Skip to main content
Top
Published in: BMC Cancer 1/2019

Open Access 01-12-2019 | Epigenetics | Research article

High methylation levels of PCDH10 predict poor prognosis in patients with pancreatic ductal adenocarcinoma

Authors: Maria Cristina Curia, Fabiana Fantini, Rossano Lattanzio, Francesca Tavano, Francesco Di Mola, Mauro Piantelli, Pasquale Battista, Pierluigi Di Sebastiano, Alessandro Cama

Published in: BMC Cancer | Issue 1/2019

Login to get access

Abstract

Background

Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal malignancies and is not a clinically homogeneous disease, but subsets of patients with distinct prognosis and response to therapy can be identified by genome-wide analyses. Mutations in major PDAC driver genes were associated with poor survival. By bioinformatics analysis, we identified protocadherins among the most frequently mutated genes in PDAC suggesting an important role of these genes in the biology of this tumor. Promoter methylation of protocadherins has been suggested as a prognostic marker in different tumors, but in PDAC this epigenetic modification has not been extensively studied. Thus, we evaluated whether promoter methylation of three frequently mutated protocadherins, PCDHAC2, PCDHGC5 and PCDH10 could be used as survival predictors in PDAC patients.

Methods

DNA extracted from 23 PDACs and adjacent non-neoplastic pancreatic tissues were bisulfite treated. Combined Bisulfite Restriction Analysis (COBRA) coupled to denaturing high-performance liquid chromatography (dHPLC) detection and bisulfite genomic sequencing (BGS) were used to determine the presence of methylated CpG dinucleotides in the promoter amplicons analyzed.

Results

In an exploratory analysis, two protocadherins showed the same pattern of CpG methylation in PDAC and adjacent non-neoplastic pancreatic tissues: lack of methylation for PCDHAC2, complete methylation for PCDHGC5. Conversely, the third protocadherin analyzed, PCDH10, showed a variable degree of CpG methylation in PDAC and absence of methylation in adjacent non-neoplastic pancreatic tissues. At Kaplan–Meier analysis, high levels of PCDH10 methylation defined according to the receiver operating characteristic (ROC) curve analysis were significantly associated with worse progression-free survival (PFS) rates (P = 0.008), but not with overall survival (OS). High levels of PCDH10 methylation were a prognostic factor influencing PFS (HR = 4.0: 95% CI, 1.3–12.3; P = 0.016), but not the OS.

Conclusions

In this study, we show for the first time that the methylation status of PCDH10 can predict prognosis in PDAC patients with a significant impact on the outcome in terms of progression-free survival. High levels of PCDH10 promoter methylation could be useful to identify patients at high risk of disease progression, contributing to a more accurate stratification of PDAC patients for personalized clinical management.
Literature
1.
go back to reference Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68:394–424.CrossRef Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68:394–424.CrossRef
2.
go back to reference Cid-Arregui A, Juare V. Perspectives in the treatment of pancreatic adenocarcinoma. World J Gastroenterol. 2015;21:9297–316.CrossRef Cid-Arregui A, Juare V. Perspectives in the treatment of pancreatic adenocarcinoma. World J Gastroenterol. 2015;21:9297–316.CrossRef
3.
go back to reference Waddell N, Pajic M, Patch AM, Chang DK, Kassahn KS, Bailey P, et al. Whole genomes redefine the mutational landscape of pancreatic cancer. Nature. 2015;518:495–501.CrossRef Waddell N, Pajic M, Patch AM, Chang DK, Kassahn KS, Bailey P, et al. Whole genomes redefine the mutational landscape of pancreatic cancer. Nature. 2015;518:495–501.CrossRef
4.
go back to reference Witkiewicz AK, McMillan EA, Balaji U, Baek G, Lin WC, Mansour J, et al. Whole-exome sequencing of pancreatic cancer defines genetic diversity and therapeutic targets. Nat Commun. 2015;6:6744.CrossRef Witkiewicz AK, McMillan EA, Balaji U, Baek G, Lin WC, Mansour J, et al. Whole-exome sequencing of pancreatic cancer defines genetic diversity and therapeutic targets. Nat Commun. 2015;6:6744.CrossRef
5.
go back to reference Bailey P, Chang DK, Nones K, Johns AL, Patch AM, Gingras MC, et al. Genomic analyses identify molecular subtypes of pancreatic cancer. Nature. 2016;531:47–52.CrossRef Bailey P, Chang DK, Nones K, Johns AL, Patch AM, Gingras MC, et al. Genomic analyses identify molecular subtypes of pancreatic cancer. Nature. 2016;531:47–52.CrossRef
6.
go back to reference Biankin AV, Waddell N, Kassahn KS, Gingras MC, Muthuswamy LB, Johns AL, et al. Pancreatic cancer genomes reveal aberrations in axon guidance pathway genes. Nature. 2012;491:399–405.CrossRef Biankin AV, Waddell N, Kassahn KS, Gingras MC, Muthuswamy LB, Johns AL, et al. Pancreatic cancer genomes reveal aberrations in axon guidance pathway genes. Nature. 2012;491:399–405.CrossRef
7.
go back to reference Vincent A, Omura N, Hong SM, Jaffe A, Eshleman J, Goggins M. Genome-wide analysis of promoter methylation associated with gene expression profile in pancreatic adenocarcinoma. Clin Cancer Res. 2011;17:4341–54.CrossRef Vincent A, Omura N, Hong SM, Jaffe A, Eshleman J, Goggins M. Genome-wide analysis of promoter methylation associated with gene expression profile in pancreatic adenocarcinoma. Clin Cancer Res. 2011;17:4341–54.CrossRef
8.
go back to reference Jones S, Zhang X, Parsons DW, Lin JC, Leary RJ, Angenendt P, et al. Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science. 2008;321:1801–6.CrossRef Jones S, Zhang X, Parsons DW, Lin JC, Leary RJ, Angenendt P, et al. Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science. 2008;321:1801–6.CrossRef
9.
go back to reference Wu Q, Zhang T, Cheng JF, Kim Y, Grimwood J, Schmutz J, et al. Comparative DNA sequence analysis of mouse and human protocadherin gene clusters. Genome Res. 2001;11:389–404.CrossRef Wu Q, Zhang T, Cheng JF, Kim Y, Grimwood J, Schmutz J, et al. Comparative DNA sequence analysis of mouse and human protocadherin gene clusters. Genome Res. 2001;11:389–404.CrossRef
10.
11.
go back to reference Morishita H, Yagi T. Protocadherin family: diversity, structure, and function. Curr Opin Cell Biol. 2007;19:584–92.CrossRef Morishita H, Yagi T. Protocadherin family: diversity, structure, and function. Curr Opin Cell Biol. 2007;19:584–92.CrossRef
12.
go back to reference Men Mah K, Weiner JA. Regulation of Wnt signaling by protocadherins. Semin Cell Dev Biol. 2017;69:158–71.CrossRef Men Mah K, Weiner JA. Regulation of Wnt signaling by protocadherins. Semin Cell Dev Biol. 2017;69:158–71.CrossRef
13.
go back to reference El Hajj N, Dittrich M, Haaf T. Epigenetic dysregulation of protocadherins in human disease. Semin Cell Dev Biol. 2017;69:158–71.CrossRef El Hajj N, Dittrich M, Haaf T. Epigenetic dysregulation of protocadherins in human disease. Semin Cell Dev Biol. 2017;69:158–71.CrossRef
14.
go back to reference Yu B, Yang H, Zhang C, Wu Q, Shao Y, Zhang J, Guan M, Wan J, Zhang W. High-resolution melting analysis of PCDH10 methylation levels in gastric, colorectal and pancreatic cancers. Neoplasma. 2010;57:247–52.CrossRef Yu B, Yang H, Zhang C, Wu Q, Shao Y, Zhang J, Guan M, Wan J, Zhang W. High-resolution melting analysis of PCDH10 methylation levels in gastric, colorectal and pancreatic cancers. Neoplasma. 2010;57:247–52.CrossRef
15.
go back to reference Hamilton SR, Aaltonen LA, editors. World Health Organization classification of Tumours. Pathology and genetics of Tumours of the digestive system. Lyon: IARC Press; 2000. Hamilton SR, Aaltonen LA, editors. World Health Organization classification of Tumours. Pathology and genetics of Tumours of the digestive system. Lyon: IARC Press; 2000.
16.
go back to reference Yu J, Cheng YY, Tao Q, Cheung KF, Lam CN, Geng H, Tian LW, Wong YP, Tong JH, Ying JM, Jin H, To KF, Chan FK, Sung JJ. Methylation of protocadherin 10, a novel tumor suppressor, is associated with poor prognosis in patients with gastric cancer. Gastroenterology. 2009;136:640–51.CrossRef Yu J, Cheng YY, Tao Q, Cheung KF, Lam CN, Geng H, Tian LW, Wong YP, Tong JH, Ying JM, Jin H, To KF, Chan FK, Sung JJ. Methylation of protocadherin 10, a novel tumor suppressor, is associated with poor prognosis in patients with gastric cancer. Gastroenterology. 2009;136:640–51.CrossRef
17.
go back to reference Ying J, Li H, Seng TJ, Langford C, Srivastava G, Tsao SW, Putti T, Murray P, Chan AT, Tao Q. Functional epigenetics identifies a protocadherin PCDH10 as a candidate tumor suppressor for nasopharyngeal, esophageal and multiple other carcinomas with frequent methylation. Oncogene. 2006;25:1070–80.CrossRef Ying J, Li H, Seng TJ, Langford C, Srivastava G, Tsao SW, Putti T, Murray P, Chan AT, Tao Q. Functional epigenetics identifies a protocadherin PCDH10 as a candidate tumor suppressor for nasopharyngeal, esophageal and multiple other carcinomas with frequent methylation. Oncogene. 2006;25:1070–80.CrossRef
18.
go back to reference Li LC, Dahiya R. MethPrimer: designing primers for methylation PCRs. Bioinformatics. 2002;18:1427–31.CrossRef Li LC, Dahiya R. MethPrimer: designing primers for methylation PCRs. Bioinformatics. 2002;18:1427–31.CrossRef
19.
go back to reference Aceto GM, Fantini F, De Iure S, Di Nicola M, Palka G, Valanzano R, Di Gregorio P, Stigliano V, Genuardi M, Battista P, Cama A, Curia MC. Correlation between mutations and mRNA expression of APC and MUTYH genes: new insight into hereditary colorectal polyposis predisposition. J Exp Clin Cancer Res. 2015;34:131.CrossRef Aceto GM, Fantini F, De Iure S, Di Nicola M, Palka G, Valanzano R, Di Gregorio P, Stigliano V, Genuardi M, Battista P, Cama A, Curia MC. Correlation between mutations and mRNA expression of APC and MUTYH genes: new insight into hereditary colorectal polyposis predisposition. J Exp Clin Cancer Res. 2015;34:131.CrossRef
20.
go back to reference Xu Y, Yang Z, Yuan H, Li Z, Li Y, Liu Q, Chen J. PCDH10 inhibits cell proliferation of multiple myeloma via the negative regulation of the Wnt/β-catenin/BCL-9 signaling pathway. Oncol Rep. 2015;34:747–54.CrossRef Xu Y, Yang Z, Yuan H, Li Z, Li Y, Liu Q, Chen J. PCDH10 inhibits cell proliferation of multiple myeloma via the negative regulation of the Wnt/β-catenin/BCL-9 signaling pathway. Oncol Rep. 2015;34:747–54.CrossRef
21.
go back to reference Blackford A, Serrano K, Wolfgang CL, Parmigiani G, Jones S, Zhang X, et al. SMAD4 gene mutations are associated with poor prognosis in pancreatic Cancer. Clin Cancer Res. 2009;15:4674–9.CrossRef Blackford A, Serrano K, Wolfgang CL, Parmigiani G, Jones S, Zhang X, et al. SMAD4 gene mutations are associated with poor prognosis in pancreatic Cancer. Clin Cancer Res. 2009;15:4674–9.CrossRef
22.
go back to reference Sato N, Fukushima N, Matsubayashi H, Iacobuzio-Donahue CA, Yeo CJ, Goggins M. Aberrant methylation of Reprimo correlates with genetic instability and predicts poor prognosis in pancreatic ductal adenocarcinoma. Cancer. 2006;107:251–7.CrossRef Sato N, Fukushima N, Matsubayashi H, Iacobuzio-Donahue CA, Yeo CJ, Goggins M. Aberrant methylation of Reprimo correlates with genetic instability and predicts poor prognosis in pancreatic ductal adenocarcinoma. Cancer. 2006;107:251–7.CrossRef
23.
go back to reference Tang X, Yin X, Xiang T, Li H, Li F, Chen L, Ren G. Protocadherin 10 is frequently downregulated by promoter methylation and functions as a tumor suppressor gene in non-small cell lung cancer. Cancer Biomark. 2013;12:11–9.CrossRef Tang X, Yin X, Xiang T, Li H, Li F, Chen L, Ren G. Protocadherin 10 is frequently downregulated by promoter methylation and functions as a tumor suppressor gene in non-small cell lung cancer. Cancer Biomark. 2013;12:11–9.CrossRef
24.
go back to reference Li Z, Chim JC, Yang M, Ye J, Wong BC, Qiao L. Role of PCDH10 and its hypermethylation in human gastric cancer. Biochim Biophys Acta. 2012;1823:298–305.CrossRef Li Z, Chim JC, Yang M, Ye J, Wong BC, Qiao L. Role of PCDH10 and its hypermethylation in human gastric cancer. Biochim Biophys Acta. 2012;1823:298–305.CrossRef
25.
go back to reference Zhong X, Shen H, Mao J, Zhang J, Han W. Epigenetic silencing of protocadherin 10 in colorectal cancer. Oncol Lett. 2017;13:2449–53.CrossRef Zhong X, Shen H, Mao J, Zhang J, Han W. Epigenetic silencing of protocadherin 10 in colorectal cancer. Oncol Lett. 2017;13:2449–53.CrossRef
26.
go back to reference Zhao Y, Yang Y, Trovik J, Sun K, Zhou L, Jiang P, Lau TS, Hoivik EA, Salvesen HB, Sun H, Wang H. A novel wnt regulatory axis in endometrioid endometrial cancer. Cancer Res. 2014;74:5103–17.CrossRef Zhao Y, Yang Y, Trovik J, Sun K, Zhou L, Jiang P, Lau TS, Hoivik EA, Salvesen HB, Sun H, Wang H. A novel wnt regulatory axis in endometrioid endometrial cancer. Cancer Res. 2014;74:5103–17.CrossRef
27.
go back to reference Yang Y, Jiang Y, Jiang M, Zhang J, Yang B, She Y, Wang W, Deng Y, Ye Y. Protocadherin 10 inhibits cell proliferation and induces apoptosis via regulation of DEP domain containing 1 in endometrial endometrioid carcinoma. Exp Mol Pathol. 2016;100:344–52.CrossRef Yang Y, Jiang Y, Jiang M, Zhang J, Yang B, She Y, Wang W, Deng Y, Ye Y. Protocadherin 10 inhibits cell proliferation and induces apoptosis via regulation of DEP domain containing 1 in endometrial endometrioid carcinoma. Exp Mol Pathol. 2016;100:344–52.CrossRef
28.
go back to reference Lin YL, Li ZG, He ZK, Guan TY, Ma JG. Clinical and prognostic significance of protocadherin-10 (PCDH10) promoter methylation in bladder cancer. J Int Med Res. 2012;40:2117–23.CrossRef Lin YL, Li ZG, He ZK, Guan TY, Ma JG. Clinical and prognostic significance of protocadherin-10 (PCDH10) promoter methylation in bladder cancer. J Int Med Res. 2012;40:2117–23.CrossRef
29.
go back to reference Narayan G, Scotto L, Neelakantan V, Kottoor SH, Wong AH, Loke SL, Mansukhani M, Pothuri B, Wright JD, Kaufmann AM, Schneider A, Arias-Pulido H, Tao Q, Murty VV. Protocadherin PCDH10, involved in tumor progression, is a frequent and early target of promoter hypermethylation in cervical cancer. Genes Chromosomes Cancer. 2009;48:983–92.CrossRef Narayan G, Scotto L, Neelakantan V, Kottoor SH, Wong AH, Loke SL, Mansukhani M, Pothuri B, Wright JD, Kaufmann AM, Schneider A, Arias-Pulido H, Tao Q, Murty VV. Protocadherin PCDH10, involved in tumor progression, is a frequent and early target of promoter hypermethylation in cervical cancer. Genes Chromosomes Cancer. 2009;48:983–92.CrossRef
30.
go back to reference Tzu-Ming J, Ming-Hong T, Hoi-Yan L, Wei-Ting W, Chun-Chieh C, Sheng-Tai T, et al. Protocadherin 10 suppresses tumorigenesis and metastasis in colorectal cancer and its genetic loss predicts adverse prognosis. Int J Cancer. 2014;135:2593–603.CrossRef Tzu-Ming J, Ming-Hong T, Hoi-Yan L, Wei-Ting W, Chun-Chieh C, Sheng-Tai T, et al. Protocadherin 10 suppresses tumorigenesis and metastasis in colorectal cancer and its genetic loss predicts adverse prognosis. Int J Cancer. 2014;135:2593–603.CrossRef
31.
go back to reference Qiu C, Bu X, Jiang Z. Protocadherin-10 acts as a tumor suppressor gene, and is frequently downregulated by promoter methylation in pancreatic cancer cells. Oncol Rep. 2016;36:383–9.CrossRef Qiu C, Bu X, Jiang Z. Protocadherin-10 acts as a tumor suppressor gene, and is frequently downregulated by promoter methylation in pancreatic cancer cells. Oncol Rep. 2016;36:383–9.CrossRef
Metadata
Title
High methylation levels of PCDH10 predict poor prognosis in patients with pancreatic ductal adenocarcinoma
Authors
Maria Cristina Curia
Fabiana Fantini
Rossano Lattanzio
Francesca Tavano
Francesco Di Mola
Mauro Piantelli
Pasquale Battista
Pierluigi Di Sebastiano
Alessandro Cama
Publication date
01-12-2019
Publisher
BioMed Central
Keyword
Epigenetics
Published in
BMC Cancer / Issue 1/2019
Electronic ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-019-5616-2

Other articles of this Issue 1/2019

BMC Cancer 1/2019 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine