compared with US patients [18, 19].

There are some concrete characteristics that are related to specific genes, for example; RASSF1 suppressor gene has been reported to have a significantly higher frequency of aberrant methylation in epithelioid MPM than in the sarcomatoid subtype [20, 21]. Methylation of MT2A gene, is shown to differ between these two histological subtypes. Epithelioid and sarcomatoid mesotheliomas also have different methylation changes at 87 CpG islands [22, 23]. MT1A and MT2A gene loci associated with DNA methylation have also been described in MPM.

CpG island methylation in the CCND2, CDKN2A, CDKN2B, HPPBP1, and RASSF1 genes has been studied in correlation with asbestos exposure. The RASSF1 DNA methylation locus is related with a higher number of asbestos bodies in the lung. There are different methylation profiles in MPM according with its exposure to asbestos and a positive association between asbestos fiber load and CDKN2A, CDKN2B, RASSF1 methylation status, and MT1A at another 100 loci.

2.3 Methylation and diagnosis through DNA

Some differences have been described in DNA methylation for sundry genes between MPM, lung adenocarcinoma, and in non-malignant lung tissues. That’s why, at these days, DNA methylation is an important tool in the diagnosis of MPM [20, 24]. Thus, the DNA methylation profile has potential helpfulness in the diagnostic of MPM and reject of other differential diagnoses. It has been demonstrated by high-throughput analyses for methylation, spanning several thousand CpG islands. It was recently suggested that DNA methylation at three specific loci: TMEM30B, KAZALD1, and MAPK13, could be useful in the differential diagnosis of MPM. In the near future, MPM diagnosis may be based on the methylation profile, but by now, further studies in larger populations are necessary before using a limited number of hypermethylated loci [19, 20, 21].

Other studies have shown alterations in the methylation status of individual genes, such as those HIC1, PYCARD, LZTS1, and SLC6A20. All of these genes have been associated with a good or bad prognosis [22, 23]. Besides, patients with MPM and a low frequency of DNA methylation had longer survival [22, 23, 24].

In view of the aberrant epigenetic events observed in MPM and the clinical value of histone deacetylase inhibitors (HDACis), the latter is currently being studied as a potential diagnostic method. However, insufficient data is yet available on the regulation of histone modifications, despite their crucial role in maintaining chromatin stability. These data are needed to support clinical trials based on HDACis [6, 7, 25, 26].

2.4 Epigenetic regulation in mesothelioma gene expression

Each nucleosome is made up of 147 base pairs (bp) of DNA wrapped twice around a histone octamer. Epigenetic regulation of gene expression occurs in the context of chromatin, the basic unit of the nucleosome. Lysine-rich histone tails extend from the nucleosome and provide sites for covalent and reversible binding, promoting processes such as acetylation, methylation, ubiquitination, phosphorylation and SUMOylation, which produce the activation or inhibition of gene expression [8, 27].

DNA methylations represent the most important mechanism regulating major changes in gene expression during normal cell cycle and tissue differentiation, as well as long-term repression of imprinted alleles, germ cell-restricted genes, repetitive DNA, and sequences. Endogenous retrovirals [27, 28, 29]. Normal somatic cells have three major DNA methyltransferases: DNMT1, DNMT3A, and DNMT3B. All these enzymes mediate the transfer of a methyl group from S-adenosyl-methionine to the 5′ position of cytosine in the context of CpG. CpG dinucleotide groups are found in the promoters of approximately 60% of genes. Furthermore, most of these islands are unmethylated, allowing for a relaxed structure (euchromatin) and active transcription [30]. Some other CpG dinucleotides and CpG islands, which are often hypermethylated in normal cells, are scattered throughout the genome [31]. Although there is considerable overlap, DNMT1 preferentially binds hypermethylated DNA and works primarily as a housekeeping methyltransferase, restoring DNA methylation patterns during the process of DNA repair or replication. On the other hand, DNMT3A and 3B mediate de novo DNA methylation after recognition of unmethylated or hypermethylated DNA [30, 31].

It is important to recapitulate that methylation-sensitive transcription factor binding is inhibited by DNA methylation, and these changes promote the recruitment of the CpG methyl-binding domain (MBD) and relevant proteins such as UHRF1, syn3a-containing repressor complexes, NCoRs and histone deacetylases (HDACs), resulting in silent transcriptional heterochromatin output [32, 33, 34].

During the process of malignant transformation, the aberrant orientation and overexpression of some factors involved in DNA methylation promote the epigenetic silencing of genes related to differentiation, many of which are tumor suppressors. On the other hand, tumor suppressor genes can be inactivated by DNA methylation through transitional mutations resulting from deamination of 5-methylcytosine (5-MC) or adduct formation with environmental carcinogens such as benzopyrene [35].

DNA demethylation occurs passively during DNA replication [36, 37]. In addition, DNA can be actively demethylated by oxidation of 5-MC to 5-hydroxymethylcytosine, a ten-eleven translocation (TET) enzyme-mediated reaction [20].

The total amount of methylated CpGs, during malignant transformation, is up to 50%, excluding CpG promoter islands. The genome-wide DNA demethylation is importantly related to a deficient DNA repair process [38, 39, 40, 41]. Besides, it can promote unrepression of imprinted alleles, endogenous retroviruses, and transposable elements, inducing genomic instability [42, 43]. On the other hand, the mechanisms that mediate this phenomenon, such as decreased expression of methyltransferase 1 [44, 45, 46] glycosylase-mediated cleavage of 5-MC and aberrant expression/orientation of TET proteins, have not been fully elucidated [38].

The most widely characterized histone modifications in normal cells and malignant cells have been the acetylation-deacetylation and the methylation-demethylation [1, 2, 43, 47]. Histone acetylation is mediated by a variety of histone acetyltransferases (HAT), increasing the net negative charge leading to DNA repulsion, chromatin relaxation, and gene expression. Some non-histone proteins, including Hsp90, SP1, p53, and HDAC1, are targets for HAT and HDAC. In the other hand, histone deacetylation is regulated by HDAC [48].

Histone lysine methylation is mediated by a variety of histone methyltransferases (KMTs), lysine mediating monomethylation/dimethylation/trimethylation of specific residues, whereas histone demethylation is mediated by histone demethylases [47, 49, 50]. Histone modifications are highly dynamic in response to environmental signals [51, 52]. Unlike histone acetylation, histone lysine methylation does not modify the charge of core histones. Furthermore, histone lysine methylation can promote or inhibit gene expression.

ATP-dependent chromatin remodeling complexes have emerged, in recent years, as critical mediators of the epigenetic regulation of gene expression in normal and malignant cells [53, 54]. To date, four gene families have been described including switch/non-fermentable sucrose (SWI/SNF), SWI mimetic (ISWI), DNA-binding helicase chromodomain (CHD), and INO80, named for their ability to regulate inositol-responsive gene expression. All these complexes have multiple subunits with diverse isoforms and exhibit pleiotropic functions including regulation of gene expression, maintenance of chromatin structure, replication of pericentromeric heterochromatin, repression of ribosomal RNA, and repair of cell damage. DNA [55]. There are several mechanisms by which different families remodel chromatin. For example, the SWI/SNF complexes expose DNA by disassembling the nucleosome, while members of the ISWI, INO80, and CDH families reposition (slide) the nucleosomes and extend the intervening DNA, promoting access to transcriptional factors. These complexes also have an important role in maintaining chromatin structure and genome stability, through mechanisms that reassemble the nucleosome [53, 55].

Studies in transcriptome analysis have revealed that almost 90% of the genome is transcribed as non-coding RNAs (IncRNAs), which are critical mediators of chromatin structure and gene expression in normal cells and malignant transformation [56, 57, 58]. Besides, lncRNAs participate in the recruitment of DNMTs and histone methyltransferases to chromatin [59], adding another layer of epigenetic regulation in normal cells which is altered in malignant tumors.

Advertisement

3. Methylation-mediated suppressor gene silencing

There are several studies that have shown a relationship between silencing suppressor gene by methylation process and the development of MPL. For example, Christensen et al. [23] examined the DNA methylation status of the promoter of six genes that regulate cell cycle progression at 70 MPM. The extent of methylation of these genes was correlated with lung asbestos burden and overall survival. Goto et al. [60] studied methylation process in more than 6000 GpC islands, comparing twenty MPM versus twenty lung adenocarcinomas, using microarray PCR technique. Their results are interesting because they found out that 387 genes (6.3%) were hypermethylated in mesotheliomas, while the number of hypermethylated gene in lung adenocarcinoma were higher with a total amount of 544 genes (8.8%).

MPL patients’ survival is related with DNA methylation levels. In this way, higher levels of DNA methylation correlate with lower patient survival. Three genes; TMEM30B, KAZALD1 and MAPK13, are specifically hypermethylated in MPM. Several reports have documented tumor suppressor gene silencing related to DNA methylation process in MPM (Table 1) and there is evidence of hypermethylation of some of these genes affecting overall survival.

APC1AP151NK4B
APC1BP16
BMP3bRARB
CDH1RASSF1A
DAPKSFRPs
ESR1SLC6A20
FHITSYK
IGFBP3TMEM30B
KAZALD1THBD
MAPK13TMEM30B
MGMTTYMP
P14ARFWIF-1

Table 1.

Hypermethylated genes related to MPM.

MPM: malignant pleural mesothelioma.

Currently, scientific evidence has shown that recurrent hypermethylation is highly related to tumor suppressor genes in MPM, however, the mechanisms behind this process have been poorly studied. Novel studies have identified TC2N gene as a tumorigenesis promoter by silencing p53. Cytokine signaling participate in modulation process of DNMT expression and mediate hypermethylation of target genes enrolled in some types of cancer such as colorectal carcinoma and erythroleukemia cells [61, 62]. Exposure to asbestos fibers leads to a cytokine cascade induced by high mobility group 1 (HMGB1) or the NLRP3 inflammasome. These cytokines use to change the regulation of the expression of DNMT and other components of the methylation machinery during the process of MPM evolution.

A study of a panel of genes encoding epigenetic regulators in a panel of cultured cell lines derived from asbestos-associated MPM relative to LP-9 (a commercially available normal mesothelial cell line) was recently carried out. Consistent with the study results, TCGA data demonstrate a spectrum of DNMT expression in MPM and suggest that overexpression of DNMT1, DNMT3A, and DNMT3B correlates with decreased survival of pleural mesothelioma patients (Figure 2).

Figure 2.

Association between intratumoral DNMT expression levels and surveillance in patients with MPM. The Kaplan Meier waves show that DNMT1, DNMT3A, and DNMT3B expression, measured by RNA-seq technique, has negative impact in patients’ surveillance.

Kim et al. [1] studied gene expression and methylation profiles in pluripotent populations (SP) and non-SP fractions in human MPM samples, using RNA-seq and methylated DNA immunoprecipitation techniques. They found 6400 hypermethylated genes and 3400 hypomethylated genes in SP. Seven hundred and ninety-five genes were upregulated, while three hundred and thirty-five were significantly repressed in SP compared to non-SP fractions. They looked at changes in DNA methylation and expression levels of 122 genes; 118 genes were hypermethylated and downregulated, while 4 were hypomethylated and upregulated. Ten other genes showed hypermethylation and low expression of CpG promoter islands.

Advertisement

4. Loss of imprinting (LI) and de-repression of cancer-germline (CG) genes

The loss of the imprinting process is largely due to DNA hypomethylation. Repression of endogenous retroviral sequences and activation of GC genes can promote malignant transformation by increasing proliferation, genomic instability, and resistance to apoptosis [63, 64].

A fascinating phenomenon can occur during the malignant transformation of somatic cells. The development of highly limited tumor antigens that induce serological and cell-mediated immune responses in cancer patients can be caused by abnormal activation of GC genes [also known as testicular cancer (TC) genes]. As a result, testicular cancer antigens (ATCs) have become popular targets for cancer immunotherapy in recent years [65]. More than 270 GC genes have been registered in the international TC database thus far. Seventy-five percent of these genes are only expressed in normal testes and malignant neoplasms, while the rest have high levels of expression in testes and varying levels of expression in other normal tissues and malignancies. On the X chromosome, around half of the GC genes are encoded. Families of cancer-testis-X (CT-X) chromosomal genes with inverted DNA repeats are common. On the other hand, inverted repetitive DNA sequences or extended families or are not linked to non-X CT genes [66, 67]. Furthermore, CT-X genes are frequently active in malignancies, and genes from families are increased in a tumor-specific manner, implying that the CT-X genes have a transcriptional coregulation and functional link.

In human malignancies, the stage at which the disease is discovered at a specific time corresponds to the degree of CG gene repression. Malignant and aggressive phenotype of cancer cells is promoted activations of this genes. BORIS/CTCFL, for example, upregulates h-TERT and suppresses apoptosis in cancer cells via processes that are still unknown [68, 69]. MAGE-A11 regulates the activity of the tumor suppressor gene RBL1/p107 [63]. MAGE-A11 inhibits the tumor suppressor gene RBL1/p107, while MAGE-B2 promotes cell cycle advancement by increasing E2F activity. MAGE-A2 and MAGE-C2 prevent p53 from binding to target promoters, changing its activities and leading to p53 deacetylation (inactivation) or enhanced ubiquitin-mediated degradation. The absence of CG gene regulation does not appear to be just a symptom of pluripotency, as it is accompanied with chromosomal hypomethylation. In human ESC, mesenchymal stem cells, and adipose-derived stem cells, Loriot et al. [70] found no overexpression of 18 different CG genes. In induced pluripotent stem cells (iPSCs) produced from normal small airway epithelial cells, transcriptional repression of CG genes such as NY-ESO-1, MAGE-A1, and MAGE-A3, which are generally located upward in thoracic malignant tumors, has been detected, which is consistent with these findings. Although these findings imply that iPSC reprogramming is partial, induction of CG genes in cancer cells may necessitate more extensive DNA hypomethylation as well as activation of tissue-specific transcription factors.

There is currently such little information on the expression of CG genes in MPM. MAGE1--4, NY-ESO-1, GAGE1-2, GAGE1-6, SSX2, SSX1-6, and RAGE-1 expression in five MPM lines was compared to normal mesothelial cells employing RT-PCR techniques, according to Sigalotti et al. [71]. In these MPM lines, diverse expressions of the CG gene were identified, with each line exhibiting a unique profile, as previously reported for lung malignancies [72]. None of these genes were found in normal mesothelial cells [71, 73].

Advertisement

5. Polycom complex-mediated epigenetic silencing

Polycomb group proteins (PcG) play an important role as regulators of stem cell pluripotency and differentiation [74], as well as inappropriate gene expression during cancer transformation [75, 76]. In mammals, two main Polycomb repressor complexes (PRCs) have been discovered. PRC-2 is an initiating complex that causes trimethylation of histone 3 lysine and contains the subunits EZH1/EZH2, SUZ12, EED, and RBAP46/48 (H3K27Me3).

PCAF, PHC, RING1, CBX, and BMI1 are components of the housekeeping complex PRC-1, which mediates the ubiquitination of H2AK119 (H2AK119Ub). CRC recruitment and heterochromatin growth are aided by these histone marks, which are frequently detected in the context of DNA hypermethylation and gene suppression [75, 76]. Several proteins, such as including JARID2 and members of the sex comb-like family (ASXL), interact with EZH2 and SUZ12 to lead PRC-2 to polykyl response elements (PRE) throughout the genome [77, 78]. Goto et al. [79] investigated gene repression in MPM and observed that a subset of genes repressed in MPM had H3K27Me3 without DNA hypermethylation, implying that disruptions in polycomb gene expression may play a role in MPM etiology [80, 81].

Several immunoblotting investigations studies revealed that MPM cells overexpress EZH2 with associated increases in H3K27Me3 levels when compared to normal mesothelial cells. Another set of tests, which included QRT-PCR, immunoblotting, and IHC, revealed that EZH2 was overexpressed in almost 80% of primary MPMs (most of which were epithelioid histology). As a result of these findings, it was identified that EZH2 is overexpressed in MPM and that PRC-2 could be considered as a potential therapeutic target in these cancers. The overexpression of EZH2 in MPM was verified by TCGA analysis, as was a strong link between EZH2 upregulation and lower MPM patient survival (Figure 2A). Further TCGA analysis reveals that SUZ12 overexpression is associated with poor survival in MPM patients (Figure 2B). On the other hand, there is no evidence about MPM patients’ survival related to EED expression (Figure 2C).

The foregoing findings are especially important in light of recent findings that inactivating mutations in BRCA-associated protein 1 (BAP1), which encodes a nuclear ubiquitin hydrolase with several functions, are found in uncommon familial MPMs as well as almost 60% of sporadic MPMs. H2AK119Ub is ubiquitinated, for example.

LaFave et al. [82] discovered that BAP1 mutations, which are linked to protein expression loss, enhanced the expression of EZH2 and SUZ12 in MPM cells in a series of experiments. Likewise, overexpression of EZH2 was related to lower levels of H4K2Me1 and less occupancy of L3MBTL2 (an unusual polycomb protein that identifies this repressive histone mark) inside the EZH2 promoter in BAP1 mutant cells. Despite the strong connection between BAP1 mutations and repression of Polycomb stem cell targets, no specific clinical manifestation of BAP1 mutant MPM has been identified. Somatic mutations in BAP1 appear to be more common in current or past smokers with MPM [83].

Advertisement

6. SWI/SNF

SWI/SNF are mammalian homologs of yeast trithorax complexes. Their major purpose is to antagonize PRC-2’s repressive effects by destroying DNA-nucleosome connections allowing movement and ejection, or by switching nucleosomes to increase factor accessibility transcription to DNA [84, 85]. In human malignancies, the genes encoding the SWI/SNF complexes are commonly altered, with various subunit mutations related to specific cancer histologies.

Yoshikawa et al. [86] used whole exome sequencing to identify a substantial number of mutations in genes involved in the SWI/SNF pathways, including homozygous SMARCA4, ARID2, and PBRM1 mutations in short-term established MPM lines [86, 87]. They also evaluated at the loss of somatic copies in the 3p21 region (which is roughly 10.7 Mb in size and contains 251 genes) in 33 MPM samples, using techniques including comparative genomic matrix high-density hybridization (a-CGH) and next-generation targeted sequencing (NGS). Bi-allelic deletions (3 Kb) were observed in 46 genes, four of which have been associated to malignant tumors, including two SWI/SNF-related genes [PBRM1 (15%) and SMARCC1 (6%)], BAP1 (48%) and SETD2 (27%). More than 200 MPM were studied in a recent thorough genomic investigation.

Bueno et al. [88] described mutations in genes encoding SWI/SNF components in 8% of the samples, as well as mutations in two histone methyltransferases (SETDB1 and SETD5) in about 3% of the samples. The discrepancies between the results reported by Yoshikawa et al. [87] and Bueno et al. [88] may be attributable to the identification of minuscule deletions by high-density, a-CGH, and specific NGS that are not detectable by conventional NGS techniques. To establish final conclusions, more studies are needed to determine the frequency and clinical significance of SWI/SNF mutations in mesothelioma.

Advertisement

7. Epigenetics in the treatment of mesothelioma

MPM inhibits tumor suppressor genes by promoting LOI and repression of CG genes by site-specific hypermethylation of DNA and/or polycomb repressor complexes in the context of hypomethylation of the genome. This “DNA methylation paradox” mimics epigenomic conditions in normal germ cells and lays the groundwork for epigenetic regimens that restore tumor suppressor gene expression and trigger growth arrest/apoptosis. Upregulation of CTAs, development of viral mimicry by derepression of endogenous retroviruses, and control of the tumor microenvironment all help to boost antitumor immunity [89, 90].

DNMTs are potential targets for MPM treatment because of their direct functions in suppressing tumor suppressor genes and maintaining pluripotency [91, 92]. Previous clinical attempts to inhibit DNMT activity in MPM, however, have failed miserably. Yogelzang et al. [93] showed a 17% objective response rate in 41 MPM patients who received 120 h of continuous dihydro-5-azacytidine infusions. Amazingly, 6 years following treatment, the single responder was disease-free. The lack of efficacy of DNA hypomethylating drugs in solid tumors could be due to their usage at maximum tolerated doses, resulting in myelosuppression, rather than prolonged use at lower doses to obtain pharmacodynamic effects without systemic toxicity. The Phase I decitabine trial (DAC) clearly demonstrates that chronic exposures are required to achieve maximum gene induction effects in cancer tissues [94].

Furthermore, 5-AZA and DAC administered IV, SQ , or PO have short half-lives (less than 5 min) and poor biodistribution, limiting their potential utility in patients with solid tumors. Cytidine deaminase (CDA), which is found in practically all organs but mainly the gastrointestinal system, quickly inactivates these molecules [95, 96]. Documented toxicity increases Cmax and t1/2 (>50 nM and 4 h, respectively) as well as biodistribution of oral decitabine, decreasing inter-patient variability in drug levels significantly [95, 96, 97, 98]. Significant increases in fetal hemoglobin, without neutropenia, thrombocytopenia, or lymphopenia, are indicative of hypomethylation of systemic DNA caused by oral DAC-THU. A phase II trial (NCT02664181) is currently underway at the Cleveland Clinic and NCI to examine whether DAC/THU can improve responses to nivolumab when given as second-line therapy to patients with non-small cell lung cancer. Despite encouraging preclinical data [26], efforts to target HDAC on MPM have also been disappointing.

As second- or third-line therapy, Krug et al. [99] randomized 661 MPM patients to receive the HDAC inhibitor vorinostat or placebo. Overall survival, as well as the drug’s safety and tolerability, were the key outcomes. Vorinostat-treated patients had a median OS of 30.7 weeks (95% CI 26.7–36.1) compared to 27 weeks (95% CI 23.1–31.9) for placebo-treated patients. Given the absence of evidence for HDAC upregulation in MPM and the limited antitumor effects of HDAC inhibitors alone in preclinical tests, the lack of efficacy of the single-agent vorinostat in patients with MPM is not surprising. Combinated techniques, such as using HDAC inhibitors to sensitize cells to TRAIL-mediated apoptosis or flavopiridol to boost romidepsin-mediated growth arrest and death, might be helpful for future clinical trials. Hypomethylating drugs, on the other hand, do not appear to lessen the incidence of mesothelioma after asbestos exposure. In fact, non-solid cancers such leukemias, lymphomas, and other myelodysplastic syndromes show the best benefits with this medicine.

It is feasible that BAP1 mutations could be used for MPM therapy in the future. BAP1 promotes the recruitment of the polychial deubiquitinase PR-DUB complex to DNA damage sites by stabilizing BRCA-1 and promoting poly (ADP-Ribose) dependent recruitment of the polychial deubiquitinase PR-DUB complex to DNA damage sites. This activity is dependent on deubiquitinase activity and BAP1 phosphorylation. BAP1 mutations, which invariably show as a loss of function, cause BRCA-1 levels to drop and double-stranded DNA repair to be inhibited [100, 101, 102]. A BAP1 isoform including part of the catalytic domain sensitized MPM cells to the PARP1 inhibitor, according to Parotta et al. [102]. (Olaparib). Concomitant treatment with GDC0980, a dual PI3K-mTOR inhibitor that is downregulated by BRCA-1, could improve this sensitivity. These strategies could improve responses to cisplatin/pemetrexed in patients with BAP1 mutant MPM and should be evaluated in future clinical trials.

There is considerable interest in chromatographic remodeling agents with adoptive cell transfer or immune checkpoint inhibitors for cancer therapy, given the extensive preclinical studies showing DNA demethylating agents, HDAC inhibitors, and KMT inhibitors in the immunomodulatory effects of potentials [103]. In a syngeneic mouse tumor model, cytolytic T lymphocytes target testicular cancer antigen in vivo using decitabine to destroy metastatic cancer. The preclinical basis for combining gene induction regimens with cancer adoptive immunotherapy was established in these studies. Furthermore, novel microenvironmental data are likely to have a substantial impact on the outcomes of clinical trials for epigenetic treatments and immunotherapies [89].

Advertisement

8. Conclusions

While malignant pleural mesothelioma is a disease with a low incidence worldwide, with aggressive behavior, its survival does not go beyond 12 months once the diagnosis is made [1, 2]. Its origin has been related to the chronic exposure of asbestos as the main factor. Also, asbestos fibers have been an essential component in structural changes at the molecular level, with much evidence about its genetic behavior and to a lesser extent, its epigenetic behavior. All of this gives it a fairly heterogeneous behavior [13, 14, 15]. New molecular techniques allow a broader understanding of the carcinogenesis of this tumor and an approach to new diagnostic tools. Epigenetic dysregulations require active maintenance and are potentially reversible, making them a therapeutic target [7, 23, 30].

The study of methylome has made it possible to carry out differential diagnoses thanks to the methylation of some specific loci, such as TMEM30B, KAZAZD1, MAPK13 and to demonstrate greater survival rates in patients with low frequencies of methylations [16, 17, 28].

It is important to mention the exposure to asbestos fibers as the main resistance factor associated with the methylation of tumor suppressor genes seen in pleural mesothelial cells such as APC and RASSF1. Additionally, there are direct cellular effects such as chronic inflammation measured by free radicals leading to DNA oxidation, hemolysis with the release of hydroxyl ions, intrachain breakdown plus subsequent chromosomal fragmentation, and production of pro-inflammatory cytokines with higher expression of angiogenic growth factors, another aspect that can be considered a potential therapeutic objective. Genomic responses related to methylation conclude in a gene silencing, most likely in tumor suppressor genes such as SFRP4, FHIT, SLCA20 [69, 71, 80]. Another diagnostic approach that can be observed by methylation is the overexpression of DNMT in patients with MPM and consequently could be an attractive therapeutic target, however, clinical efforts for its inhibition have been disappointing and future studies should focus on the therapeutic approach to the inhibition of DNMT.

A greater association of methylation has been seen in advanced ages and ethnic groups such as the Japanese population. However, the greater association related to histological changes in proliferation, differentiation, invasion, and reduction of apoptosis has been seen with the increased methylation of CpG islands in genes such as CCND2, CDKN2A, and associated with asbestos bodies with RASSF1.

Although methylation is the most studied epigenetic mechanism, there are other modifications that lead to the silencing of tumor suppressor genes, such as the activation of the Polycomb complex and the mutation of the SWI/SNF pathway [82, 83]. Deacetylation mediated by HDAC has been seen in the p53 gene and other aspects such as HAT-mediated acetylation or demethylation by KDMs.

The modification in histone features such as stability in chromatin has a great relationship with HDCAs, thus making them a potential therapeutic target. There are few studies with inhibitors such as vorinostat, however, where there are no positive results due to the low expression in MPM.

Finally, it is clear that there is much to know about the modifications and/or epigenetic changes in MPM. The current evidence of the molecular mechanisms opens up another panorama for us to adjust personalized therapeutic strategies aimed at reversing normal changes and thus be able to identify in a timely manner those patients who are susceptible to such treatments. Therefore, clinical trials should focus on those epigenetic markers that at some point in their disease are overexpressed or silenced.

References

  1. 1. Kim MC, Kim NY, Seo YR, Kim Y. An integrated analysis of the genome-wide profiles of DNA methylation and mRNA expression defining the side population of a human malignant mesothelioma cell line. Journal of Cancer. 2016;7(12):1668-1679
  2. 2. Sage AP, Martinez VD, Minatel BC, et al. Genomics and epigenetics of malignant mesothelioma. High-Throughput. 2018;7(20):2-23
  3. 3. Feinberg AP, Ohlsson R, Henikoff S. The epigenetic progenitor origin of human cancer. Nature Reviews Genetics. 2006;7:21-33
  4. 4. Magee JA, Piskounova E, Morrison SJ. Cancer stem cells: Impact, heterogeneity, and uncertainty. Cancer Cell. 2012;21:283-296
  5. 5. Pietras A. Cancer stem cells in tumor heterogeneity. Advances in Cancer Research. 2011;112:255-282
  6. 6. Sekido Y. Molecular pathogenesis of malignant mesothelioma. Carcinogenesis. 2013;34(7):1413-1419
  7. 7. Heintz NH et al. Asbestos, lung cancers, and mesotheliomas: From molecular approaches to targeting tumor survival pathways. American Journal of Respiratory Cell and Molecular Biology. 2010;42:133-139
  8. 8. Toyokuni S. Mechanisms of asbestos-induced carcinogenesis. Nagoya Journal of Medical Science. 2009;71(1-2):1-10
  9. 9. Laird PW. Principles and challenges of genome-wide DNA methylation analysis. Nature Reviews Genetics. 2010;11:191-203
  10. 10. Jacinto FV, Ballestar E, Esteller M. Methyl-DNA immunoprecipitation (MeDIP): Hunting down the DNA methylome. BioTechniques. 2008;44:35
  11. 11. Blayney JK, Ceresoli GL, Castagneto B, O’Brien ME, Hasan B, et al. Response to chemotherapy is predictive in relation to longer overall survival in an individual patient combined-analysis with pleural mesothelioma. European Journal of Cancer. 2010;48:2983-2992
  12. 12. Vandermeers F, Neelature Sriramareddy S, Costa C, Hubaux R, Cosse JP, et al. The role of epigenetics in malignant pleural mesothelioma. Lung Cancer. 2013;81:311-318
  13. 13. Shukla A, Shukla A. Current therapies for malignant mesothelioma. Journal of Cancer Science and Therapy. 2014;6:306-309
  14. 14. Lee AY, He B, You L, et al. Expression of the secreted frizzled-related protein gene family is downregulated in human mesothelioma. Oncogene. 2004;23(39):6672-6676
  15. 15. Kohno H, Amatya VJ, Takeshima Y, et al. Aberrant promoter methylation of WIF-1 and SFRP1, 2, 4 genes in mesothelioma. Oncology Reports. 2010;24(2):423-431
  16. 16. Christensen BC, Houseman EA, Godleski JJ, et al. Epigenetic profiles distinguish pleural mesothelioma from normal pleura and predict lung asbestos burden and clinical outcome. Cancer Research. 2009;69(1):227-234
  17. 17. Goto Y, Shinjo K, Kondo Y, et al. Epigenetic profiles distinguish malignantpleural mesothelioma from lung adenocarcinoma. Cancer Research. 2009;69(23):9073-9082
  18. 18. Tomii K, Tsukuda K, Toyooka S, et al. Aberrant promoter methylation of insulin-like growth factor binding protein-3 gene in human cancers. International Journal of Cancer. 2007;120(3):566-573
  19. 19. Kimura K, Toyooka S, Tsukuda K, et al. The aberrant promoter methylation of BMP3b and BMP6 in malignant pleural mesotheliomas. Oncology Reports. 2008;20(5):1265-1268
  20. 20. Toyooka S, Pass HI, Shivapurkar N, et al. Aberrant methylation and simian virus 40 tag sequences in malignant mesothelioma. Cancer Research. 2001;61(15):5727-5730
  21. 21. Shivapurkar N, Toyooka S, Toyooka KO, et al. Aberrant methylation of trail decoy receptor genes is frequent in multiple tumor types. International Journal of Cancer. 2004;109(5):786-792
  22. 22. Tsou JA, Galler JS, Wali A, et al. DNA methylation profile of 28 potential marker loci in malignant mesothelioma. Lung Cancer. 2007;58(2):220-230
  23. 23. Christensen BC, Marsit CJ, Houseman EA, et al. Differentiation of lung adenocarcinoma, pleural mesothelioma, and nonmalignant pulmonary tissues using DNA methylation profiles. Cancer Research. 2009;69(15):6315-6321
  24. 24. Tsou JA, Shen LY, Siegmund KD, et al. Distinct DNA methylation profiles in malignant mesothelioma, lung adenocarcinoma, and non-tumor lung. Lung Cancer. 2005;47(2):193-204
  25. 25. Suzuki M, Toyooka S, Shivapurkar N, et al. Aberrant methylation profile of human malignant mesotheliomas and its relationship to SV40 infection. Oncogene. 2005;24(7):1302-1308
  26. 26. Paik PK, Krug LM. Histone deacetylase inhibitors in malignant pleural mesothelioma: Preclinical rationale and clinical trials. Journal of Thoracic Oncology. 2010;5(2):275-279
  27. 27. Dawson MA, Kouzarides T. Cancer epigenetics: From mechanism to therapy. Cell. 2012;150(1):12-27
  28. 28. Tessarz P, Kouzarides T. Histone core modifications regulating nucleosome structure and dynamics. Nature Reviews. Molecular Cell Biology. 2014;15:703-708
  29. 29. Jin B, Robertson KD. DNA methyltransferases, DNA damage repair, and cancer. Advances in Experimental Medicine and Biology. 2013;754:3-29
  30. 30. Baylin SB, Jones PA. Epigenetic determinants of cancer. Cold Spring Harbor Perspectives in Biology. 2016;8(9):a019505
  31. 31. Pruitt K. Molecular and cellular changes during cancer progression resulting from genetic and epigenetic alterations. Progress in Molecular Biology and Translational Science. 2016;144:3-47
  32. 32. Yin Y, Morgunova E, Jolma A, et al. Impact of cytosine methylation on DNA binding specificities of human transcription factors. Science. 2017;356:eaaj2239
  33. 33. Zhu H, Wang G, Qian J. Transcription factors as readers and effectors of DNA methylation. Nature Reviews. Genetics. 2016;17(9):551-565
  34. 34. Shimbo T, Wade PA. Proteins that read DNA methylation. Advances in Experimental Medicine and Biology. 2016;945:303-320
  35. 35. Yoon JH, Smith LE, Feng Z, et al. Methylated CpG dinucleotides are the preferential targets for G-to-T transversion mutations induced by benzo[a]pyrene diol epoxide in mammalian cells: Similarities with the p53 mutation spectrum in smoking-associated lung cancers. Cancer Research. 2001;61(19):7110-7117
  36. 36. Bochtler M, Kolano A, Xu GL. DNA demethylation pathways: Additional players and regulators. BioEssays. 2017;39:1-13
  37. 37. Yin X, Xu Y. Structure and function of TET enzymes. Advances in Experimental Medicine and Biology. 2016;945:275-302
  38. 38. An J, Rao A, Ko M. TET family dioxygenases and DNA demethylation in stem cells and cancers. Experimental & Molecular Medicine. 2017;49:323
  39. 39. Zoghbi HY, Beaudet AL. Epigenetics and human disease. Cold Spring Harbor Perspectives in Biology. 2016;8(2):a019497
  40. 40. Schomacher L, Niehrs C. DNA repair and erasure of 5-methylcytosine in vertebrates. BioEssays. 2017;39(3)
  41. 41. Schuermann D, Weber AR, Schar P. Active DNA demethylation by DNA repair: Facts and uncertainties. DNA Repair (Amst). 2016;44:92-102
  42. 42. Van Tongelen A, Loriot A, De Smet C. Oncogenic roles of DNA hypomethylation through the activation of cancer-germline genes. Cancer Letters. 2017;396:130-137
  43. 43. Audia JE, Campbell RM. Histone modifications and cancer. Cold Spring Harbor Perspectives in Biology. 2016;8(4):a019521
  44. 44. Ha K, Lee GE, Palii SS, et al. 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. Human Molecular Genetics. 2011;20:126-140
  45. 45. Cai Y, Tsai HC, Yen RC, et al. Critical threshold levels of DNA methyltransferase 1 are required to maintain DNA methylation across the genome in human cancer cells. Genome Research. 2017;27:533-544
  46. 46. Bagwe AN, Kay PH, Spagnolo DV. Evidence that DNA methylation imbalance is not involved in the development of malignant mesothelioma. Anticancer Research. 1997;17(5A):3341-3343
  47. 47. Zhao Y, Garcia BA. Comprehensive catalog of currently documented histone modifications. Cold Spring Harbor Perspectives in Biology. 2015;7(9):a025064
  48. 48. Wapenaar H, Dekker FJ. Histone acetyltransferases: Challenges in targeting bi-substrate enzymes. Clinical Epigenetics. 2016;8:59
  49. 49. Hyun K, Jeon J, Park K, et al. Writing, erasing and reading histone lysine methylations. Experimental & Molecular Medicine. 2017;49:e324
  50. 50. Yi X, Jiang XJ, Li XY, et al. Histone methyltransferases: Novel targets for tumor and developmental defects. American Journal of Translational Research. 2015;7:2159-2175
  51. 51. Filipp FV. Crosstalk between epigenetics and metabolism-Yin and Yang of histone demethylases and methyltransferases in cancer. Briefings in Functional Genomics. 2017;16(6):320-325
  52. 52. Liu F, Killian JK, Yang M, et al. Epigenomic alterations and gene expression profiles in respiratory epithelia exposed to cigarette smoke condensate. Oncogene. 2010;29:3650-3664
  53. 53. Kumar R, Li DQ , Muller S, et al. Epigenomic regulation of oncogenesis by chromatin remodeling. Oncogene. 2016;35:4423-4436
  54. 54. Längst G, Manelyte L. Chromatin remodelers: From function to dysfunction. Genes (Basel). 2015;6(2):299-324
  55. 55. Thapa B, Salcedo A, Lin X, et al. The immune microenvironment, genome-wide copy number aberrations, and survival in mesothelioma. Journal of Thoracic Oncology. 2017;12:850-859
  56. 56. Rinn JL, Chang HY. Genome regulation by long noncoding RNAs. Annual Review of Biochemistry. 2012;81:145-166
  57. 57. Rinn JL. lncRNAs: Linking RNA to chromatin. Cold Spring Harbor Perspectives in Biology. 2014;6(8):a018614
  58. 58. Meseure D, Drak Alsibai K, Nicolas A, et al. Long noncoding RNAs as new architects in cancer epigenetics, prognostic biomarkers, and potential therapeutic targets. BioMed Research International. 2015;2015:320214
  59. 59. Davidovich C, Cech TR. The recruitment of chromatin modifiers by long noncoding RNAs: Lessons from PRC2. RNA. 2015;21(12):2007-2022
  60. 60. Goto Y, Shinjo K, Kondo Y, et al. Epigenetic profiles distinguish malignant pleural mesothelioma from lung adenocarcinoma. Cancer Research. 2009;69:9073-9082
  61. 61. Foran E, Garrity-Park MM, Mureau C, et al. Upregulation of DNA methyltransferase-mediated gene silencing, anchorage-independent growth, and migration of colon cancer cells by interleukin-6. Molecular Cancer Research. 2010;8:471-481
  62. 62. Qureshi MA, Khan S, Tauheed MS, et al. Pan-cancer multiomics analysis of TC2N gene suggests its important Role(s) in tumourigenesis of many cancers. Asian Pacific Journal of Cancer Prevention. 2020;21(11):3199-3209
  63. 63. Gaudet F, Hodgson JG, Eden A, et al. Induction of tumors in mice by genomic hypomethylation. Science. 2003;300:489-492
  64. 64. Salmaninejad A, Zamani MR, Pourvahedi M, et al. Cancer/testis antigens: Expression, regulation, tumor invasion, and use in immunotherapy of cancers. Immunological Investigations. 2016;45:619-640
  65. 65. De Smet C, Loriot A. DNA hypomethylation and activation of germline-specific genes in cancer. Advances in Experimental Medicine and Biology. 2013;754:149-166
  66. 66. Simpson AJ, Caballero OL, Jungbluth A, et al. Cancer/testis antigens, gametogenesis and cancer. Nature Reviews. Cancer. 2005;5:615-625
  67. 67. Zendman AJ, Ruiter DJ, Van Muijen GN. Cancer/testis-associated genes: Identification, expression profile, and putative function. Journal of Cellular Physiology. 2003;194(3):272-288
  68. 68. Renaud S, Loukinov D, Alberti L, et al. BORIS/CTCFL-mediated transcriptional regulation of the hTERT telomerase gene in testicular and ovarian tumor cells. Nucleic Acids Research. 2011;39:862-873
  69. 69. Zhou J, Cheng T, Li X, et al. Epigenetic imprinting alterations as effective diagnostic biomarkers for early-stage lung cancer and small pulmonary nodules. Clin. Epigenetics. 2021;13(1):220
  70. 70. Loriot A, Reister S, Parvizi GK, et al. DNA methylation-associated repression of cancer-germline genes in human embryonic and adult stem cells. Stem Cells. 2009;27:822-824
  71. 71. Sigalotti L, Coral S, Altomonte M, et al. Cancer testis antigens expression in mesothelioma: Role of DNA methylation and bioimmunotherapeutic implications. British Journal of Cancer. 2002;86:979-982
  72. 72. Schrump DS. Targeting epigenetic mediators of gene expression in thoracic malignancies. Biochimica et Biophysica Acta. 2012;1819(7):836-845
  73. 73. Weiser TS, Guo ZS, Ohnmacht GA, et al. 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. Journal of Immunotherapy. 2001;24:151-161
  74. 74. Oliviero G, Brien GL, Waston A, et al. Dynamic protein interactions of the polycomb repressive complex 2 during differentiation of pluripotent cells. Molecular & Cellular Proteomics. 2016;15:3450-3460
  75. 75. Pasini D, Di Croce L. Emerging roles for Polycomb proteins in cancer. Current Opinion in Genetics & Development. 2016;36:50-58
  76. 76. Poynter ST, Kadoch C. Polycomb and trithorax opposition in development and disease. Wiley Interdisciplinary Reviews: Developmental Biology. 2016;5(6):659-688
  77. 77. Pasini D, Cloos PA, Walfridsson J, et al. JARID2 regulates binding of the Polycomb repressive complex 2 to target genes in ES cells. Nature. 2010;464:306-310
  78. 78. Katoh M. Functional proteomics of the epigenetic regulators ASXL1, ASXL2 and ASXL3: A convergence of proteomics and epigenetics for translational medicine. Expert Review of Proteomics. 2015;12(3):317-328
  79. 79. Mantsoki A, Devailly G, Joshi A. CpG island erosion, polycomb occupancy and sequence motif enrichment at bivalent promoters in mammalian embryonic stem cells. Scientific Reports. 2015;5:16791
  80. 80. Harikumar A, Meshorer E. Chromatin remodeling and bivalent histone modifications in embryonic stem cells. EMBO Reports. 2015;16(12):1609-1619
  81. 81. Kemp CD, Rao M, Xi S, et al. Polycomb repressor complex-2 is a novel target for mesothelioma therapy. Clinical Cancer Research. 2012;18:77-90
  82. 82. LaFave LM, Beguelin W, Koche R, et al. Loss of BAP1 function leads to EZH2-dependent transformation. Nature Medicine. 2015;21:1344-1349
  83. 83. Zauderer MG, Bott M, McMillan R, et al. Clinical characteristics of patients with malignant pleural mesothelioma harboring somatic BAP1 mutations. Journal of Thoracic Oncology. 2013;8:1430-1433
  84. 84. Kadoch C, Copeland RA, Keilhack H. PRC2 and SWI/SNF chromatin remodeling complexes in health and disease. Biochemistry. 2016;55(11):1600-1614
  85. 85. Pulice JL, Kadoch C. Composition and function of mammalian SWI/SNF chromatin remodeling complexes in human disease. Cold Spring Harbor Symposia on Quantitative Biology. 2016;81:53-60
  86. 86. Yoshikawa Y, Sato A, Tsujimura T, et al. Biallelic germline and somatic mutations in malignant mesothelioma: Multiple mutations in transcription regulators including mSWI/SNF genes. International Journal of Cancer. 2015;136:560-571
  87. 87. Yoshikawa Y, Emi M, Hashimoto-Tamaoki T, et al. High-density array-CGH with targeted NGS unmask multiple noncontiguous minute deletions on chromosome 3p21 in mesothelioma. Proceedings of the National Academy of Sciences of the United States of America. 2016;113:13432-13437
  88. 88. Bueno R, Stawiski EW, Goldstein LD, et al. Comprehensive genomic analysis of malignant pleural mesothelioma identifies recurrent mutations, gene fusions and splicing alterations. Nature Genetics. 2016;48:407-416
  89. 89. Wolff F, Leisch M, Greil R, et al. The double-edged sword of (re)expression of genes by hypomethylating agents: From viral mimicry to exploitation as priming agents for targeted immune checkpoint modulation. Cell Communication and Signaling: CCS. 2017;15:13
  90. 90. Kim K, Skora AD, Li Z, et al. Eradication of metastatic mouse cancers resistant to immune checkpoint blockade by suppression of myeloid-derived cells. Proceedings of the National Academy of Sciences of the United States of America. 2014;111:11774-11779
  91. 91. Pathania R, Ramachandran S, Elangovan S, et al. DNMT1 365 is essential for mammary and cancer stem cell maintenance and tumorigenesis. Nature Communications. 2015;6:6910
  92. 92. Yang J, Corsello TR, Ma Y. Stem cell gene SALL4 suppresses transcription through recruitment of DNA methyltransferases. The Journal of Biological Chemistry. 2012;287(3):1996-2005
  93. 93. Yogelzang NJ, Herndon JE 2nd, Cirrincione C, et al. Dihydro-5-azacytidine in malignant mesothelioma. A phase II trial demonstrating activity accompanied by cardiac toxicity. Cancer and Leukemia Group B Cancer. 1997;79:2237-2242
  94. 94. Schrump DS, Fischette MR, Nguyen DM, et al. Phase I study of decitabine-mediated gene expression in patients with cancers involving the lungs, esophagus, or pleura. Clinical Cancer Research. 2006;12:5777-5785
  95. 95. Chabner BA, Johns DG, Coleman CN, et al. Purification and properties of cytidine deaminase from normal and leukemic granulocytes. The Journal of Clinical Investigation. 1974;53:922-931
  96. 96. Lavelle D, Vaitkus K, Ling Y, et al. Effects of tetrahydrouridine on pharmacokinetics and pharmacodynamics of oral decitabine. Blood. 2012;119:1240-1247
  97. 97. Wentworth DF, Wolfenden R. On the interaction of 3,4,5,6-tetrahydrouridine with human liver cytidine deaminase. Biochemistry. 1975;14(23):5099-5105
  98. 98. Newman EM, Morgan RJ, Kummar S, et al. A phase I, pharmacokinetic, and pharmacodynamic evaluation of the DNA methyltransferase inhibitor 5-fluoro-2′-deoxycytidine, administered with tetrahydrouridine. Cancer Chemotherapy and Pharmacology. 2015;75:537-546
  99. 99. Krug LM, Kindler HL, Calvert H, et al. Vorinostat in patients with advanced malignant pleural mesothelioma who have progressed on previous chemotherapy (VANTAGE-014): A phase 3, double-blind, randomised, placebo-controlled trial. The Lancet Oncology. 2015;16:447-456
  100. 100. Ismail IH, Davidson R, Gagne JP, et al. Germline mutations in BAP1 impair its function in DNA double-strand break repair. Cancer Research. 2014;74:4282-4294
  101. 101. Fukuda T, Tsuruga T, Kuroda T, et al. Functional link between BRCA1 and BAP1 through histone H2A, heterochromatin and DNA damage response. Current Cancer Drug Targets. 2016;16:101-109
  102. 102. Yu H, Pak H, Hammond-Martel I, et al. Tumor suppressor and deubiquitinase BAP1 promotes DNA double-strand break repair. Proceedings of the National Academy of Sciences of the United States of America. 2014;111:285-290
  103. 103. Parrotta R, Okonska A, Ronner M, et al. A novel BRCA1-associated protein-1 isoform affects response of mesothelioma cells to drugs impairing BRCA1-mediated DNA repair. Journal of Thoracic Oncology. 2017;12(8):1309-1319

Written By

Aldo Manuel Alvarez Moran, Pablo Alejandro Ávila Sánchez, Jorge Alejandro Torres Ríos and Lorena Vega Castillo

Submitted: 29 November 2021 Reviewed: 13 May 2022 Published: 29 June 2022