bioRxiv preprint doi: https://doi.org/10.1101/2021.03.01.433329; this version posted March 1, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

1 DNMT family induced down-regulation of NDRG1 via DNA methylation and

2 clinicopathological significance in gastric cancer

3 Xiaojing Chang1*, Jinguo MA2*, Xiaoying Xue1, Guohui Wang1, Linlin Su1, Xuetao

4 Han1, Huandi Zhou3, Liubing Hou3.

5 1 Department of Radiotherapy, The Second Hospital of Hebei Medical University,

6 Shijiazhuang , China.

7 2 Hulun Buir People’s hospital, Hulun Buir Medical school in nationalities University

8 of inner Mongolia, Hulun Buir, China.

9 3Department of Central Laboratory, The Second Hospital of Hebei Medical

10 University, Shijiazhuang, China

11 *Xiaojing Chang and Jinguo MA have contributed equally to this manuscript.

12 Corresponding author: Xiaoying Xue, MD, PhD, Professor, Department of

13 Radiotherapy, The Second Hospital of Hebei Medical University, NO.215 Heping

14 West Road, Shijiazhuang, China. E-mail:[email protected]. 15 E-mail addresses of authors:

16 Xiaojing Chang: [email protected] 17 Jinguo MA: [email protected] 18 Xiaoying Xue: [email protected]/[email protected] 19 Guohui Wang: [email protected] 20 Linlin Su:[email protected] 21 Xuetao Han :[email protected] 22 Huandi Zhou: [email protected] 23 Liubing Hou: [email protected]

24 Running title:

25 The relationship of DNA methylation of NDRG1and DNMT family in gastric cancer .

26 Funding statement: The authors received funding through the National Natural

27 Science Foundation of Hebei province of China(#H2018206180).

28 Conflict of interest statement: No conflicts of interest are declared by the authors.

29 Ethical approval statement: Our study did not require an ethical board approval

30 because it did not contain human or animal trials.

1 bioRxiv preprint doi: https://doi.org/10.1101/2021.03.01.433329; this version posted March 1, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

31 DNMT family induced down-regulation of NDRG1 via DNA methylation and

32 clinicopathological significance in gastric cancer

33 Abstract Background Aberrant DNA methylation of tumor suppressor is a

34 common event in the development and progression of gastric cancer(GC). Our

35 previous study showed NDRG1, which could suppress cell invasion and migration,

36 was frequently down-regulated by DNA methylation of its promoter in GC. Purpose

37 and Methods To analyze the relationship between the expression and DNA

38 methylation of NDRG1 and DNA methyltransferase (DNMT) family. We performed

39 a comprehensive comparison analysis using 407 patients including sequencing

40 analysis data of GC from TCGA. Results NDRG1 was negatively correlative to

41 DNMT1(p =0.03), DNMT3A(p =0.01), DNMT3B(p =0.88), respectively.

42 Whereas, the DNA methylation of NDRG1 was positively correlative to DNMT

43 family(DNMT1 p<0.01, DNMT3A p<0.001 , DNMT3B p=0.57, respectively).

44 NDRG1 expression was significantly inverse correlated with invasion depth (p

45 =0.023), and DNMT1 was significantly positive correlated with the degree of tumor

46 cell differentiation (p =0.049). DNMT3B was significantly correlated with tumor cell

47 differentiation (p =0.030). However, there was no association between the expression

48 of DNMT3A and clinicopathological features. The univariate analysis showed that

49 NDRG1and DNMTs had no association with prognosis of GC patients. But,

50 multivariate analysis showed DNMT1 was significantly correlated with prognosis of

51 GC patients. Conclusion These data suggest that down-regulation of NDRG1 in

52 gastric cancer is due to DNA methylation of NDRG1 promoter via DNMT

2 bioRxiv preprint doi: https://doi.org/10.1101/2021.03.01.433329; this version posted March 1, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

53 family. The demethylating agent maybe a potential target drug for GC patients.

54 Keywords: Gastric carcinoma, tumor suppressor gene, NDRG1, DNMT, DNA

55 methylation, metastasis

56 Introduction

57 Gastric carcinoma (GC) is one of the most common digestive malignancies

58 worldwide with the third most common cause of cancer-related death1, 2. The poor

59 prognosis and high recurrence rate are mainly due to lymph node metastasis in its

60 early stage.

61 To date, a number of laboratories have shown that the occurrence of GC is usually

62 caused by oncogenes activation and tumor suppressor genes(TSGs) inactivation.

63 TSGs could suppress tumor cells migration and invasion. Studies have shown that the

64 inactivation of most TSGs are related to DNA methylation of CpG, while the

65 silencing of these genes possibly contributes to the development and progression of

66 tumors3-5. There is increasing evidence that aberrant DNA promoter methylation of

67 TSGs is a major event in the development and progression of gastric cance6. Our

68 previous studies showed NDRG1(N-myc downstream-regulated gene 1), a

69 differentiation-related and tumor suppressor gene, which could suppress cancer cells

70 invasion and migration, was frequently down-regulated by DNA methylation of its

71 promoter in GC,but there were not any mutations in NDRG1 cDNA, and also not

72 significant correlation with histone acetylation 7, 8.

73 DNA methylation of CpG islands, which is carried out

74 by DNA methyltransferase (DNMT) enzymes, is the most widely and the best well

75 studied epigenetic modification event, and leads to transcriptional gene silencing9.

76 DNA methyltransferase (DNMT) enzymes include DNMT1, DNMT2, DNMT3A,

77 DNMT3B, and DNMT3L, among which DNMT1(a maintenance DNMT), DNMT3A

78 and DNMT3B(de novo methyltransferases) are the most important10. In the current

79 study, we focused on the relationship between NDRG1 and DNMT family(DNMT1,

80 DNMT3A and DNMT3B) and their clinical significance in GC by analyzing

81 high-throughput data obtained from TCGA. 3 bioRxiv preprint doi: https://doi.org/10.1101/2021.03.01.433329; this version posted March 1, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

82 Materials and methods

83 The data that we investigated the mRNA expression of NDRG1 and DNMTs, and

84 the DNA methylation of NDRG1 in GC was obtained from the data portal of TCGA

85 (https://portal.gdc.cancer.gov/), which is a well-known cancer research project that

86 collects and analyzes high-quality tumor samples and makes the related data available

87 to researchers. We obtained the GC data set (March 1st, 2018 updated), which

88 encompasses 407 gastric tumor samples, and of which 338 cases encompasses the

89 data of DNA methylation of NDRG1. Total of 315 cases contain the complete

90 clinicopathological information.

91 Gastric Cells and Culture

92 Human gastric cancer cell lines, SGC7901 and MKN45 were obtained from the

93 Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences (Shanghai,

94 China), and one immortalized normal gastric cell line, GES1 was obtained from the

95 Oncology Institute of China Medical University. Cells were cultured with RPMI 1640

96 (Invitrogen/Gibco, Grand Island, NY, USA) containing 10 % fetal bovine serum

97 (Invitrogen/Gibco) in a humidified atmosphere of 5 % CO2 at 37 C.

98 Isolation and Western Blot

99 Total cellular protein was extracted from cultured cells and western bolt was

100 performed as previous description7. Protein bands were scanned and quantified using

101 densitometric software (Bio-Rad, California, USA). A rabbit monoclonal

102 anti-NDRG1 antibody was at a dilution of 1:1,000 (Cell Signaling Technology,

103 Danvers, MA, USA). DNMT1, DNMT3A and DNMT3B antibody at a dilution of

104 1:500 were purchased from Bioss, Beijing, China.

105 Statistics

106 All statistical analyses were conducted using SPSS 24.0 (Chicago, IL, USA) and R

107 3.6.1 (https://www.r-project. org/). R language and GraphPad Prism 7 (San Diego,

108 CA, USA) were performed to draw plots. X2 test and Fisher’s exact test were used to

109 generate P values, Values with p < 0.05 are considered as statistically significant.

110 Results

4 bioRxiv preprint doi: https://doi.org/10.1101/2021.03.01.433329; this version posted March 1, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

111 NDRG1 was negatively correlative to DNMT family

112 Total of 407 gastric cancer cases ( 375 was gastric cancer tissues and 32 was

113 nonmalignant gastric tissues ) which included sequencing analysis data were

114 downloaded form TCGA, then we performed a comprehensive comparison analysis

115 about the mRNA expression of NDRG1 and DNMTs and their clinicopathological

116 features. As shown in Figure 1A-C, NDRG1 mRNA expression was negatively

117 correlative to DNMT1(r=-0.1067, p =0.03), DNMT3A(r=-0.1015, p =0.01),

118 DNMT3B(r=-0.0069, p =0.88). Noteworthy, NDRG1 was significantly associated

119 with DNMT1(p =0.03) and DNMT3A ( p =0.01 ) , and DNMT3A showed the

120 strongest association. However, there was no significant association between NDRG1

121 and DNMT3B (p=0.8885).

122 To confirm these results, we further assessed the protein levels of NDRG1 and

123 DNMTs in one immortalized normal gastric cell line, GES1 and two gastric cell lines,

124 SGC7901 and MKN45 by western blot. Results showed DNMT1, DNMT3A and

125 DNMT3B were up-regulated in SGC7901 and MKN45 cells compared with GES1,

126 while NDRG1 expression was inversed (p < 0.05, Fig. 2). This was in accordance to

127 the results of the relationship between NDRG1 and DNMT family mRNA expression

128 from TCGA.

129 Methylation of NDRG1 gene promoter was positively correlative to the

130 expression DNMT family

131 Among all of 407 GC cases, 338 cases encompass the data of DNA methylation of

132 NDRG1. Results showed DNA methylation of NDRG1 gene promoter was positively

133 correlative to the expression DNMT family, and was significantly associated with

134 DNMT1 (p =0.002, Fig.1D) and DNMT3A (p <0.001, Fig.1E). However, no

135 statistical significance was found between NDRG1 and DNMT3B (p =0.57, Fig. 1F).

136 The association of NDRG1 and DNMT family with the clinicopathological

137 parameters and prognosis of GC

138 The association of NDRG1 and DNMT family with the clinicopathological

139 parameters of gastric cancer patients were shown in Table 1. Total of 315 cases which

5 bioRxiv preprint doi: https://doi.org/10.1101/2021.03.01.433329; this version posted March 1, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

140 contain the complete clinicopathological and survival information were analyzed. The

141 high and low expression of NDRG1 and DNMTs was based on the median value of

142 their mRNA levels. We found NDRG1 expression was significantly inverse correlated

143 with invasion depth (p =0.023), whereas DNMT1 was statistically significantly

144 positive correlated with invasion depth (p = 0.049), DNMT3B was significantly

145 positive correlated with the degree of tumor cell differentiation (p =0.030). There was

146 no significant association between DNMT3A and the clinicopathological parameters

147 of the GC patients (Table 1).

148 Univariate analysis of prognosis showed that age, invasion depth, lymph metastasis

149 and stage were significantly inverse correlated with prognosis of GC patients. But

150 NDRG1and DNMTs showed no association with prognosis of GC patients(Fig. 3).

151 Whereas, Multivariate analysis of prognosis showed age, stage and DNMT1 were

152 significantly correlated with prognosis of GC patients. But NDRG1, DNMT3A

153 andDNMT3B showed no association with prognosis(Fig. 4).

154 Discussion

155 NDRG1, a differentiation-related gene, which belongs to NDRG family, is

156 down-regulated in many tumors, and proposed as a metastasis suppressor gene11-14.

157 Our previous studies confirmed the anti-cancer effect of NDRG1 in GC7, 8, 15. Gene

158 network analysis shows that the 5 'end of the NDRG1 gene contains a large CpG

159 island. Some studies have confirmed the downregulation of NDRG1 caused by DNA

160 methylation of CpG islands of its promoter in breast cancer, prostate cancer cells and

161 pancreatic cancer16-18. In GC, the down-regulation of NDRG1 was also found to be

162 regulated by DNA methylation of its promoter in our previous study. DNA

163 methylation is carried out by DNA methyltransferase (DNMT) enzymes. To date, not

164 any studies reported the relationship of NDRG1 and DNMTs in GC.

165 In the current study, total 407 GC cases which included sequencing analysis data

166 of NDRG1 mRNA were analyzed. We found that NDRG1 mRNA level was

167 negatively correlative to DNMT1, DNMT3A and DNMT3B, and DNMT3A showed

168 the strongest association. No significant association was found between NDRG1 and

6 bioRxiv preprint doi: https://doi.org/10.1101/2021.03.01.433329; this version posted March 1, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

169 DNMT3B. The results of western blot analysis confirmed these data from TCGA.

170 This is the first time for analyzing the relationship between NDRG1 and DNMT

171 family. NDRG2, another member of NDRG family which includes four members

172 NDRG1-4, was reported to be significantly decreased in human cancers, and in GC, H.

173 pylori silenced Ndrg2 by activating the NF-κB pathway and up-regulating DNMT3b,

174 promoting gastric cancer progression19. Our current data were similar to those of

175 NDRG2. Noteworthy, our data showed that aberrant DNA methylation of NDRG1was

176 mainly due to DNMT3A.

177 We further investigate the association of DNA methylation of NDRG1 and DNMTs.

178 Total 338 of all 407 GC cases encompasses the data of DNA methylation of NDRG1.

179 Conversely, DNA methylation of NDRG1 was positively correlative to the expression

180 of DNMTs, and was significantly associated with DNMT1 and DNMT3A.These

181 results once again confirmed the relationship of NDRG1 mRNA expression and

182 DNMTs in GC. Whereas, NDRG1 expression was significantly inverse correlated

183 with invasion depth, and DNMT1 was positively correlated with invasion depth.

184 DNMT3B was significantly positive correlated with the degree of tumor cell

185 differentiation. But, the univariate analysis of prognosis showed no association

186 between NDRG1and DNMTs with the prognosis of GC patients. While, multivariate

187 analysis of prognosis showed only DNMT1 was significantly correlated with

188 prognosis of GC patients. These results were mainly based on the data of

189 bioinformatics, the current data remain suggest that high NDRG1 expression and low

190 DNMTs expression could suppress cell invasion and migration, may improved the

191 rate of recurrence and metastasis. Our previous study had showed GC patients with

192 high expression of NDRG1 had better overall survival rate than those with low

193 NDRG1 expression8. So large sample data of immunohistochemistry, western blot

194 and RT-PCR of GC tissues were urgently needed.

195 DNMT inhibitor, including azacitidine, 5-aza-2′-deoxycytidine (5-Aza-Dc,

196 decitabine) , guadecitabine, hydralazine, procaine, MG98 and/or zebularine, among

197 which 5-Aza-Dc is the most common used, could cause demethylating and reactivate

7 bioRxiv preprint doi: https://doi.org/10.1101/2021.03.01.433329; this version posted March 1, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

198 the expression of TSGs, then suppress the metastasis of tumor cell20-22. NDRG1

199 expression was found to be increased after treatment with 5-Aza-Dc in breast cancer,

200 prostate cancer and pancreatic cancer16-18. Lee E et al23 found that the epidrug 5-Aza

201 could up-regulated NDRG1 expression by reduction of suppressive histone marks,

202 H3K9me3 and H3K27me3 on NDRG1 promoter in prostate cancer.

203 In the current study, the aberrant DNA methylation of NDRG1was found to be

204 significantly associated with DNMT family, especially DNMT1 and DNMT3A. Our

205 previous study showed that 5-Aza-Dc could up-regulate the expression of NDRG1 in

206 GC cells. It suggests that the demethylating agent maybe a potential target drug in GC.

207 Up to now, demethylating treatment has been used for the treatment of myeloid

208 leukemias24, 25. In vitro studies of solid tumors, demethylating agents combined

209 chemotherapy and/or immunotherapy could enhances the therapeutic effect in cancer

210 cells26-29. In a case report, azacitidine was used to treat a 57-year-old woman newly

211 diagnosed with MDS during palliative chemotherapy for metastatic breast cancer,

212 azacitidine showed promising effects for MDS, and also stabilized the patient's lung

213 and lymph node metastases without any major toxicity30. Now, An increasing number

214 of clinical trials of demethylating treatment is ongoing,some have presented exciting

215 effects31-34(Table 2). A Phase I study showed the security of decitabine treated by

216 hepatic arterial infusionin, and a dose level of 20 mg/m2/day on five consecutive days

217 every 4 weeks could be considered for further investigation in combinatorial

218 immunotherapy regimens in patients with Unresectable Liver-predominant Metastases

219 From Colorectal Cancer33. In a phase I/II clinical trial, 15 patients with metastatic

220 castration-resistant prostate cancer (mCRPC) were enrolled in phase I which is dose

221 climbing experiment of 5-Aza-Dc, and 7 patients were enrolled in phase II. In phase I,

222 no dose-limiting toxicity (DLT) was observed, and most patients were well tolerated.

223 The highest level reached was 5-Aza-Dc with 150 mg/m2 daily for 5 days followed

224 by docetaxel with 75 mg/m2 on day 6, which was considered as the recommended

225 phase II dose. In phase II, 6 patients received the recommended phase II dose during

226 46 cycles. Two episodes of Grade 3 hematologic and three Grade 3 nonhematologic

8 bioRxiv preprint doi: https://doi.org/10.1101/2021.03.01.433329; this version posted March 1, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

227 toxicities were observed, and one patient died from neutropenic sepsis. Subsequently,

228 5-Aza was reduced to 75 mg/m2, and no treatment-related>Grade 3 toxicities was

229 observed in one patient. In this clinical trial, PSA response was observed in 10 of 19

230 (52.6 %) patients, and the median duration of response was 20.5 weeks.

231 Kaplane-Meier estimate of median PFS was 4.9 months, and median OS was 19.5

232 months , which were both favorable34. It suggests that the combination of azacitidine

233 and chemotherapy is active in mCRPC patients, and it maybe a new treatment in

234 future for tumors.

235 This study is mainly based on the data of bioinformatics, large sample data of

236 immunohistochemistry, western blot and RT-PCR of GC tissues were urgently

237 needed.

238 We will further detect and confirm the relationship of NDRG1 and DNMTs in GC

239 tissues.

240 In conclusion, the current data showed that aberrant DNA methylation of NDRG1

241 which could suppress cell invasion and migration, was regulated by DNMT family in

242 GC, especially DNMT1 and DNMT3A. Furthermore, the demethylating agent

243 5-Aza-Dc, a DNMT inhibitor, maybe a potential target drug. Therefore, further

244 clinical studies are warranted to evaluate and confirm the effect of demethylating

245 treatment in GC.

246 Acknowledgments This study was supported in part by a grant from the National

247 Natural Science Foundation of Hebei province of China(#H2018206180).

248 Disclosures No conflicts of interest are declared by the authors.

249 References: 250 1. Ferlay J, Colombet M, Soerjomataram I, Mathers C, Parkin DM, Piñeros M, Znaor A, Bray F. 251 Estimating the global cancer incidence and mortality in 2018:GLOBOCAN sources and methods. INT 252 J CANCER 2019; 144(8): 1941-1953. 253 254 2. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: 255 GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA 256 Cancer J Clin 2018; 68(6): 394-424. 257 258 3. Song J, Teplova M, Ishibe-Murakami S, Patel DJ. Structure-based mechanisitic insights into 9 bioRxiv preprint doi: https://doi.org/10.1101/2021.03.01.433329; this version posted March 1, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

259 DNMT1-mediated maintenance DNA methylation. SCIENCE 2012; 335(6069): 709-712. 260 261 4. Celarain N, Tomas-Roig J. Aberrant DNA methylation profile exacerbates inflammation and 262 neurodegeneration in multiple sclerosis patients. J Neuroinflammation 2020; 17: 21. 263 264 5. Hackett JA, Surani MA. DNA methylation dynamics during the mammalian life cycle. Philos 265 Trans R Soc Lond B Biol Sci 2013; 368(1609): 20110328. 266 267 6. Qu Y, Dang S, Hou P. Gene methylation in gastric cancer. CLIN CHIM ACTA 2013; 424: 53-65. 268 269 7. Chang X. Association of NDRG1 gene promoter methylation with reduced NDRG1 expression in 270 gastric cancer cells and tissue specimens. CELL BIOCHEM BIOPHYS 2013; 66(1): 93-101. 271 272 8. Chang X. NDRG1 expression is related to the progression and prognosis of gastric cancer patients 273 through modulating proliferation, invasion and cell cycle of gastric cancer cells. MOL BIOL REP 2014; 274 41(9): 6215-6223. 275 276 9. Lyko F. The DNA Methyltransferase Family: A Versatile Toolkit for Epigenetic Regulation. NAT 277 REV GENET 2018; 19(2): 81-92. 278 279 10. Baylin SB, Jones PA. A decade of exploring the cancer 280 epigenome—Biological and translational implications. NAT REV CANCER 2011; 11(10): 726-734. 281 282 11. Wang J, Lv W, Lin Z, Wang X, Bu J, Su Y. Hsa_circ_0003159 inhibits gastric cancer progression 283 by regulating miR-223-3p/NDRG1 axis. CANCER CELL INT 2020; 20: 57. 284 285 12. Chen Z, Sun J, Li T, Liu Y, Gao S, Zhi X, Zheng M. Iron chelator-induced up-regulation of 286 Ndrg1 inhibits proliferation and EMT process by targeting Wnt/β-catenin pathway in colon cancer 287 cells. Biochem Biophys Res Commun 2018; 506(1): 114-121. 288 289 13. Park KC, Paluncic J, Kovacevic Z, Richardson DR. Pharmacological targeting and the diverse 290 functions of the metastasis suppressor, NDRG1, in cancer. Free 2019; S0891-5849(19): 30254-30256. 291 292 14. Menezes SV, Kovacevic Z, Richardson DR. The metastasis suppressor NDRG1 down- 293 regulates the epidermal growth factor receptor via a lysosomal mechanism by up- 294 regulating mitogen-inducible gene 6. J BIOL CHEM 2019; 294(11): 4045-4064. 295 296 15. Chang X, Xu X, Xue X, Ma J, Li Z, Deng P, Chen J, Zhang S, Zhi Y, Dai D. NDRG1 controls 297 gastric cancer migration and invasion through regulating MMP-9. PATHOL ONCOL RES 2016; 22(4): 298 789-796. 299 300 16. Han LL. Aberrant NDRG1 methylation associated with its decreased expression and 301 clinicopathological significance in breast cancer. J BIOMED SCI 2013; 20: 52. 302 10 bioRxiv preprint doi: https://doi.org/10.1101/2021.03.01.433329; this version posted March 1, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

303 17. Li Y. Downregulation of N-myc downstream regulated gene 1 caused by the methylation of CpG 304 islands of NDRG1 promoter promotes proliferation and invasion of prostate cancer cells. INT J 305 ONCOL 2015; 47(3): 1001-1008. 306 307 18. Angst E. Epigenetic regulation affects N-myc downstream-regulated gene 1 expression indirectly 308 in pancreatic cancer cells. PANCREAS 2010; 39(5): 675-679. 309 310 19. Ling ZQ. Ndrg2 promoter hypermethylation triggered by helicobacter pylori infection correlates 311 with poor patients survival in human gastric carcinoma. Oncotarget 2015; 6(10): 8210-8225. 312 313 20. Cheishvili D. DNA Demethylation and Invasive Cancer: Implications for Therapeutics. Br J 314 Pharmacol 2015; 172(11): 2705-2715. 315 316 21. Zhou Z. DNA Methyltransferase Inhibitors and their Therapeutic Potential. CURR TOP MED 317 CHEM 2018; 18(28): 2448-2457. 318 319 22. Rygiel Katarzyna. Epigenetic therapies in patients with solid tumors: Focus on monotherapy with 320 deoxyribonucleic acid methyltransferase inhibitors and histone deacetylase inhibitors. 2019; 15(5): 321 961-970. 322 323 23. Lee E, Wang J, Jung Y, Cackowski FC, Taichman RS. Reduction of two histone marks, H3k9me3 324 and H3k27me3 by epidrug induces neuroendocrine differentiation in prostate cancer. J CELL 325 BIOCHEM 2018; 119(4): 3697-3705. 326 327 24. Sato T, Issa JJ, Kropf P. DNA Hypomethylating Drugs in Cancer Therapy. Cold Spring Harb 328 Perspect Med 2017; 7(5): a26948. 329 330 25. Dombret H, Itzykson R. How and when to decide between epigenetic therapy and 331 chemotherapy in patients with AML. Hematology Am Soc Hematol Educ Program 2017; 2017(1): 332 45-53. 333 334 26. Li XY, Wu JZ, Cao HX, Ma R, Wu JQ, Zhong YJ, Feng JF. Blockade of DNA methylation 335 enhances the therapeutic effect of gefitinib in non-small cell lung cells 336 . ONCOL REP 2013; 29(5): 1975-1982. 337 338 27. Lou YF, Zou ZZ, Chen PJ, Huang GB, Li B, Zheng DQ, Yu XR, Luo XY. Combination of 339 Gefitinib and DNA Methylation Inhibitor Decitabine Exerts Synergistic Anti-Cancer Activity in Colon 340 Cancer Cells. PLOS ONE 2014; 9(5): e97719. 341 342 28. Vijayaraghavalu S, Labhasetwar V. Nanogel-mediated delivery of a cocktail of epigenetic drugs 343 plus doxorubicin overcomes drug resistance in breast cancer cells. Drug Deliv Transl Res 2018; 8(5): 344 1289-1299. 345 346 29. Luo N, Sugiura A, Balko JM. Therapeutic potential of DNA methyltransferase inhibitors with 11 bioRxiv preprint doi: https://doi.org/10.1101/2021.03.01.433329; this version posted March 1, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

347 immune checkpoint inhibitor therapy in breast cancer. Cell Stress 2018; 2(3): 69-71. 348 349 30. Baek DW, Lee SJ, Sohn SK, Moon JH, Chae YS. Clinical Effects of Hypomethylating Agents in 350 Patients with Newly Diagnosed Myelodysplastic Syndrome Who Received DNA-Damaging 351 Chemotherapy for Metastatic Breast Cancer. J Breast Cancer. 2019; 22(4): 647-652. 352 353 31. Linnekamp JF, Butter R, Spijker R, Medema JP, van Laarhoven HWM. Clinical and biological 354 effects of demethylating agents on solid tumours - A systematic review. CANCER TREAT REV 2017; 355 54: 10-23. 356 357 32. Perrard J, Morel A, Meznad K, Paget-Bailly P, Dalstein V, Guenat D, Mourareau C, Clavel C, 358 Fauconnet S, Baguet A, Mougin C, Pretet JL. DNA Demethylation Agent 5azadC Downregulates 359 HPV16 E6 Expression in Cervical Cancer Cell Lines Independently of TBX2 Expression. ONCOL 360 LETT 2020; 19(1): 1074-1081. 361 33. Jansen YJL, Verset G, Schats K, Van Dam PJ, Seremet T, Kockx M, Van Laethem JB, B. Phase I 362 clinical trial of decitabine (5-aza-2'-deoxycytidine) administered by hepatic arterial infusion in patients 363 with unresectable liver-predominant metastases. ESMO Open. 2019; 4(2): e464. 364 365 34. Singal R, Ramachandran K, Gordian E, Quintero C, Zhao W, Reis IM. Phase I/II study of 366 azacitidine, docetaxel, and prednisone in patients with metastatic castration-resistant prostate cancer 367 previously treated with docetaxel-based therapy. Clin Genitourin Cancer 2015; 13(1): 22-31. 368 369

370

371

372

373

374

375

376

377

378

379

380

381

382

383 12 bioRxiv preprint doi: https://doi.org/10.1101/2021.03.01.433329; this version posted March 1, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

384 Caption:

385 Figure.1 Association between NDRG1 and DNMTs. A-C, NDRG1 mRNA expression

386 was negatively correlative to DNMT1, DNMT3A and DNMT3B. D-F, DNA

387 methylation of NDRG1 gene promoter was positively correlative to the expression

388 DNMT family.

389 Figure.2 The protein levels of NDRG1 and DNMTs in GES1 and two gastric cell

390 lines, SGC7901 and MKN45.A, the protein level of NDRG1 and DNMTs; B.

391 Histogram of the protein level.

392 Figure.3 Univariate analysis of prognosis of GC patients.

393 Figure.4 Multivariate analysis of prognosis of GC patients.

394 395 Table.1 Clinicopathological parameters of NDRG1 and DNMTs mRNA expression 396 from TCGA cohort 397 Table.2 Ongoing clinical trials evaluating decitabine drug in solid tumors 398

13 bioRxiv preprint doi: https://doi.org/10.1101/2021.03.01.433329; this version posted March 1, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. bioRxiv preprint doi: https://doi.org/10.1101/2021.03.01.433329; this version posted March 1, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. bioRxiv preprint doi: https://doi.org/10.1101/2021.03.01.433329; this version posted March 1, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. bioRxiv preprint doi: https://doi.org/10.1101/2021.03.01.433329; this version posted March 1, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. bioRxiv preprint doi: https://doi.org/10.1101/2021.03.01.433329; this version posted March 1, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. bioRxiv preprint doi: https://doi.org/10.1101/2021.03.01.433329; this version posted March 1, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.