Skip to main content

REVIEW article

Front. Genet., 04 March 2022
Sec. Epigenomics and Epigenetics
This article is part of the Research Topic Protein Modifications in Epigenetic Dysfunctional Diseases: Mechanisms and Potential Therapeutic Strategies View all 15 articles

DNA Methylation: An Important Biomarker and Therapeutic Target for Gastric Cancer

Yunqing ZengYunqing Zeng1Huimin RongHuimin Rong2Jianwei XuJianwei Xu3Ruyue CaoRuyue Cao1Shuhua LiShuhua Li1Yanjing GaoYanjing Gao1Baoquan ChengBaoquan Cheng1Tao Zhou
Tao Zhou4*
  • 1Department of Gastroenterology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
  • 2Department of Reconstructive Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
  • 3Department of Pancreatic Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
  • 4Department of Geriatric Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China

Gastric cancer (GC) is a very common malignancy with a poor prognosis, and its occurrence and development are closely related to epigenetic modifications. Methylation of DNA before or during gastric cancer is an interesting research topic. This article reviews the studies on DNA methylation related to the cause, diagnosis, treatment, and prognosis of gastric cancer and aims to find cancer biomarkers to solve major human health problems.

1 Introduction

Gastric cancer (GC) is a major unresolved clinical problem, with over a million new cases globally in 2020 (Sexton et al., 2020). It is the fourth most common cancer in men (Sexton et al., 2020), and the third most frequent cause of cancer-related deaths worldwide (Global, 2019). Most patients are diagnosed at an advanced stage and have a poor prognosis (Digklia and Wagner, 2016). Recently, multiple studies showed that epigenetic dysregulations, including DNA methylation, histone post-translational modifications, chromatin remodeling and non-coding RNAs, play a vital role in the oncogenesis and progression of GC. Among the epigenetic modifications mentioned, DNA methylation is the earliest known and well-investigated epigenetic change (Skvortsova et al., 2019).

The types of aberrant DNA methylation in human cancers include global DNA hypomethylation and local hypermethylation of genes. Genome-wide hypomethylation mainly occurs in repetitive elements that are normally hypermethylated to maintain genomic integrity and stability. Long Interspersed Nucleotide Element 1 (LINE-1), Alu repetitive elements and human endogenous retroviruses (HERVs) are the major constituents of interspersed repetitive sequences (IRS) (Chansangpetch et al., 2018). Regional hypermethylation of genes occurs in CpG (5′-cytosine-phosphate-guanine-3′) islands, which are normally unmethylated and cause silencing of tumor-suppressor genes, cell cycle regulator genes, and DNA repair genes (Eyvazi et al., 2019; Moore et al., 2013; Shao et al., 2018; Zeng et al., 2017). CpG islands (CGI) are frequently found in mammalian promoters (Field et al., 2018). Three types of DNA methyltransferases including DNMT1, DNMT3A, and DNMT3B, are responsible for DNA methylation. DNMT3A and DNMT3B are primarily ab initio methyltransferases, while DNMT1 maintains the methylation of symmetrically methylated CpGs during DNA duplication (Ebrahimi et al., 2020; Zeng et al., 2017).

Although many studies explored the prospect of DNA methylation as a biomarker with the aim of decreasing GC deaths, the methylation levels in those studies were mainly detected from the tissues by invasive methods. There are still many aberrantly methylated genes whose roles in GC have not been fully investigated, especially those detected by non-invasive methods. Novel non-invasive biomarkers are necessary for early detection, the prediction of prognosis and recurrence, and the evaluation of treatment efficacy. In this review, from the perspective of clinical practicality, we briefly described DNA methylation associated with pathogens of GC. Then, we highlighted the value of aberrant DNA methylation as a non-invasive biomarker for GC management.

2 DNA Methylation Associated With Pathogenesis

Pathogens invade host cells and cause epigenetic changes, such as DNA methylation, making it a safer environment for the pathogen. This allows the infection to persist and promotes the development of GC (Fattahi et al., 2018). The most important pathogens associated with gastric carcinogenesis are Helicobacter pylori (H. pylori, HP) and the Epstein-Barr virus (EBV).

2.1 Infection Mediated by Helicobacter pylori

Infection by H. pylori induces hypermethylation in the promoter regions of many DNA repair genes and tumor suppressor genes (MHL1, RUNX 3, APC, and PTEN), thus silencing the genes and facilitating carcinogenesis (Muhammad et al., 2019). Kosumi et al. found that LINE-1 hypomethylation of non-cancerous gastric mucosae in gastric cancer patients was significantly correlated with H. pylori infection (p = 0.037) and prospectively confirmed the similar result in non-gastric cancer patients (p = 0.010) (Kosumi et al., 2015). Yoshida et al. found that compared to the gastric mucosae of H. pylori-negative healthy volunteers, the Alu methylation level was significantly lower in the gastric mucosae of H. pylori-positive healthy volunteers and H. pylori-positive gastric cancer patients (Yoshida et al., 2011).

When chronic inflammation, triggered by H. pylori infection in Mongolian gerbils, was repressed by cyclosporin A, aberrant DNA methylation was substantially suppressed; however, the abundance of H. pylori in the gastric mucosa was not reduced. Therefore, it was concluded that the inflammation, rather than H. pylori, was responsible for inducing abnormal DNA methylation (Niwa et al., 2013). However, recurrent inflammation caused by alcohol or saturated NaCl did not induce abnormal DNA methylation (Hur et al., 2011). Helicobacter pylori infection activates the secretion of IL-1β and TNF-α and the production of reactive oxygen species. Together, they induce DNA methyltransferase 1 (DNMT1) and cause aberrant DNA methylation in gastric epithelial cells (Kim, 2019). Aggregation of aberrantly methylated DNA in the gastric mucosa might favor cancerogenesis (Maeda et al., 2017).

Although eliminating H. pylori significantly decreases methylation of tumor suppressor genes, DNA methylation does not return to the same level as that in individuals who are never infected by H. pylori, and the higher levels of methylated DNA in the previously infected individuals have adverse effects on the gastric mucosa in the long term (Nakajima et al., 2010). Therefore, individuals with ongoing presence of aberrant DNA methylation would face a higher risk of GC even after the eradication of H. pylori (Shin et al., 2012). From this perspective, determination of DNA methylation in H. pylori-negative subjects, including subjects whose H. pylori has been eliminated, can also act as a helpful diagnostic biomarker for assessing the risk of gastric cancer (Tahara and Arisawa, 2015).

2.2 EBV Infection

Based on The Cancer Genome Atlas (TCGA) project, gastric cancer was classified into four molecular subtypes: Epstein-Barr virus, microsatellite instability, genomically stable, and chromosomal instability (Sohn et al., 2017). Epstein-Barr virus-associated gastric carcinoma (EBVaGC), which comprises nearly 10% of gastric carcinomas (Fukayama and Ushiku, 2011), shows the most extreme DNA hypermethylation in all human malignancies (Usui et al., 2021). Matsusaka et al. divided GC into three epigenotypes according to DNA methylation patterns: EBV−/low methylation, EBV−/high methylation, and EBV+/high methylation (Matsusaka et al., 2011). CXXC4, TIMP2, and PLXND1 genes are methylated only in the EBV + tumors. COL9A2, EYA1, and ZNF365 genes are methylated both in EBV+ and EBV−/high tumors. AMPH, SORCS3, and AJAP1 genes are methylated in all gastric cancers (Matsusaka et al., 2011). Many studies have shown that promoter hypermethylation plays a crucial role in the carcinogenesis of EBVaGC (Kaneda et al., 2012; Matsusaka et al., 2014; Okada et al., 2013; Yau et al., 2014; Zong and Seto, 2014). EBVaGC exhibited promoter hypermethylation in multiple genes (e.g., p15, p16, p73, hMLH1, MGMT, GSTP1, CDH1, TIMP1, TIMP3, DAPK, bcl-2, APC, PTEN, and RASSF1A) associated with regulation of cell proliferation (Shinozaki-Ushiku et al., 2015). In contrast, the relationship between EBV infection and demethylation in interspersed DNA repeats is unclear.

Unlike HP infection, EBV infection-induced hypermethylation is considered to be caused not by the intermediate pathways related to inflammation induced by infection but by the pathogen itself (Kaneda et al., 2012). The molecular mechanism of aberrant DNA methylation triggered by EBV infection remains unclear. The proposed mechanism includes the upregulation of the expression of DNMT1 and DNMT3b and the downregulation of the activity of TET2 demethylase by EBV (Hino et al., 2009; Namba-Fukuyo et al., 2016; Zhao et al., 2013).

3 Diagnostic Biomarkers

Exploring the role of DNA methylation in early diagnosis of gastric cancer is important for reducing mortality. Most investigations of DNA methylation are based on the evaluation of disparities in methylation levels between the tumor and the adjacent tissues. Many studies have found that repetitive element hypomethylation and site-specific CpG island promoter hypermethylation are associated with increased risk of GC (Hu X et al., 2021; Min et al., 2017; Saito et al., 2012). In such cases, surgery is required to access the affected tissue, which is a major limitation for clinical application. Growing evidence has shown that methylation-related alterations in cancer patients arise systematically and might be measured in surrogate tissues (Yuasa, 2010). Developing non-invasive detection techniques is quite important for GC patients. Therefore, increasing researchers are devoted to exploring the clinical significance of aberrant DNA methylation detected in body fluids (including peripheral blood, gastric juice, etc.) and stool. Methylation of tumor suppressor genes in peripheral blood has been studied most extensively. In contrast, non-invasive tests related to genome-wide hypomethylation are performed less frequently. Aberrant methylation of multiple genes in body fluids and stool could be a valuable non-invasive biomarker for the early screening and diagnosis of gastric cancer (Qu et al., 2013; Yamamoto et al., 2020) (Table 1) (Figure 1).

TABLE 1
www.frontiersin.org

TABLE 1. Aberrant DNA methylation as diagnostic biomarkers in body fluids and stool of GC patients.

FIGURE 1
www.frontiersin.org

FIGURE 1. Biomarkers for diagnosis, therapy, and prognosis of gastric cancer. Hypermethylated (in red) and Hypomethylated (in blue) genes are shown. Sensitivity and specificity are shown in square brackets. GW: gastric washes. GJ: gastric juice. WBC: white blood cell. PPW: preoperative peripheral washes.

3.1 Biomarkers in Peripheral Blood

Various blood biomarker-based tests can be used for the early diagnosis of cancer (Huang et al., 2021). Tumor cells can release DNA into peripheral blood, causing abundant circulating DNA levels in the blood of cancer patients to be several-fold higher than that of individuals without cancer (Tahara and Arisawa, 2015). Increasing evidence indicates that the detection of methylated DNA in peripheral blood is more well-developed and stable than detection of gene mutation (Qi et al., 2016). Testing DNA methylation in the blood as a risk marker for carcinoma is of special significance, since the non-invasive and convenient collection of peripheral blood DNA is easily accepted by patients.

3.1.1 Circulating Cell-free DNA Methylation

Circulating cell-free DNA (cfDNA), derived from both normal and tumor cells, is present in the blood. In particular, the cfDNA that is derived from tumors and possesses tumor-specific mutations is called circulating tumor DNA (ctDNA) (Pessoa et al., 2020). Numerous studies have investigated the feasibility of measuring serum or plasma DNA methylation to detect methylation of tumor-derived circulating DNA as a latent diagnostic biomarker for gastric cancer (Tahara and Arisawa, 2015). Methylation of p16, CDH1, RARβ, Reprimo, Rassf1A, hMLH1, RUNX3, APC, E-cadherin, SFRP1, KCNA4, p15, SFRP2, HSULF1, PCDH10, TFPI2, TIMP3, CYP26B1, DAPK, DLEC1, FLNC, THBS1, UCHL1, GSTP1, HLTF, RPRML, SLC19A3, RNF180, etc. are markedly higher in the DNA from peripheral blood of GC subjects than in that of control subjects (Wang et al., 2015; Abbaszadegan et al., 2008; Alarcón et al., 2020; Balgkouranidou et al., 2013; Bernal et al., 2008; Chen et al., 2015a; Chen et al., 2009; Cheng et al., 2007; Cheung et al., 2012; Guo et al., 2010, 2011,; Hibi et al., 2011; Huang et al., 2021; Ikoma et al., 2006; Kanyama et al., 2003; Lee et al., 2002; Leung et al., 2005; Liu et al., 2015; Lu et al., 2012; Miao et al., 2020; Ng et al., 2011; Pimson et al., 2016; Sakakura et al., 2009; Tahara et al., 2013; Wang et al., 2008; Xue et al., 2016; Zhang et al., 2010, 2014; Chen et al., 2015b; Zheng et al., 2011b). Lin et al. evaluated the methylation state of three genes (ZIC1, HOXD10, and RUNX3) from the blood samples of GC patients using methylation-specific polymerase chain reaction. They discovered that the Odds ratios of ZIC1, HOXD10, and RUNX3 methylation for predicting GC were 4.285 (95%CI: 2.435–7.542), 3.133 (95%CI: 1.700–5.775), and 2.674 (95%CI: 1.441–4.960), respectively. The joint detection sensitivity of these three genes was 91.6%. Therefore, the combined detection of multiple gene promoter hypermethylation exhibited a cooperative effect compared to a single biomarker used to predict GC (Lin et al., 2017).

What is noteworthy is that even for the same methylated gene, there are significant discrepancies between the results of different studies, which might be attributed to differences in the sample size, detection methods, and study regions (Huang et al., 2021). To understand this heterogeneity and evaluate the accuracy of DNA methylation markers in the blood for identifying gastric cancer patients, Hu et al. conducted a meta-analysis with 32 studies, containing 69 analyses of blood DNA methylation tests that were conducted to evaluate GC. The 32 studies included 2,098 GC patients and 2,098 control subjects. The blood test based on DNA methylation had an overall sensitivity of 57% and specificity of 97% for gastric cancer. Plasma-based tests showed a sensitivity of 71% and specificity of 89%. Serum-based tests showed a sensitivity of 50% and specificity of 98%. The sensitivity of using multiple methylation genes was 76% and specificity was 85%. These results suggested that the blood-based DNA methylation test has high specificity but moderate sensitivity for the detection of gastric cancer. The determination of various methylation genes or the use of plasma samples might improve the sensitivity of the diagnosis (Hu et al., 2017).

3.1.2 DNA Methylation in Peripheral Blood Leukocytes

Unlike tumor DNA, leukocyte DNA can be obtained non-invasively and relatively inexpensively (Tahara et al., 2018). Studies which determine whether selected tumor suppressor genes and genome-wide repetitive sequence methylation in peripheral blood leukocytes of subjects with gastric cancer and healthy controls are different are rapidly emerging. Hypermethylation of KIBRA, DLEC1, FAT4, WT1, H19, MALAT1, APC, ACIN1, BCL2, CD44, TNFRSF10C and RARB promoters in peripheral blood leukocytes was found to be statistically significant in GC patients (Dauksa et al., 2014; Hu D. et al., 2021; Sun et al., 2018; Xie et al., 2020; Zhang et al., 2018; Zhou et al., 2019). To date, only a few studies have examined GC risk associated with Alu and LINE-1 methylation in peripheral blood leukocytes and the results are variable. Dauksa et al. found that the mean methylation level in Alu and LINE-1 repeats of GC patients was slightly lower than the mean level in the controls (Dauksa et al., 2014). Hou et al. demonstrated that GC risk increased with a decrease in the methylation of Alu or LINE-1, although the trends were not statistically significant (Hou et al., 2010). Gao et al. found that Alu methylation in blood leukocyte DNA was inversely associated with GC risk, but LINE-1 methylation levels were not correlated with GC risk (Gao et al., 2012). Barchitta et al. also showed that the LINE-1 methylation levels were significantly different in tissue samples but not in blood samples (Barchitta et al., 2014). These results suggested that studies with more individuals must be performed to determine the clinical applicability of leukocyte DNA methylation to detect gastric cancer non-invasively.

3.1.3 DNA Methylation in Whole Blood

Several studies evaluated the association of aberrant DNA methylation with the risk of gastric cancer. SOCS3, SPG20, and SFRP1 promoter hypermethylation in whole blood significantly increased GC risk (Han et al., 2020; Tahara et al., 2013; Zhang et al., 2014).

3.2 Biomarkers in Gastric Washes

Since a large number of mucosal cells could be extracted from gastric juice (GJ)), DNA biomarkers in gastric juice might be used to detect gastric cancer. However, DNA is easily degraded in an acidic environment; thus, gastric wash (GW) is used as an alternative source for determining aberrant DNA methylation (Yamamoto et al., 2020). Unfortunately, only several early studies showed that the methylation levels of MINT25, RORA, GDNF, ADAM23, PRDM5, CDH1, and MLF1 in gastric washes of GC patients were significantly higher than those of control subjects (Muretto et al., 2008; Watanabe et al., 2009). Among them, MINT25 methylation has the optimal sensitivity (90.0%) and specificity (95.8%), and thus, can distinguish GC from non-GC and be a potential biomarker for screening GC. Yoshiyuki et al. investigated the relationship of the methylation levels between biopsy and gastric washes. The methylation levels of all six genes were tightly associated by statistical analysis (MINT25: p = 0.001; RORA: p = 0.03; PRDM5: p < 0.001; MLF1: p < 0.001; ADAM23 p < 0.001; GDNF: p < 0.001). These results indicated that DNA from gastric washes can be used as a suitable substitute for DNA from biopsied tissues to determine the accumulation of DNA methylation in GC patients (Watanabe et al., 2009).

Hypermethylation of BARHL2 was detected in gastric wash-derived and gastric juice-derived exosomal DNA in early-stage GC patients before endoscopic treatment, whereas methylation levels considerably decreased with a curative endoscopic therapy. These results indicated that BARHL2 methylation might contribute to the detection of residual cancer after endoscopic resection and the potential prediction of tumor relapse (Yamamoto et al., 2020). Some disturbing factors such as aging, HP infection, and chronic inflammation can also induce aberrant DNA methylation. BARHL2 methylation is not affected by those factors. Therefore, GW or GJ exoDNA-based methylation analysis of BARH2 is expected to be an accurate biomarker for detecting early and advanced gastric cancer (Yamamoto et al., 2016).

3.3 Biomarkers in Stool

Guo et al. (Guo et al., 2021) evaluated the feasibility of gene methylation in feces for screening gastric cancer. All GC patients and normal controls were divided into training sets and test sets. The sensitivity and specificity of a single marker for gastric cancer detection in the training set for SDC2 were 40.9 and 93.3%, for TERT were 36.4 and 90.0%, for RASSF2 were 31.8 and 93.3%, for SFRP2 were 22.7 and 90.0%, and for Hb were 27.3 and 90.0%. The sensitivity and specificity of the three markers for methylation of SDC2, TERT, and Hb in the test set for gastric cancer detection were 40.9 and 91.7%, 34.1 and 91.7%, and 25.0 and 86.7%, respectively. The results showed that the methylation of SDC2, SFRP2, TERT, and RASSF2 has a certain sensitivity and high specificity in GC screening, which is a potential fecal biomarker of gastric cancer. Another study (Liu et al., 2017) also suggested the feasibility of stool TERT promoter methylation analyses for the non-invasive screening of gastric cancer.

4 Therapeutic Target

DNA methylation is also critical for the adjuvant treatment of gastric cancer. Chemotherapy is one of the major methods for treating GC. The main problem with chemotherapy is drug resistance, which is primarily related to DNA methylation. Correcting aberrant methylation patterns can improve chemotherapy response and patient survival (Housman et al., 2014). Animal studies have also shown that direct repression of aberrant DNA methylation can inhibit gastric carcinogenesis (Maeda et al., 2017). Therefore, DNMT inhibitors (DNMTi) are being actively investigated as novel cancer treatments. Additionally, adjuvant radiotherapy of GC has been debated over the past few decades. However, it has been suggested that hypermethylation and inactivation of certain genes associated with cell cycle regulation, DNA repair, apoptosis, and signal transduction can lead to radiotherapy resistance in GC cells (Zhang et al., 2006). In the future, it may be possible to improve radiotherapy response by altering DNA methylation patterns to benefit GC patients.

4.1 Chemotherapy

Studies have shown that DNA methylation in gastric cancer cells is related to the sensitivity of chemotherapy and resistance of anticancer agents such as 5-FU and cisplatin. The biomarkers used to identify resistance or sensitivity to chemotherapeutic drugs can be investigated. Hypermethylated TFAP2E, TMS1, PYCARD (ASC/TMS1), and DAPK might be appropriate biomarkers for 5-FU-resistant gastric cancer. Hypermethylated CDKN2A (p16INK4a) and DCTPP1 might be useful biomarkers for 5-FU-sensitive gastric cancer. Hypomethylated ADGRL2 (LPHN2) and GTSE1 are potential biomarkers of cisplatin-sensitive gastric cancer. Regardless of the type of drug, the hypomethylated ATP-binding cassette gene B1 (ABCB1) could be an effective biomarker for chemotherapy-resistant gastric cancer (Choi et al., 2017) (Figure 1). This is because ABCB1 hypermethylation silences genes that encode cellular factors necessary for cancer cell resistance to the chemotherapeutic drugs 5-FU and cisplatin (Shitara et al., 2010).

4.2 DNMT Inhibitors

DNMT inhibitors are either nucleoside analogs or non-nucleoside analogs (Erdmann et al., 2015). Azacytidine and decitabine are nucleoside analogs of cytosine that cannot accept a methyl donor at the 5′ position of the pyrimidine ring and depletes cellular DNMT1 (Zeng et al., 2017). Decitabine is integrated into DNA instead of cytidine during duplication, and azacitidine can be incorporated directly into RNA, inhibiting protein synthesis, which causes a substantial reduction in DNMT activity (Navada et al., 2014). Azacitidine was found to suppress the proliferation of GC cell lines and alter DNA methylation (Chen & Wang et al., 2015). Decitabine treatment can cause growth inhibition and reduction in DNMT3A and DNMT3B levels, accompanied by demethylation of the P16 INK4A gene (Liu et al., 2013). Zebularine, another kind of nucleoside analog, is a novel DNMT inhibitor that reduces the expression of the DNMT protein and reactivates epigenetically silenced genes (Tan et al., 2013).

At present, neither azacytidine nor decitabine has been identified as monotherapy for gastric cancer in the clinical setting. This might be because DNMT inhibitors alone cannot reactivate gene expression (Si et al., 2010). However, growing evidence suggests that the combination of DNMT inhibitors and traditional chemotherapy can improve chemosensitization by restoring aberrant epigenetic changes. For example, the combined administration of decitabine and 5-FU showed that TFAP2E is reactively expressed in GC by demethylation and an increase in chemosensitization (Wu F. L. et al., 2015). Additionally, azacytidine upregulates DAPK2, DAPK3, RASSF1, and THBS1 genes which might synergize with chemotherapeutic agents (Tan et al., 2013; Wu F et al., 2015; Zhang et al., 2006). In a clinical trial (Phase 1) (Schneider et al., 2017), researchers investigated pre-treatment with 5-azacitidine as a demethylation reagent in late-stage gastric cancer. They used 5-azacytidine (V) before EOX (epirubicin, oxaliplatin, capecitabine) neoadjuvant chemotherapy in GC patients and the result showed hypomethylation of tumor-related loci such as HPP1, TIMP3, CDKN2A, ESR1, and MGMT. Most patients easily tolerate neoadjuvant VEOX therapy with significant clinical and epigenetic responses. More randomized studies are required to further determine whether the efficacy of this combination is better than chemotherapy alone.

Other types of non-nucleoside analogs such as procaine, hydrazone-gallate, genistein, miRNA-21, miRNA-335, miRNA-148a, and miRNA-155-5p have also been investigated, but further studies need to be performed before approval for clinical use (Erdmann et al., 2016; Li et al., 2014, 2018,; Pan et al., 2010; Xie et al., 2014; Zhang et al., 2015; Zuo et al., 2013).

4.3 Radiotherapy

Despite active anticancer treatment, the overall prognosis of advanced GC is not ideal. Hence, an effective biomarker is required for selecting suitable patients who might benefit from adjuvant radiotherapy. Unfortunately, such predictive indicators have not been determined.

An et al. (An et al., 2020) analyzed methylation maps of 397 gastric cancer samples downloaded from The Cancer Genome Atlas (TCGA) and established a new biomarker called promoter methylation burden of DNA repair genes (RPMB), which meant the ratio of methylated DNA repair genes to the number of all DNA repair genes in order to identify patients who were sensitive to radiotherapy. Subgroup analyses based on overall survival (OS) and disease-free survival (DFS) showed that most of the subgroups tended toward the high-RMPB group. High-RPMB patients receiving radiotherapy with both ≥ T2 tumor and positive lymph nodes showed longer DFS than the low -RPMB group (p = 0.010). High-RPMB patients receiving radiotherapy with both ≥ T2 tumor and positive lymph nodes survived low-RPMB patients in disease-free status (p = 0.010). Therefore, RPMB might be a promising biomarker to evaluate the indications for adjuvant radiotherapy in GC. Furthermore, treatment with 5-aza-CdR can positively affect radiotherapy sensitivity of gastric cancer cells by enhancing the expression of some genes such as p53, RASSF1, and DAPK (Qiu et al., 2009).

5 Prognostic Biomarkers

Aberrant DNA methylation in peripheral blood is also related to multiple prognostic results of gastric cancer. Therefore, it could be used as a prognostic biomarker of GC (Figure 1). Hypermethylation of most genes such as APC, E-cadherin, UCHL1, SPG20, RASSF1A, SFRP2, CDH1, THBS1, SOX17, Wif-1, RNF-180, MED12L, HMLH1, MGMT, FLNC, LOX, HOXD10, BNIP3, and PCDH10 was significantly associated with adverse prognosis in GC (Balgkouranidou et al., 2015; Cheung et al., 2012; Hu X et al., 2021; Ikoma et al., 2006; Karamitrousis et al., 2021; Necula et al., 2019; Pimson et al., 2016; Wang et al., 2015; Yan et al., 2021; Yu et al., 2012; Zhang et al., 2014) (Table 2). Additionally, Xie et al. found that PBX3 methylation in peripheral blood leukocytes was associated with poorer GC prognosis only in the elderly group (HR = 1.678, 95% CI = 1.046–2.693) and the female group (HR = 2.058, 95% CI = 1.024–4.137) (Xie et al., 2020).

TABLE 2
www.frontiersin.org

TABLE 2. Aberrantly methylated genes as prognostic biomarkers in GC patients.

Ko et al. evaluated the prognostic value of LINE-1 methylation level in cfDNA in gastric cancer patients undergoing radical surgery and chemotherapy. The overall survival (OS) of patients with low methylation levels before starting treatment was significantly worse than those with high methylation levels. But methylation level before surgery had no effect on recurrence-free survival (RFS) and OS (Ko et al., 2021). However, another study showed that the methylation status of LINE-1 in leukocyte DNA was not an independent prognostic factor of GC (Tahara et al., 2018).

The association between aberrant DNA methylation and the prognosis of GC needs to be evaluated in larger cohorts and diverse populations. Additionally, more intensive studies are required to determine the potential molecular biomarkers for predicting prognosis in GC patients.

6 Conclusion

Helicobacter pylori and EBV are the most important pathogens associated with gastric cancer, which can cause carcinogenesis by inducing aberrant DNA methylation. DNA methylation has high clinical application value. Aberrant methylation of various genes in body fluids and feces can be used as a non-invasive method for early screening and diagnosis of gastric cancer. Specifically, Reprimo, RUNX3, PCDH10, BARHL2, and MINT25 hypermethylation have both high sensitivity and specificity, which indicates their value in the diagnosis of GC. However, the sensitivity of detecting other types of DNA methylation from peripheral blood and stool is not satisfactory. Using a combination of multiple genes can yield higher sensitivity. DNA methylation can also affect the response to chemoradiotherapy in gastric cancer patients. A combination of DNMT inhibitors and chemotherapy drugs seems to have a better therapeutic effect. Therefore, more DNMT inhibitors that have lower toxicity, an effective response, and a low price need to be developed. Furthermore, DNA methylation can predict a variety of prognostic results for GC patients, such as overall survival (OS) and disease-free survival (DFS). Aberrant methylation of APC, SFRP2, LINE-1, E-cadherin, SOX17, Wif-1, RASSF1A, RNF-180, UCHL1, and SPG20 in peripheral blood was significantly associated with shorter OS in GC. The methylation levels of SFRP2, SOX17, Wif-1, and RASSF1A in peripheral blood had an impact on progression-free survival (PFS). THBS1 methylation in the serum and CDH1 methylation in preoperative peripheral washes (PPW) were related to worse DFS. Additionally, aberrantly methylated genes such as SFRP2, THBS1, UCHL1, SOX17, APC, E-cadherin, RASSF1A, RNF-180, and SPG20 could play an important role in both diagnosis and prognosis of GC (Table 3). New tests with improved sensitivity, simplicity, standardization, and cost-effectiveness need to be developed, and new biomarkers need to be validated in larger prospective clinical studies.

TABLE 3
www.frontiersin.org

TABLE 3. Aberrantly methylated genes that play an important role in both diagnosis and prognosis of GC.

Author Contributions

YZ designed the study, searched the literature, and wrote the manuscript. HR designed the study and searched the literature. JX contributed to manuscript revision. RC and SL searched the literature. YG, BC, and TZ contributed to thesis supervision. All authors approved the final version of the manuscript.

Funding

This work was supported by the Key Technology Research and Development Program of Shandong Province (2019GSF108254).

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s Note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors, and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

Abbaszadegan, M. R., Moaven, O., Sima, H. R., Ghafarzadegan, K., A'Rabi, A., Forghani, M. N., et al. (2008). P16 Promoter Hypermethylation: A Useful Serum Marker for Early Detection of Gastric Cancer. World J. Gastroenterol. 14 (13), 2055–2060. doi:10.3748/wjg.14.2055

CrossRef Full Text | Google Scholar

Alarcón, M. A., Olivares, W., Córdova-Delgado, M., Muñoz-Medel, M., de Mayo, T., Carrasco-Aviño, G., et al. (2020). The Reprimo-like Gene Is an Epigenetic-Mediated Tumor Suppressor and a Candidate Biomarker for the Non-invasive Detection of Gastric Cancer. Int. J. Mol. Sci. 21 (24). doi:10.3390/ijms21249472

CrossRef Full Text | Google Scholar

An, N., Yu, Z., He, X. J., Zhao, Y. Y., Yu, L., Zhang, Y. C., et al. (2020). Promoter Methylation of DNA Repair Genes Predicts Disease-free Survival of Gastric Adenocarcinoma after Adjuvant Radiotherapy. Mol. Ther. Oncolytics 18, 109–117. doi:10.1016/j.omto.2020.06.006

PubMed Abstract | CrossRef Full Text | Google Scholar

Balgkouranidou, I., Karayiannakis, A., Matthaios, D., Bolanaki, H., Tripsianis, G., Tentes, A. A., et al. (2013). Assessment of SOX17 DNA Methylation in Cell Free DNA from Patients with Operable Gastric Cancer. Association with Prognostic Variables and Survival. Clin. Chem. Lab. Med. 51(7), 1505–1510. doi:10.1515/cclm-2012-0320

PubMed Abstract | CrossRef Full Text | Google Scholar

Balgkouranidou, I., Matthaios, D., Karayiannakis, A., Bolanaki, H., Michailidis, P., Xenidis, N., et al. (2015). Prognostic Role of APC and RASSF1A Promoter Methylation Status in Cell Free Circulating DNA of Operable Gastric Cancer Patients. Mutat. Research/Fundamental Mol. Mech. Mutagenesis 778, 46–51. doi:10.1016/j.mrfmmm.2015.05.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Barchitta, M., Quattrocchi, A., Maugeri, A., Vinciguerra, M., and Agodi, A. (2014). LINE-1 Hypomethylation in Blood and Tissue Samples as an Epigenetic Marker for Cancer Risk: A Systematic Review and Meta-Analysis. [Journal Article; Meta-Analysis; Review; Systematic Review. PLoS One 9 (10), e109478. doi:10.1371/journal.pone.0109478

PubMed Abstract | CrossRef Full Text | Google Scholar

Bernal, C., Aguayo, F., Villarroel, C., Vargas, M., Díaz, I., Ossandon, F. J., et al. (2008). Reprimo as a Potential Biomarker for Early Detection in Gastric Cancer. [Journal Article; Research Support, Non-U.S. Gov't]. Clin. Cancer Res. 14 (19), 6264–6269. doi:10.1158/1078-0432.CCR-07-4522

PubMed Abstract | CrossRef Full Text | Google Scholar

Chansangpetch, S., Prombhul, S., Tantisevi, V., Sodsai, P., Manassakorn, A., Hirankarn, N., et al. (2018). DNA Methylation Status of the Interspersed Repetitive Sequences for LINE-1, Alu, HERV-E, and HERV-K in Trabeculectomy Specimens from Glaucoma Eyes. J. Ophthalmol. 2018, 9171536. doi:10.1155/2018/9171536

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, X., Lin, Z., Xue, M., Si, J., and Chen, S. (2015a). Zic1 Promoter Hypermethylation in Plasma DNA Is a Potential Biomarker for Gastric Cancer and Intraepithelial Neoplasia. PLoS One 10 (7), e133906. doi:10.1371/journal.pone.0133906

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, X., Wang, F., Li, J., He, X., Liu, X., and Ma, L. (2015b). The Effect of Two Nucleoside Antitumor Drugs on the Proliferation and DNA Methylation of Human Gastric Cancer Cells. Oncol. Lett. 10 (3), 1919–1923. doi:10.3892/ol.2015.3427

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, Z., Fan, J. Q., Li, J., Li, Q. S., Yan, Z., Jia, X. K., et al. (2009). Promoter Hypermethylation Correlates with the Hsulf-1 Silencing in Human Breast and Gastric cancerResearch Support, Non-U.S. Gov't]. [Journal Articleint. J. Cancer 124 (3), 739–744. doi:10.1002/ijc.23960

CrossRef Full Text | Google Scholar

Cheng, Y. Y., Yu, J., Wong, Y. P., Man, E. P., To, K. F., Jin, V. X., et al. (2007). Frequent Epigenetic Inactivation of Secreted Frizzled-Related Protein 2 (SFRP2) by Promoter Methylation in Human Gastric cancerResearch Support, Non-U.S. Gov't]. [Journal Articlebr J. Cancer 97 (7), 895–901. doi:10.1038/sj.bjc.6603968

CrossRef Full Text | Google Scholar

Cheung, K. F., Lam, C. N., Wu, K., Ng, E. K., Chong, W. W., Cheng, A. S., et al. (2012). Characterization of the Gene Structure, Functional Significance, and Clinical Application of RNF180, a Novel Gene in Gastric Cancer. Cancer-am. Cancer Soc. 118 (4), 947–959. doi:10.1002/cncr.26189

PubMed Abstract | CrossRef Full Text | Google Scholar

Choi, S. J., Jung, S. W., Huh, S., Chung, Y. S., Cho, H., and Kang, H. (2017). Alteration of DNA Methylation in Gastric Cancer with Chemotherapy. J. Microbiol. Biotechnol. 27 (8), 1367–1378. doi:10.4014/jmb.1704.04035

PubMed Abstract | CrossRef Full Text | Google Scholar

Dauksa, A., Gulbinas, A., Endzinas, Z., Oldenburg, J., and El-Maarri, O. (2014). DNA Methylation at Selected CpG Sites in Peripheral Blood Leukocytes Is Predictive of Gastric Cancer. Anticancer Res. 34 (10), 5381–5388.

PubMed Abstract | Google Scholar

Digklia, A., and Wagner, A. D. (2016). Advanced Gastric Cancer: Current Treatment Landscape and Future Perspectives. [Journal Article; Review]world J. Gastroenterol. 22 (8), 2403–2414. doi:10.3748/wjg.v22.i8.2403

CrossRef Full Text | Google Scholar

Ebrahimi, V., Soleimanian, A., Ebrahimi, T., Azargun, R., Yazdani, P., Eyvazi, S., et al. (2020). Epigenetic Modifications in Gastric Cancer: Focus on DNA Methylation. Gene 742, 144577. doi:10.1016/j.gene.2020.144577

PubMed Abstract | CrossRef Full Text | Google Scholar

Erdmann, A., Halby, L., Fahy, J., and Arimondo, P. B. (2015). Targeting DNA Methylation with Small Molecules: What’s Next? J. Med. Chem. 58 (6), 2569–2583. doi:10.1021/jm500843d

CrossRef Full Text | Google Scholar

Erdmann, A., Menon, Y., Gros, C., Masson, V., Aussagues, Y., Ausseil, F., et al. (2016). Identification and Optimization of Hydrazone-Gallate Derivatives as Specific Inhibitors of DNA Methyltransferase 3A. Future Med. Chem. 8 (4), 373–380. doi:10.4155/fmc.15.192

PubMed Abstract | CrossRef Full Text | Google Scholar

Eyvazi, S., Khamaneh, A. M., Tarhriz, V., Bandehpour, M., Hejazi, M. S., Sadat, A. T. E., et al. (2020). CpG Islands Methylation Analysis of CDH11, EphA5, and HS3ST2 Genes in Gastric Adenocarcinoma Patients. J. Gastrointest. Cancer. 51 (2), 579–583. doi:10.1007/s12029-019-00290-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Fattahi, S., Kosari-Monfared, M., Ghadami, E., Golpour, M., Khodadadi, P., Ghasemiyan, M., et al. (2018). Infection-associated epigenetic alterations in gastric cancer: New insight in cancer therapy. J. Cell. Physiol. 233 (12), 9261–9270. doi:10.1002/jcp.27030

PubMed Abstract | CrossRef Full Text | Google Scholar

Field, A. E., Robertson, N. A., Wang, T., Havas, A., Ideker, T., and Adams, P. D. (2018). DNA Methylation Clocks in Aging: Categories, Causes, and Consequences. Mol. Cel. 71 (6), 882–895. doi:10.1016/j.molcel.2018.08.008

PubMed Abstract | CrossRef Full Text | Google Scholar

Fukayama, M., and Ushiku, T. (2011). Epstein-Barr Virus-Associated Gastric Carcinoma. Pathol. Res. Pract. 207 (9), 529–537. doi:10.1016/j.prp.2011.07.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Gao, Y., Baccarelli, A., Shu, X. O., Ji, B. T., Yu, K., Tarantini, L., et al. (2012). Blood Leukocyte Alu and LINE-1 Methylation and Gastric Cancer Risk in the Shanghai Women's Health Study[Journal Article; Research Support. Br. J. Cancer 106 (3), 585–591. doi:10.1038/bjc.2011.562

PubMed Abstract | CrossRef Full Text | Google Scholar

Global, B. O. D. C. (2019). Global, Regional, and National Cancer Incidence, Mortality, Years of Life Lost, Years Lived with Disability, and Disability-Adjusted Life-Years for 29 Cancer Groups, 1990 to 2017: A Systematic Analysis for the Global burden of Disease Study. JAMA Oncol. 5 (12), 1749–1768. doi:10.1001/jamaoncol.2019.2996

PubMed Abstract | CrossRef Full Text | Google Scholar

Guo, P., Zhou, X. J., Xu, L., Chen, H., Zhao, L., Sun, M. H., et al. (2021). [Application of Fecal DNA Methylation Biomarkers Detection in Gastric Cancer Screening. Zhonghua Yi Xue Za Zhi 101 (11), 808–812. doi:10.3760/cma.j.cn112137-20200916-02659

PubMed Abstract | CrossRef Full Text | Google Scholar

Guo, W., Dong, Z., Guo, Y., Chen, Z., Kuang, G., and Yang, Z. (2011). Aberrant Methylation of the CpG Island of HLTF Gene in Gastric Cardia Adenocarcinoma and dysplasiaResearch Support, Non-U.S. Gov't]. [Journal Articleclin. Biochem. 44 (10-11), 784–788. doi:10.1016/j.clinbiochem.2011.04.006

CrossRef Full Text | Google Scholar

Guo, X., Liu, W., Pan, Y., Ni, P., Ji, J., Guo, L., et al. (2010). Homeobox Gene IRX1 Is a Tumor Suppressor Gene in Gastric Carcinoma. Oncogene 29 (27), 3908–3920. doi:10.1038/onc.2010.143

PubMed Abstract | CrossRef Full Text | Google Scholar

Han, Q., Zhou, H., Xie, W., Sun, T., Wei, R., Nie, C., et al. (2020). Association between the Methylation of the STAT1 and SOCS3 in Peripheral Blood and Gastric Cancer. J. Gastroen. Hepatol. 35 (8), 1347–1354. doi:10.1111/jgh.15021

CrossRef Full Text | Google Scholar

Hibi, K., Goto, T., Shirahata, A., Saito, M., Kigawa, G., Nemoto, H., et al. (2011). Detection of TFPI2 Methylation in the Serum of Gastric Cancer Patients. [Comparative Study; Journal Article]. Anticancer Res. 31 (11), 3835–3838.

PubMed Abstract | Google Scholar

Hino, R., Uozaki, H., Murakami, N., Ushiku, T., Shinozaki, A., Ishikawa, S., et al. (2009). Activation of DNA Methyltransferase 1 by EBV Latent Membrane Protein 2A Leads to Promoter Hypermethylation of PTEN Gene in Gastric Carcinoma. Cancer Res. 69 (7), 2766–2774. doi:10.1158/0008-5472.CAN-08-3070

PubMed Abstract | CrossRef Full Text | Google Scholar

Hou, L., Wang, H., Sartori, S., Gawron, A., Lissowska, J., Bollati, V., et al. (2010). Blood Leukocyte DNA Hypomethylation and Gastric Cancer Risk in a High-Risk Polish Population. [Comparative Study; Journal Article; Randomized Controlled Trial; Research Support, N.I.H., Intramural]. Int. J. Cancer 127 (8), 1866–1874. doi:10.1002/ijc.25190

CrossRef Full Text | Google Scholar

Housman, G., Byler, S., Heerboth, S., Lapinska, K., Longacre, M., Snyder, N., et al. (2014). Drug Resistance in Cancer: An Overview. [Journal Article; Review]Cancers (Basel). 6 (3), 1769–1792. doi:10.3390/cancers6031769

PubMed Abstract | CrossRef Full Text | Google Scholar

Hu, D., Lou, X., Meng, N., Li, Z., Teng, Y., Zou, Y., et al. (2021). Peripheral Blood-Based DNA Methylation of Long Non-coding RNA H19 and Metastasis-Associated Lung Adenocarcinoma Transcript 1 Promoters Are Potential Non-invasive Biomarkers for Gastric Cancer Detection. Cancer Control 28, 1399505581. doi:10.1177/10732748211043667

CrossRef Full Text | Google Scholar

Hu, W., Zheng, W., Liu, Q., Chu, H., Chen, S., Kim, J. J., et al. (2017). Diagnostic Accuracy of DNA Methylation in Detection of Gastric Cancer: A Meta-Analysis. Oncotarget 8 (68), 113142–113152. doi:10.18632/oncotarget.22613

PubMed Abstract | CrossRef Full Text | Google Scholar

Hu, X., Ling, Z., Hong, L., Yu, Q., Li, P., and Ling, Z. (2021). Circulating Methylated THBS1 DNAs as a Novel Marker for Predicting Peritoneal Dissemination in Gastric Cancer. J. Clin. Lab. Anal. 35 (9), e23936. doi:10.1002/jcla.23936

CrossRef Full Text | Google Scholar

Huang, Z. B., Zhang, H. T., Yu, B., and Yu, D. H. (2021). Cell-free DNA as a Liquid Biopsy for Early Detection of Gastric Cancer. [Journal Article; Review]. Oncol. Lett. 21 (1), 3. doi:10.3892/ol.2020.12264

PubMed Abstract | CrossRef Full Text | Google Scholar

Hur, K., Niwa, T., Toyoda, T., Tsukamoto, T., Tatematsu, M., Yang, H. K., et al. (2011). Insufficient Role of Cell Proliferation in Aberrant DNA Methylation Induction and Involvement of Specific Types of Inflammation. Carcinogenesis 32 (1), 35–41. doi:10.1093/carcin/bgq219

PubMed Abstract | CrossRef Full Text | Google Scholar

Ikoma, H., Ichikawa, D., Koike, H., Ikoma, D., Tani, N., Okamoto, K., et al. (2006). Correlation between Serum DNA Methylation and Prognosis in Gastric Cancer Patients. Anticancer Res. 26 (3 B), 2313–2316.

PubMed Abstract | Google Scholar

Kaneda, A., Matsusaka, K., Aburatani, H., and Fukayama, M. (2012). Epstein-Barr Virus Infection as an Epigenetic Driver of Tumorigenesis. Cancer Res. 72 (14), 3445–3450. doi:10.1158/0008-5472.CAN-11-3919

PubMed Abstract | CrossRef Full Text | Google Scholar

Kanyama, Y., Hibi, K., Nakayama, H., Kodera, Y., Ito, K., Akiyama, S., et al. (2003). Detection of P16 Promoter Hypermethylation in Serum of Gastric Cancer Patients. [Comparative Study; Journal Article]. Cancer Sci. 94 (5), 418–420. doi:10.1111/j.1349-7006.2003.tb01457.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Karamitrousis, E. I., Balgkouranidou, I., Xenidis, N., Amarantidis, K., Biziota, E., Koukaki, T., et al. (2021). Prognostic Role of RASSF1A, SOX17 and Wif-1 Promoter Methylation Status in Cell-free DNA of Advanced Gastric Cancer Patients. Technol. Cancer Res. Treat. 20, 1079240927. doi:10.1177/1533033820973279

CrossRef Full Text | Google Scholar

Kim, N. (2019). Chemoprevention of Gastric Cancer by Helicobacter pylori Eradication and its Underlying Mechanism. [Journal Article; Review]. J. Gastroenterol. Hepatol. 34 (8), 1287–1295. doi:10.1111/jgh.14646

CrossRef Full Text | Google Scholar

Ko, K., Kananazawa, Y., Yamada, T., Kakinuma, D., Matsuno, K., Ando, F., et al. (2021). Methylation Status and Long-Fragment Cell-free DNA Are Prognostic Biomarkers for Gastric Cancer. [Journal Article; Observational Study; Research Support, Non-U.S. Gov't]. Cancer Med. 10 (6), 2003–2012. doi:10.1002/cam4.3755

PubMed Abstract | CrossRef Full Text | Google Scholar

Kosumi, K., Baba, Y., Ishimoto, T., Harada, K., Miyake, K., Izumi, D., et al. (2015). Relationship between LINE-1 Hypomethylation and Helicobacter pylori Infection in Gastric Mucosae. [Comparative Study; Journal Article; Randomized Controlled Trial]. Med. Oncol. 32 (4), 117. doi:10.1007/s12032-015-0571-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Lee, H. S., Hwang, S. M., Kim, T. S., Kim, D. W., Park, D. J., Kang, S. B., et al. (2013). Circulating Methylated Septin 9 Nucleic Acid in the Plasma of Patients with Gastrointestinal Cancer in the Stomach and colon. Transl Oncol. 6 (3), 290–296. doi:10.1593/tlo.13118

PubMed Abstract | CrossRef Full Text | Google Scholar

Lee, T., Leung, W. K., Chan, M. W. Y., Ng, E. K. W., Tong, J. H. M., Lo, K., et al. (2002). Detection of Gene Promoter Hypermethylation in the Tumor and Serum of Patients with Gastric Carcinoma. Clin. Cancer Res. 8 (6), 1761.

PubMed Abstract | Google Scholar

Leung, W. K., To, K. F., Chu, E. S., Chan, M. W., Bai, A. H., Ng, E. K., et al. (2005). Potential Diagnostic and Prognostic Values of Detecting Promoter Hypermethylation in the Serum of Patients with Gastric Cancer. Br. J. Cancer 92 (12), 2190–2194. doi:10.1038/sj.bjc.6602636

CrossRef Full Text | Google Scholar

Li, Y., Wang, Y., Li, D., Zhang, Y., Zhao, T., and Li, C. (2018). Procaine Is a Specific DNA Methylation Inhibitor with Anti-tumor Effect for Human Gastric Cancer. J. Cel. Biochem. 119 (2), 2440–2449. doi:10.1002/jcb.26407

CrossRef Full Text | Google Scholar

Li, Z., Li, D., Zhang, G., Xiong, J., Jie, Z., Cheng, H., et al. (2014). Methylation-associated Silencing of MicroRNA-335 Contributes Tumor Cell Invasion and Migration by Interacting with RASA1 in Gastric Cancer. [Journal Article]. Am. J. Cancer Res. 4 (6), 648–662.

Google Scholar

Lin, Z., Luo, M., Chen, X., He, X., Qian, Y., Lai, S., et al. (2017). Combined Detection of Plasma ZIC1, HOXD10 and RUNX3 Methylation Is a Promising Strategy for Early Detection of Gastric Cancer and Precancerous Lesions. J. Cancer 8 (6), 1038–1044. doi:10.7150/jca.18169

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, C., Li, N., Lu, H., Wang, Z., Chen, C., Wu, L., et al. (2015). Circulating SFRP1 Promoter Methylation Status in Gastric Adenocarcinoma and Esophageal Squamous Cell Carcinoma. Biomed. Rep. 3 (1), 123–127. doi:10.3892/br.2014.388

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, J., Xie, Y., Wang, F., Zhang, L., Zhang, Y., and Luo, H. (2013). Cytotoxicity of 5-Aza-2′-Deoxycytidine against Gastric Cancer Involves DNA Damage in an ATM-P53 Dependent Signaling Pathway and Demethylation of P16INK4A. Biomed. Pharmacother. 67 (1), 78–87. doi:10.1016/j.biopha.2012.10.015

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, L., Liu, C., Fotouhi, O., Fan, Y., Wang, K., Xia, C., et al. (2017). TERT Promoter Hypermethylation in Gastrointestinal Cancer: A Potential Stool Biomarker. [Journal Article; Res. Support, NonOncologist 22 (10), 1178–1188. doi:10.1634/theoncologist.2017-0064

PubMed Abstract | CrossRef Full Text | Google Scholar

Lu, X., Yu, J., Ying, L., Han, J., Wang, S., Yu, Q., et al. (2012). Stepwise Cumulation of RUNX3 Methylation Mediated by Helicobacter pylori Infection Contributes to Gastric Carcinoma Progression. Cancer-am. Cancer Soc. 118 (22), 5507–5517. doi:10.1002/cncr.27604

PubMed Abstract | CrossRef Full Text | Google Scholar

Maeda, M., Moro, H., and Ushijima, T. (2017). Mechanisms for the Induction of Gastric Cancer by Helicobacter pylori Infection: Aberrant DNA Methylation Pathway. Gastric Cancer 20 (1), 8–15. doi:10.1007/s10120-016-0650-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Matsusaka, K., Funata, S., Fukayama, M., and Kaneda, A. (2014). DNA Methylation in Gastric Cancer, Related to Helicobacter pylori and Epstein-Barr Virus. World J. Gastroenterol. 20 (14), 3916–3926. doi:10.3748/wjg.v20.i14.3916

CrossRef Full Text | Google Scholar

Matsusaka, K., Kaneda, A., Nagae, G., Ushiku, T., Kikuchi, Y., Hino, R., et al. (2011). Classification of Epstein–Barr Virus–Positive Gastric Cancers by Definition of DNA Methylation Epigenotypes. Cancer Res. 71 (23), 7187. doi:10.1158/0008-5472.CAN-11-1349

PubMed Abstract | CrossRef Full Text | Google Scholar

Miao, J., Liu, Y., Zhao, G., Liu, X., Ma, Y., Li, H., et al. (2020). Feasibility of Plasma-Methylated SFRP2 for Early Detection of Gastric Cancer. [Journal Article]. Cancer Control 27 (2), 1148357823. doi:10.1177/1073274820922559

PubMed Abstract | CrossRef Full Text | Google Scholar

Min, J., Choi, B., Han, T. S., Lee, H. J., Kong, S. H., Suh, Y. S., et al. (2017). Methylation Levels of LINE-1 as a Useful Marker for Venous Invasion in Both FFPE and Frozen Tumor Tissues of Gastric Cancer. [Journal Article]. Mol. Cell 40 (5), 346–354. doi:10.14348/molcells.2017.0013

PubMed Abstract | CrossRef Full Text | Google Scholar

Moore, L. D., Le, T., and Fan, G. (2013). DNA Methylation and its Basic Function. Neuropsychopharmacol 38 (1), 23–38. doi:10.1038/npp.2012.112

PubMed Abstract | CrossRef Full Text | Google Scholar

Muhammad, J. S., Eladl, M. A., and Khoder, G. (2019). Helicobacter Pylori-Induced DNA Methylation as an Epigenetic Modulator of Gastric Cancer: Recent Outcomes and Future Direction. Pathogens 8, 23. doi:10.3390/pathogens8010023

PubMed Abstract | CrossRef Full Text | Google Scholar

Muretto, P., Ruzzo, A., Pizzagalli, F., Graziano, F., Maltese, P., Zingaretti, C., et al. (2008). Endogastric Capsule for E-Cadherin Gene (CDH1) Promoter Hypermethylation Assessment in DNA from Gastric Juice of Diffuse Gastric Cancer Patients. Ann. Oncol. 19 (3), 516–519. doi:10.1093/annonc/mdm493

PubMed Abstract | CrossRef Full Text | Google Scholar

Nakajima, T., Enomoto, S., Yamashita, S., Ando, T., Nakanishi, Y., Nakazawa, K., et al. (2010). Persistence of a Component of DNA Methylation in Gastric Mucosae after Helicobacter pylori Eradication. J. Gastroenterol. 45 (1), 37–44. doi:10.1007/s00535-009-0142-7

CrossRef Full Text | Google Scholar

Namba-Fukuyo, H., Funata, S., Matsusaka, K., Fukuyo, M., Rahmutulla, B., Mano, Y., et al. (2016). TET2 Functions as a Resistance Factor against DNA Methylation Acquisition during Epstein-Barr Virus Infection. Oncotarget 7, 49. doi:10.18632/oncotarget.13130

PubMed Abstract | CrossRef Full Text | Google Scholar

Navada, S. C., Steinmann, J., Lübbert, M., and Silverman, L. R. (2014). Clinical Development of Demethylating Agents in Hematology. J. Clin. Invest. 124 (1), 40–46. doi:10.1172/JCI69739

CrossRef Full Text | Google Scholar

Necula, L., Matei, L., Dragu, D., Neagu, A. I., Mambet, C., Nedeianu, S., et al. (2019). Recent Advances in Gastric Cancer Early Diagnosis. World J. Gastroentero. 25 (17), 2029–2044. doi:10.3748/wjg.v25.i17.2029

CrossRef Full Text | Google Scholar

Ng, E. K., Leung, C. P., Shin, V. Y., Wong, C. L., Ma, E. S., Jin, H. C., et al. (2011). Quantitative Analysis and Diagnostic Significance of Methylated SLC19A3 DNA in the Plasma of Breast and Gastric Cancer patientsResearch Support, Non-U.S. Gov't]. [Journal ArticlePLoS One 6 (7), e22233. doi:10.1371/journal.pone.0022233

CrossRef Full Text | Google Scholar

Niwa, T., Toyoda, T., Tsukamoto, T., Mori, A., Tatematsu, M., and Ushijima, T. (2013). Prevention of <em>Helicobacter pylori</em>–Induced Gastric Cancers in Gerbils by a DNA Demethylating Agent. Cancer Prev. Res. 6 (4), 263. doi:10.1158/1940-6207.CAPR-12-0369

CrossRef Full Text | Google Scholar

Okada, T., Nakamura, M., Nishikawa, J., Sakai, K., Zhang, Y., Saito, M., et al. (2013). Identification of Genes Specifically Methylated in Epstein-Barr Virus-Associated Gastric carcinomasResearch Support, Non-U.S. Gov't]. [Journal Articlecancer Sci. 104 (10), 1309–1314. doi:10.1111/cas.12228

CrossRef Full Text | Google Scholar

Pan, W., Zhu, S., Yuan, M., Cui, H., Wang, L., Luo, X., et al. (2010). MicroRNA-21 and MicroRNA-148a Contribute to DNA Hypomethylation in Lupus CD4+ T Cells by Directly and Indirectly Targeting DNA Methyltransferase 1. J. Immunol. 184 (12), 6773. doi:10.4049/jimmunol.0904060

PubMed Abstract | CrossRef Full Text | Google Scholar

Pessoa, L. S., Heringer, M., and Ferrer, V. P. (2020). CtDNA as a Cancer Biomarker: A Broad Overview. [Journal Article; Review]. Crit. Rev. Oncol. Hematol. 155, 103109. doi:10.1016/j.critrevonc.2020.103109

PubMed Abstract | CrossRef Full Text | Google Scholar

Pimson, C., Ekalaksananan, T., Pientong, C., Promthet, S., Putthanachote, N., Suwanrungruang, K., et al. (2016). Aberrant Methylation of PCDH10 and RASSF1A Genes in Blood Samples for Non-invasive Diagnosis and Prognostic Assessment of Gastric Cancer. PeerJ 4, e2112. doi:10.7717/peerj.2112

PubMed Abstract | CrossRef Full Text | Google Scholar

Qi, Q., Pan, Y. F., Shen, J. J., Gu, X. Q., Han, S. W., Liao, H. H., et al. (2016). Circulating DNA for Detection of Gastric Cancer. Eur. Rev. Med. Pharmacol. Sci. 20 (12), 2558–2564.

PubMed Abstract | Google Scholar

Qiu, H., Yashiro, M., Shinto, O., Matsuzaki, T., and Hirakawa, K. (2009). DNA Methyltransferase Inhibitor 5-Aza-CdR Enhances the Radiosensitivity of Gastric Cancer Cells. Cancer Sci. 100 (1), 181–188. doi:10.1111/j.1349-7006.2008.01004.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Qu, Y., Dang, S., and Hou, P. (2013). Gene Methylation in Gastric Cancer. [Journal Article; Research Support, Non-U.S. Gov't; Review]. Clin. Chim. Acta 424, 53–65. doi:10.1016/j.cca.2013.05.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Saito, M., Suzuki, K., Maeda, T., Kato, T., Kamiyama, H., Koizumi, K., et al. (2012). The Accumulation of DNA Demethylation in Sat α in normal Gastric Tissues with Helicobacter pylori Infection Renders Susceptibility to Gastric Cancer in Some Individuals. [Journal Article; Research Support, Non-U.S. Gov't]. Oncol. Rep. 27 (6), 1717–1725. doi:10.3892/or.2012.1718

PubMed Abstract | CrossRef Full Text | Google Scholar

Sakakura, C., Hamada, T., Miyagawa, K., Nishio, M., Miyashita, A., Nagata, H., et al. (2009). Quantitative Analysis of Tumor-Derived Methylated RUNX3 Sequences in the Serum of Gastric Cancer Patients. Anticancer Res. 29 (7), 2619–2625.

PubMed Abstract | Google Scholar

Schneider, B. J., Shah, M. A., Klute, K., Ocean, A., Popa, E., Altorki, N., et al. (2017). Phase I Study of Epigenetic Priming with Azacitidine Prior to Standard Neoadjuvant Chemotherapy for Patients with Resectable Gastric and Esophageal Adenocarcinoma: Evidence of Tumor Hypomethylation as an Indicator of Major Histopathologic Response. Clin. Cancer Res. 23 (11), 2673–2680. doi:10.1158/1078-0432.CCR-16-1896

PubMed Abstract | CrossRef Full Text | Google Scholar

Sexton, R. E., Al, H. M., Diab, M., and Azmi, A. S. (2020). Gastric Cancer: A Comprehensive Review of Current and Future Treatment Strategies. Cancer Metastasis Rev. 39 (4), 1179–1203. doi:10.1007/s10555-020-09925-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Shao, L., Chen, Z., Peng, D., Soutto, M., Zhu, S., Bates, A., et al. (2018). Methylation of the HOXA10 Promoter Directs miR-196b-5p-dependent Cell Proliferation and Invasion of Gastric Cancer Cells. Mol. Cancer Res. 16 (4), 696–706. doi:10.1158/1541-7786.MCR-17-0655

PubMed Abstract | CrossRef Full Text | Google Scholar

Shin, C. M., Kim, N., Park, J. H., Kang, G. H., Kim, J. S., Jung, H. C., et al. (2012). Prediction of the Risk for Gastric Cancer Using Candidate Methylation Markers in the Non-neoplastic Gastric Mucosae. J. Pathol. 226 (4), 654–665. doi:10.1002/path.2990

CrossRef Full Text | Google Scholar

Shinozaki-Ushiku, A., Kunita, A., and Fukayama, M. (2015). Update on Epstein-Barr Virus and Gastric Cancer (Review). Int. J. Oncol. 46 (4), 1421–1434. doi:10.3892/ijo.2015.2856

CrossRef Full Text | Google Scholar

Shitara, K., Matsuo, K., Ito, S., Sawaki, A., Kawai, H., Yokota, T., et al. (2010). Effects of Genetic Polymorphisms in the ABCB1 Gene on Clinical Outcomes in Patients with Gastric Cancer Treated by Second-Line Chemotherapy. Asian Pac. J. Cancer Prev. 11 (2), 447–452.

Google Scholar

Si, J., Boumber, Y. A., Shu, J., Qin, T., Ahmed, S., He, R., et al. (2010). Chromatin Remodeling Is Required for Gene Reactivation after Decitabine-Mediated DNA Hypomethylation. [Journal Article; Research Support, N.I.H., Extramural]. Cancer Res. 70 (17), 6968–6977. doi:10.1158/0008-5472.CAN-09-4474

PubMed Abstract | CrossRef Full Text | Google Scholar

Skvortsova, K., Stirzaker, C., and Taberlay, P. (2019). The DNA Methylation Landscape in Cancer. Essays Biochem. 63 (6), 797–811. doi:10.1042/EBC20190037

PubMed Abstract | CrossRef Full Text | Google Scholar

Sohn, B. H., Hwang, J. E., Jang, H. J., Lee, H. S., Oh, S. C., Shim, J. J., et al. (2017). Clinical Significance of Four Molecular Subtypes of Gastric Cancer Identified by the Cancer Genome Atlas Project. [Journal Article]. Clin. Cancer Res. 23 (15), 4441–4449. doi:10.1158/1078-0432.CCR-16-2211

PubMed Abstract | CrossRef Full Text | Google Scholar

Sun, H., Zhou, H., Zhang, Y., Chen, J., Han, X., Huang, D., et al. (2018). Aberrant Methylation of FAT4 and SOX11 in Peripheral Blood Leukocytes and Their Association with Gastric Cancer Risk. J. Cancer 9 (13), 2275–2283. doi:10.7150/jca.24797

PubMed Abstract | CrossRef Full Text | Google Scholar

Tahara, T., and Arisawa, T. (2015). DNA Methylation as a Molecular Biomarker in Gastric Cancer. Epigenomics-UK. 7 (3), 475–486. doi:10.2217/epi.15.4

CrossRef Full Text | Google Scholar

Tahara, T., Maegawa, S., Chung, W., Garriga, J., Jelinek, J., Estécio, M. R. H., et al. (2013). Examination of Whole Blood DNA Methylation as a Potential Risk Marker for Gastric Cancer. Cancer Prev. Res. 6 (10), 1093–1100. doi:10.1158/1940-6207.CAPR-13-0034

CrossRef Full Text | Google Scholar

Tahara, T., and Arisawa, T. (2015). DNA methylation as a molecular biomarker in gastric cancer. 608 Epigenomics-UK 7 (3), 476–486. doi:10.2217/epi.15.4

PubMed Abstract | CrossRef Full Text | Google Scholar

Tahara, T., Tahara, S., Horiguchi, N., Kawamura, T., Okubo, M., Yamada, H., et al. (2018). Methylation Status of IGF2 DMR and LINE1 in Leukocyte DNA Provides Distinct Clinicopathological Features of Gastric Cancer Patients. Clin. Exp. Med. 18 (2), 215–220. doi:10.1007/s10238-017-0471-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Tan, W., Zhou, W., Yu, H., Luo, H., and Shen, L. (2013). The DNA Methyltransferase Inhibitor Zebularine Induces Mitochondria-Mediated Apoptosis in Gastric Cancer Cells In Vitro and In Vivo. Biochem. Bioph. Res. Co. 430 (1), 250–255. doi:10.1016/j.bbrc.2012.10.143

CrossRef Full Text | Google Scholar

Usui, G., Matsusaka, K., Mano, Y., Urabe, M., Funata, S., Fukayama, M., et al. (2021). DNA Methylation and Genetic Aberrations in Gastric Cancer. Digestion 102 (1), 25–32. doi:10.1159/000511243

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, G., Zhang, W., Zhou, B., Jin, C., Wang, Z., Yang, Y., et al. (2015). The Diagnosis Value of Promoter Methylation of UCHL1 in the Serum for Progression of Gastric Cancer. Biomed. Res. Int. 2015, 741030. doi:10.1155/2015/741030

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, Y. C., Yu, Z. H., Liu, C., Xu, L. Z., Yu, W., Lu, J., et al. (2008). Detection of RASSF1A Promoter Hypermethylation in Serum from Gastric and Colorectal Adenocarcinoma Patients. [Journal Article]world J. Gastroenterol. 14 (19), 3074–3080. doi:10.3748/wjg.14.3074

CrossRef Full Text | Google Scholar

Watanabe, Y., Kim, H. S., Castoro, R. J., Chung, W., Estecio, M. R. H., Kondo, K., et al. (2009). Sensitive and Specific Detection of Early Gastric Cancer with DNA Methylation Analysis of Gastric Washes. Gastroenterology 136 (7), 2149–2158. doi:10.1053/j.gastro.2009.02.085

PubMed Abstract | CrossRef Full Text | Google Scholar

Wu, F., Li, R., Yang, M., Yue, G., Wang, H., Liu, Q., et al. (2015). Gelatinases-stimuli Nanoparticles Encapsulating 5-fluorouridine and 5-Aza-2′-Deoxycytidine Enhance the Sensitivity of Gastric Cancer Cells to Chemical Therapeutics. Cancer Lett. 363 (1), 7–16. doi:10.1016/j.canlet.2015.01.006

PubMed Abstract | CrossRef Full Text | Google Scholar

Wu, F. L., Li, R. T., Yang, M., Yue, G. F., Wang, H. Y., Liu, Q., et al. (2015). Gelatinases-stimuli Nanoparticles Encapsulating 5-fluorouridine and 5-Aza-2'-Deoxycytidine Enhance the Sensitivity of Gastric Cancer Cells to Chemical therapeuticsResearch Support, Non-U.S. Gov't]. [Journal Articlecancer Lett. 363 (1), 7–16. doi:10.1016/j.canlet.2015.01.006

PubMed Abstract | CrossRef Full Text | Google Scholar

Xie, Q., Bai, Q., Zou, L., Zhang, Q., Zhou, Y., Chang, H., et al. (2014). Genistein Inhibits DNA Methylation and Increases Expression of Tumor Suppressor Genes in Human Breast Cancer Cells. Genes, Chromosomes and Cancer 53 (5), 422–431. doi:10.1002/gcc.22154

PubMed Abstract | CrossRef Full Text | Google Scholar

Xie, W., Zhou, H., Han, Q., Sun, T., Nie, C., Hong, J., et al. (2020). Relationship between DLEC1 and PBX3 Promoter Methylation and the Risk and Prognosis of Gastric Cancer in Peripheral Blood Leukocytes. [Journal Article]. J. Cancer Res. Clin. Oncol. 146 (5), 1115–1124. doi:10.1007/s00432-020-03171-4

CrossRef Full Text | Google Scholar

Xue, W., Feng, Y., Wang, F., Li, P., Liu, Y., Guo, Y., et al. (2016). The Value of Serum RASSF10 Hypermethylation as a Diagnostic and Prognostic Tool for Gastric Cancer. Tumor Biol. 37 (8), 11249–11257. doi:10.1007/s13277-016-5001-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Yamamoto, H., Watanabe, Y., Oikawa, R., Morita, R., Yoshida, Y., Maehata, T., et al. (2016). BARHL2 Methylation Using Gastric Wash DNA or Gastric Juice Exosomal DNA Is a Useful Marker for Early Detection of Gastric Cancer in an H. Pylori-independent Manner. Clin. translational Gastroenterol. 7 (7), e184. doi:10.1038/ctg.2016.40

PubMed Abstract | CrossRef Full Text | Google Scholar

Yamamoto, H., Watanabe, Y., Sato, Y., Maehata, T., and Itoh, F. (2020). Non-Invasive Early Molecular Detection of Gastric Cancers. Cancers (Basel). 12 (10). doi:10.3390/cancers12102880

PubMed Abstract | CrossRef Full Text | Google Scholar

Yan, H., Chen, W., Ge, K., Mao, X., Li, X., Liu, W., et al. (2021). Value of Plasma Methylated SFRP2 in Prognosis of Gastric Cancer. Dig. Dis. Sci. 66 (11), 3854–3861. doi:10.1007/s10620-020-06710-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Yau, T. O., Tang, C. M., and Yu, J. (2014). Epigenetic Dysregulation in Epstein-Barr Virus-Associated Gastric Carcinoma: Disease and Treatments. World J. Gastroenterol. 20 (21), 6448–6456. doi:10.3748/wjg.v20.i21.6448

CrossRef Full Text | Google Scholar

Yoshida, T., Yamashita, S., Takamura-Enya, T., Niwa, T., Ando, T., Enomoto, S., et al. (2011). Alu and Satα Hypomethylation in Helicobacter Pylori-Infected Gastric Mucosae. Int. J. Cancer 128 (1), 33–39. doi:10.1002/ijc.25534

CrossRef Full Text | Google Scholar

Yu, Q., Wang, X., Luo, J., Wang, S., Fang, X., Yu, J., et al. (2012). CDH1 Methylation in Preoperative Peritoneal Washes Is an Independent Prognostic Factor for Gastric Cancer. J. Surg. Oncol. 106 (6), 765–771. doi:10.1002/jso.23116

CrossRef Full Text | Google Scholar

Yuasa, Y. (2010). Epigenetics in Molecular Epidemiology of Cancer a New scopeResearch Support, Non-U.S. Gov't]. [Journal Articleadv. Genet. 71, 211–235. doi:10.1016/B978-0-12-380864-6.00007-9

CrossRef Full Text | Google Scholar

Zeng, X. Q., Wang, J., and Chen, S. Y. (2017). Methylation Modification in Gastric Cancer and Approaches to Targeted Epigenetic Therapy (Review). Int. J. Oncol. 50 (6), 1921–1933. doi:10.3892/ijo.2017.3981

CrossRef Full Text | Google Scholar

Zhang, G., Estève, P., Chin, H. G., Terragni, J., Dai, N., Corrêa, I. R. J., et al. (2015). Small RNA-Mediated DNA (Cytosine-5) Methyltransferase 1 Inhibition Leads to Aberrant DNA Methylation. Nucleic Acids Res. 43 (12), 6112–6124. doi:10.1093/nar/gkv518

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, H., Song, Y., Xia, P., Cheng, Y., Guo, Q., Diao, D., et al. (2014). Detection of Aberrant Hypermethylated Spastic Paraplegia-20 as a Potential Biomarker and Prognostic Factor in Gastric Cancer. Med. Oncol. 31 (2), 830. doi:10.1007/s12032-013-0830-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, X., Yashiro, M., Ohira, M., Ren, J., and Hirakawa, K. (2006). Synergic Antiproliferative Effect of DNA Methyltransferase Inhibitor in Combination with Anticancer Drugs in Gastric Carcinoma. Cancer Sci. 97 (9), 938–944. doi:10.1111/j.1349-7006.2006.00253.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, Y., Ye, X., Geng, J., and Chen, L. (2010). Epigenetic Inactivation of Deleted in Lung and Esophageal Cancer 1 Gene by Promoter Methylation in Gastric and Colorectal Adenocarcinoma. [Journal Article]Hepatogastroenterology 57 (104), 1614–1619. doi:10.4149/neo_2010_03_228

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, Y., Zhou, H., Sun, H., Chen, J., Huang, D., Han, X., et al. (2018). Association of Peripheral Blood Leukocyte KIBRA Methylation with Gastric Cancer Risk: A Case-Control Study. Cancer Med. 7 (6), 2682–2690. doi:10.1002/cam4.1474

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhao, J., Liang, Q., Cheung, K. F., Kang, W., Lung, R. W., Tong, J. H., et al. (2013). Genome-wide Identification of Epstein-Barr Virus-Driven Promoter Methylation Profiles of Human Genes in Gastric Cancer Cells. Cancer-am. Cancer Soc. 119 (2), 304–312. doi:10.1002/cncr.27724

CrossRef Full Text | Google Scholar

Zheng, Y., Chen, L., Li, J., Yu, B., Su, L., Chen, X., et al. (2011a). Hypermethylated DNA as Potential Biomarkers for Gastric Cancer Diagnosis. Clin. Biochem. 44 (17-18), 1405–1411. doi:10.1016/j.clinbiochem.2011.09.006

PubMed Abstract | CrossRef Full Text | Google Scholar

Zheng, Y., Zhang, Y., Huang, X., and Chen, L. (2011b). Analysis of the RUNX3 Gene Methylation in Serum DNA from Esophagus Squamous Cell Carcinoma, Gastric and Colorectal Adenocarcinoma Patients. Hepatogastroenterology 58 (112), 2007–2011. doi:10.5754/hge10016

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhou, H., Sun, H., Liu, X., Chen, J., Zhang, L., Lin, S., et al. (2019). Combined Effect between WT1 Methylation and Helicobacter pylori Infection, Smoking, and Alcohol Consumption on the Risk of Gastric Cancer. Helicobacter 24 (5), e12650. doi:10.1111/hel.12650

PubMed Abstract | CrossRef Full Text | Google Scholar

Zong, L., and Seto, Y. (2014). CpG Island Methylator Phenotype, Helicobacter pylori, Epstein-Barr Virus, and Microsatellite Instability and Prognosis in Gastric Cancer: A Systematic Review and Meta-Analysis. [Journal Article; Meta-Analysis; Review; Systematic Review]. PLoS One 9 (1), e86097. doi:10.1371/journal.pone.0086097

PubMed Abstract | CrossRef Full Text | Google Scholar

Zuo, J., Xia, J., Ju, F., Yan, J., Zhu, A., Jin, S., et al. (2013). MicroRNA-148a Can Regulate Runt-Related Transcription Factor 3 Gene Expression via Modulation of DNA Methyltransferase 1 in Gastric Cancer. Mol. Cell 35 (4), 313–319. doi:10.1007/s10059-013-2314-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: DNA methylation, gastric cancer, diagnosis, therapy, prognosis

Citation: Zeng Y, Rong H, Xu J, Cao R, Li S, Gao Y, Cheng B and Zhou T (2022) DNA Methylation: An Important Biomarker and Therapeutic Target for Gastric Cancer. Front. Genet. 13:823905. doi: 10.3389/fgene.2022.823905

Received: 28 November 2021; Accepted: 07 February 2022;
Published: 04 March 2022.

Edited by:

Trygve Tollefsbol, University of Alabama at Birmingham, United States

Reviewed by:

Apiwat Mutirangura, Chulalongkorn University, Thailand
Fazlur Rahman Talukdar, International Agency For Research On Cancer (IARC), France

Copyright © 2022 Zeng, Rong, Xu, Cao, Li, Gao, Cheng and Zhou. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Tao Zhou, zhoutao@sdu.edu.cn

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.