A panel of DNA methylated markers predicts metastasis of pN0M0 gastric carcinoma: a prospective cohort study

Zhaojun Liu, Xiaojing Cheng, Lianhai Zhang, Jing Zhou, Dajun Deng, Jiafu Ji, Zhaojun Liu, Xiaojing Cheng, Lianhai Zhang, Jing Zhou, Dajun Deng, Jiafu Ji

Abstract

Background: The aim of this prospective study was to evaluate the feasibility of predicting GC metastasis using CDH1, GFRA1, P16 and ZNF382 DNA methylation as biomarkers.

Methods: 198 GC patients without metastasis at the time of surgery resection were recruited into the double-blind cohort (NCT02159339). Gene methylation was analysed using MethyLight assays. GC metastasis and survival data were obtained from 178 patients with 94.7% compliance during follow-up.

Results: Twenty six cases of metastasis and 5 cases of recurrence were observed in 178 cases (17.4%) during the follow-up (median, 62.7 months). The GC metastasis rate for GFRA1 methylation-positive patients was significantly reduced compared with GFRA1 methylation-negative patients (odds ratio [OR]: 0.23, 95% confidence interval [CI] 0.08-0.66). Similar results were also observed using ZNF382 methylation as a predictor (OR: 0.17, 95% CI 0.06-0.47). A risk score including methylation of GFRA1 and ZNF382 was generated. The metastasis rate was significantly increased in high-risk GC patients (OR: 4.71, 95% CI: 1.85-12.00). GC patients with high risk had a shorter overall survival, especially for patients with stage I GC (P = 0.024).

Conclusions: The combination of GFRA1 and ZNF382 methylation is a biomarker panel for the prediction of GC metastasis.

Conflict of interest statement

The authors declare no competing interests.

Figures

Fig. 1
Fig. 1
Participant flow diagram
Fig. 2
Fig. 2
Comparison of the proportions of methylated-gene alleles in gastric carcinoma (GC) and corresponding surgical margin (SM) samples from 151 GC patients using MethyLight analysis. a CDH1; b GFRA1; c P16; d ZNF382
Fig. 3
Fig. 3
ROC curves for the prediction of GC metastasis by GFRA1, ZNF382, CDH1 and P16 methylation positivity as biomarkers. The cut-off value to define methylation-positive patients is noted. a GFRA1; b ZNF382; c CDH1; d P16
Fig. 4
Fig. 4
Kaplan–Meier survival curves of gastric carcinoma (GC) patients with different GFRA1/ZNF382 methylation states and numbers of risk factors in GC tissues. a, c, e Relapse-free survival (RFS); (b, d, f) Overall survival (OS)

References

    1. Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int. J. Cancer. 2015;136:E359–E386. doi: 10.1002/ijc.29210.
    1. Bang YJ, Kim YW, Yang HK, Chung HC, Park YK,, Lee KH, et al. Adjuvant capecitabine and oxaliplatin for gastric cancer after D2 gastrectomy (CLASSIC): a phase 3 open-label, randomised controlled trial. Lancet. 2012;379:315–321. doi: 10.1016/S0140-6736(11)61873-4.
    1. Ajani JA, D'Amico TA, Almhanna K, Bentrem DJ, Chao J, Das P, et al. Gastric cancer, version 3.2016, NCCN clinical practice guidelines in oncology. J. Natl. Compr. Canc. Netw. 2016;14:1286–1312. doi: 10.6004/jnccn.2016.0137.
    1. Aguirre-Ghiso JA, Bragado P, Sosa MS. Metastasis awakening: targeting dormant cancer. Nat. Med. 2013;19:276–277. doi: 10.1038/nm.3120.
    1. Polzer B, Klein CA. Metastasis awakening: the challenges of targeting minimal residual cancer. Nat. Med. 2013;19:274–275. doi: 10.1038/nm.3121.
    1. Bass AJ. Comprehensive molecular characterization of gastric adenocarcinoma. Nature. 2014;513:202–209. doi: 10.1038/nature13480.
    1. Shimizu T, Marusawa H, Matsumoto Y, Inuzuka T, Ikeda A, Fujii Y, et al. Accumulation of somatic mutations in TP53 in gastric epithelium with Helicobacter pylori infection. Gastroenterology. 2014;147:407–417. doi: 10.1053/j.gastro.2014.04.036.
    1. Shigematsu Y, Niwa T, Yamashita S, Taniguchi H, Kushima R, Katai H, et al. Identification of a DNA methylation marker that detects the presence of lymph node metastases of gastric cancers. Onco. Lett. 2012;4:268–274. doi: 10.3892/ol.2012.708.
    1. Terashima M, Ichikawa W, Ochiai A, Katada K, Kurahashi I, Sakuramoto S, et al. TOP2A, GGH, PECAM1 are associated with hemotaogenous, lymph node, and peritoneal recurrence in stage II/III gastric cancer patients enrolled in the ACTS-GC study. Oncotarget. 2017;8:57574–57582. doi: 10.18632/oncotarget.15895.
    1. Belinsky SA. Gene-promoter hypermethylation as a biomarker in lung cancer. Nat. Rev. Cancer. 2004;4:707–717. doi: 10.1038/nrc1432.
    1. Liu ZJ, Zhang J, Gao YH, Pei LR, Zhou J, Gu LK, et al. Large-scale characterization of DNA methylation changes in human gastric carcinomas with and without metastasis. Clin. Cancer. Res. 2014;20:4598–4612. doi: 10.1158/1078-0432.CCR-13-3380.
    1. Beroukhim R, Mermel CH, Porter D, Wei G, Raychaudhuri S, Donovan J, et al. The landscape of somatic copy-number alteration across human cancers. Nature. 2010;463:899–905. doi: 10.1038/nature08822.
    1. Chen S, Sanjana NE, Zheng K, Shalem O, Lee K, Shi X, et al. Genome-wide CRISPR screen in a mouse model of tumor growth and metastasis. Cell. 2015;160:1246–1260. doi: 10.1016/j.cell.2015.02.038.
    1. Cui CH, Gan Y, Gu LK, Wilson J, Liu ZJ, Zhang BZ, et al. P16-specific DNA methylation by engineered zinc finger methyltransferase inactivates gene transcription and promotes cancer metastasis. Genome. Biol. 2015;16:252. doi: 10.1186/s13059-015-0819-6.
    1. Grady W, Willis J, Guilford P, Dunbier A, Toro T, Lynch H, et al. Methylation of the CDH1 promoter as the second genetic hit in hereditary diffuse gastric cancer. Nat. Genet. 2000;26:16–17. doi: 10.1038/79120.
    1. Corso G, Carvalho J, Marrelli D, Vindigni C, Carvalho B, Seruca R, et al. Somatic mutations and deletions of the E-cadherin gene predict poor survival of patients with gastric cancer. J. Clin. Oncol. 2013;31:868–875. doi: 10.1200/JCO.2012.44.4612.
    1. Sobin L, Gospodarowicz M, Wittekind C, editors. TNM Classification of Malignant Tumours, 7th edn. International Union Against Cancer (UICC). New York, USA: Wiley Press; 2009.
    1. International Cancer Genome C, Hudson TJ, Anderson W, Artez A, Barker AD, Bell C, et al. International network of cancer genome projects. Nature. 2010;464:993–998. doi: 10.1038/nature08987.
    1. Eads C. & Laird P. Combined bisulfite restriction analysis (COBRA). in DNA Methylation Protocols (Methods in Molecular Biology, (eds Mills K., Ramsahoye B.) vol 200, pp 53–70 (Humana Press, Totowa, New Jersey, USA, 2002).
    1. Toyooka KO, Toyooka S, Maitra A, Feng Q, Kiviat NC, Smith A, et al. Establishment and validation of real-time polymerase chain reaction method for CDH1 promoter methylation. Am J Pathol. 2002;161:629–634. doi: 10.1016/S0002-9440(10)64218-6.
    1. Zhou J, Cao J, Lu ZM, Liu HW, Deng DJ. A 115-bp MethyLight assay for detection ofp16 (CDKN2A) methylation as a diagnostic biomarker in human tissues. BMC Medical Genetics. 2011;12:67. doi: 10.1186/1471-2350-12-67.
    1. Widschwendter M, Siegmund KD, Müller HM, Fiegl H, Marth C, Muller-Holzner E, et al. Association of breast cancer DNA methylation profiles with hormone receptor status and response to tamoxifen. Cancer Res. 2004;64:3807–3813. doi: 10.1158/0008-5472.CAN-03-3852.
    1. Liu Z, Zhou J, Gu L, Deng D. Significant impact of amount of PCR input templates on various PCR-based DNA methylation analysis and countermeasure. Oncotarget. 2016;7:56447–56455.
    1. Donohue MC, Overholser R, Xu R, Vaida F. Conditional Akaike information under generalized linear and proportional hazards mixed models. Biometrika. 2011;98:685–700. doi: 10.1093/biomet/asr023.
    1. Wang YW, Zhu ML, Wang RF, Xue WJ, Zhu XR, Wang LF, et al. Predictable factors for lymph node metastasis in early gastric cancer analysis of clinicopathologic factors and biological markers. Tumor Biol. 2016;37:8567–8578. doi: 10.1007/s13277-015-4721-3.
    1. Motoyama K, Inoue H, Mimori K, Tanaka F, Kojima K, Uetake H, et al. Clinicopathological and prognostic significance of PDCD4 and microRNA-21 in human gastric cancer. Int. J. Oncol. 2010;36:1089–1095.
    1. Tanaka M, Kitajima Y, Edakuni G, Sato S, Miyazaki K. Abnormal expression of E-cadherin and beta-catenin may be a molecular marker of submucosal invasion and lymph node metastasis in early gastric cancer. Br. J. Surg. 2002;89:236–244.
    1. Arigami T, Natsugoe S, Uenosono Y, Yanagita S, Arima H, Hirata M, et al. CCR7 and CXCR4 expression predicts lymph node status including micrometastasis in gastric cancer. Int. J. Oncol. 2009;35:19–24. doi: 10.3892/ijo_00000308.
    1. Shen Z, Ye Y, Dong L, Vainionpää S, Mustonen H, Puolakkainen P, et al. Kindlin-2: a novel adhesion protein related to tumor invasion, lymph node metastasis, and patient outcome in gastric cancer. Am. J. Surg. 2012;203:222–229. doi: 10.1016/j.amjsurg.2011.06.050.
    1. Cheng Y, Geng H, Cheng SH, Liang P, Bai Y, Li J, et al. KRAB zinc finger protein ZNF382 is a proapoptotic tumor suppressor that represses multiple oncogenes and is commonly silenced in multiple carcinomas. Cancer Res. 2010;70:6516–6526. doi: 10.1158/0008-5472.CAN-09-4566.
    1. Airaksinen MS, Saarma M. The GDNF family: signalling, biological functions and therapeutic value. Nat. Rev. Neurosci. 2002;3:383–394. doi: 10.1038/nrn812.
    1. Esseghir S, Todd SK, Hunt T, Poulsom R, Plaza-Menacho I, Reis-Filho JS, et al. A role for glial cell derived neurotrophic factor induced expression by inflammatory cytokines and RET/GFR alpha1 receptor up-regulation in breast cancer. Cancer Res. 2007;67:11732–11741. doi: 10.1158/0008-5472.CAN-07-2343.
    1. Cavel O, Shomron O, Shabtay A, Vital J, Trejo-Leider L, Weizman N, et al. Endoneurial macrophages induce perineural invasion of pancreatic cancer cells by secretion of GDNF and activation of RET tyrosine kinase receptor. Cancer Res. 2012;72:5733–5743. doi: 10.1158/0008-5472.CAN-12-0764.

Source: PubMed

3
Subskrybuj