Skip to main content
Top
Published in: Cancer Cell International 1/2022

Open Access 01-12-2022 | Pancreatic Cancer | Research

DNA methylome in pancreatic cancer identified novel promoter hyper-methylation in NPY and FAIM2 genes associated with poor prognosis in Indian patient cohort

Authors: Ankita Chatterjee, Akash Bararia, Debopriyo Ganguly, Pronoy Kanti Mondal, Paromita Roy, Sudeep Banerjee, Shibajyoti Ghosh, Sumit Gulati, Supriyo Ghatak, Bitan Kumar Chattopadhay, Priyadarshi Basu, Aniruddha Chatterjee, Nilabja Sikdar

Published in: Cancer Cell International | Issue 1/2022

Login to get access

Abstract

Background

Pancreatic ductal adenocarcinoma (PDAC) is one of the leading cancers worldwide and has a poor survival, with a 5-year survival rate of only 8.5%. In this study we investigated altered DNA methylation associated with PDAC severity and prognosis.

Methods

Methylome data, generated using 450 K bead array, was compared between paired PDAC and normal samples in the TCGA cohort (n = 9) and our Indian cohort (n = 7). The total Indian Cohort (n = 75) was split into cohort 1 (n = 7), cohort 2 (n = 22), cohort 3 (n = 26) and cohort 4 (n = 20).Validation of differential methylation (6 selected CpG loci) and associated gene expression for differentially methylated genes (10 selected gDMs) were carried out in separate validation cohorts, using MSP, RT-PCR and IHC correlations between methylation and gene expression were observed in TCGA, GTEx cohorts and in validation cohorts. Kaplan–Meier survival analysis was done to study differential prognosis, during 2–5 years of follow-up.

Results

We identified 156 DMPs, mapped to 91 genes (gDMs), in PDAC; 68 (43.5%) DMPs were found to be differentially methylated both in TCGA cohort and our cohort, with significant concordance at hypo- and hyper-methylated loci. Enrichments of “regulation of ion transport”, “Interferon alpha/beta signalling”, “morphogenesis and development” and “transcriptional dysregulation” pathways were observed among 91 gDMs. Hyper-methylation of NPY and FAIM2 genes with down-regulated expression in PDAC, were significantly associated with poor prognosis in the Indian patient cohort.

Conclusions

Ethnic variations among populations may determine the altered epigenetic landscape in the PDAC patients of the Indian cohort. Our study identified novel differentially methylated genes (mainly NPY and FAIM2) and also validated the previously identified differentially methylated CpG sites associated with PDAC cancer patient’s survival. Comparative analysis of our data with TCGA and CPTAC cohorts showed that both NPY and FAIM2 hyper-methylation and down-regulations can be novel epigenetically regulated genes in the Indian patient population, statistically significantly associated with poor survival and advanced tumour stages.
Appendix
Available only for authorised users
Literature
1.
go back to reference Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2022. CA Cancer J Clin. 2022;72:7–33.PubMedCrossRef Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2022. CA Cancer J Clin. 2022;72:7–33.PubMedCrossRef
2.
go back to reference Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71:209–49.PubMedCrossRef Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71:209–49.PubMedCrossRef
3.
go back to reference Lucas AL, Malvezzi M, Carioli G, Negri E, La Vecchia C, Boffetta P, et al. Global trends in pancreatic cancer mortality from 1980 through 2013 and predictions for 2017. Clin Gastroenterol Hepatol. 2016;14:1452-1462.e4.PubMedPubMedCentralCrossRef Lucas AL, Malvezzi M, Carioli G, Negri E, La Vecchia C, Boffetta P, et al. Global trends in pancreatic cancer mortality from 1980 through 2013 and predictions for 2017. Clin Gastroenterol Hepatol. 2016;14:1452-1462.e4.PubMedPubMedCentralCrossRef
4.
go back to reference McGranahan N, Swanton C. Clonal heterogeneity and tumor evolution: past, present, and the future. Cell. 2017;168:613–28.PubMedCrossRef McGranahan N, Swanton C. Clonal heterogeneity and tumor evolution: past, present, and the future. Cell. 2017;168:613–28.PubMedCrossRef
5.
go back to reference Chatterjee A, Rodger EJ, Eccles MR. Epigenetic drivers of tumourigenesis and cancer metastasis. Semin Cancer Biol. 2018;51:149–59.PubMedCrossRef Chatterjee A, Rodger EJ, Eccles MR. Epigenetic drivers of tumourigenesis and cancer metastasis. Semin Cancer Biol. 2018;51:149–59.PubMedCrossRef
9.
go back to reference Henriksen SD, Thorlacius-Ussing O. Cell-free DNA methylation as blood-based biomarkers for pancreatic adenocarcinoma—a literature update. Epigenomes. 2021;5:8.PubMedPubMedCentralCrossRef Henriksen SD, Thorlacius-Ussing O. Cell-free DNA methylation as blood-based biomarkers for pancreatic adenocarcinoma—a literature update. Epigenomes. 2021;5:8.PubMedPubMedCentralCrossRef
10.
go back to reference Bararia A, Dey S, Gulati S, Ghatak S, Ghosh S, Banerjee S, et al. Differential methylation landscape of pancreatic ductal adenocarcinoma and its precancerous lesions. Hepatobiliary Pancreat Dis Int. 2020;19:205–17.PubMedCrossRef Bararia A, Dey S, Gulati S, Ghatak S, Ghosh S, Banerjee S, et al. Differential methylation landscape of pancreatic ductal adenocarcinoma and its precancerous lesions. Hepatobiliary Pancreat Dis Int. 2020;19:205–17.PubMedCrossRef
11.
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.PubMedPubMedCentralCrossRef 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.PubMedPubMedCentralCrossRef
13.
go back to reference Ansari D, Tingstedt B, Andersson B, Holmquist F, Sturesson C, Williamsson C, et al. Pancreatic cancer: Yesterday, today and tomorrow. Futur Oncol. 2016;12:1929–46.CrossRef Ansari D, Tingstedt B, Andersson B, Holmquist F, Sturesson C, Williamsson C, et al. Pancreatic cancer: Yesterday, today and tomorrow. Futur Oncol. 2016;12:1929–46.CrossRef
14.
go back to reference Storz P, Crawford HC. Carcinogenesis of pancreatic ductal adenocarcinoma. Gastroenterology. 2020;158:2072–81.PubMedCrossRef Storz P, Crawford HC. Carcinogenesis of pancreatic ductal adenocarcinoma. Gastroenterology. 2020;158:2072–81.PubMedCrossRef
15.
go back to reference Nones K, Waddell N, Song S, Patch AM, Miller D, Johns A, et al. Genome-wide DNA methylation patterns in pancreatic ductal adenocarcinoma reveal epigenetic deregulation of SLIT-ROBO, ITGA2 and MET signaling. Int J Cancer. 2014;135:1110–8.PubMedCrossRef Nones K, Waddell N, Song S, Patch AM, Miller D, Johns A, et al. Genome-wide DNA methylation patterns in pancreatic ductal adenocarcinoma reveal epigenetic deregulation of SLIT-ROBO, ITGA2 and MET signaling. Int J Cancer. 2014;135:1110–8.PubMedCrossRef
16.
go back to reference Aryee MJ, Jaffe AE, Corrada-Bravo H, Ladd-Acosta C, Feinberg AP, Hansen KD, et al. Minfi: a flexible and comprehensive bioconductor package for the analysis of infinium DNA methylation microarrays. Bioinformatics. 2014;30:1363–9.PubMedPubMedCentralCrossRef Aryee MJ, Jaffe AE, Corrada-Bravo H, Ladd-Acosta C, Feinberg AP, Hansen KD, et al. Minfi: a flexible and comprehensive bioconductor package for the analysis of infinium DNA methylation microarrays. Bioinformatics. 2014;30:1363–9.PubMedPubMedCentralCrossRef
17.
go back to reference Qin Y, Feng H, Chen M, Wu H, Zheng X. InfiniumPurify: an R package for estimating and accounting for tumor purity in cancer methylation research. Genes Dis. 2018;5:43–5.PubMedPubMedCentralCrossRef Qin Y, Feng H, Chen M, Wu H, Zheng X. InfiniumPurify: an R package for estimating and accounting for tumor purity in cancer methylation research. Genes Dis. 2018;5:43–5.PubMedPubMedCentralCrossRef
18.
go back to reference Lonsdale J, Thomas J, Salvatore M, Phillips R, Lo E, Shad S, et al. The Genotype-Tissue Expression (GTEx) project. Nat Genet. 2013;45:580–5.CrossRef Lonsdale J, Thomas J, Salvatore M, Phillips R, Lo E, Shad S, et al. The Genotype-Tissue Expression (GTEx) project. Nat Genet. 2013;45:580–5.CrossRef
19.
go back to reference Tang Z, Li C, Kang B, Gao G, Li C, Zhang Z. GEPIA: a web server for cancer and normal gene expression profiling and interactive analyses. Nucleic Acids Res. 2017;45:W98-102.PubMedPubMedCentralCrossRef Tang Z, Li C, Kang B, Gao G, Li C, Zhang Z. GEPIA: a web server for cancer and normal gene expression profiling and interactive analyses. Nucleic Acids Res. 2017;45:W98-102.PubMedPubMedCentralCrossRef
20.
go back to reference Zhou Y, Zhou B, Pache L, Chang M, Khodabakhshi AH, Tanaseichuk O, et al. Metascape provides a biologist-oriented resource for the analysis of systems-level datasets. Nat Commun. 2019;10:1523.PubMedPubMedCentralCrossRef Zhou Y, Zhou B, Pache L, Chang M, Khodabakhshi AH, Tanaseichuk O, et al. Metascape provides a biologist-oriented resource for the analysis of systems-level datasets. Nat Commun. 2019;10:1523.PubMedPubMedCentralCrossRef
22.
go back to reference Barretina J, Caponigro G, Stransky N, Venkatesan K, Margolin AA, Kim S, et al. The Cancer Cell Line Encyclopedia enables predictive modelling of anticancer drug sensitivity. Nature. 2012;483:603–7.PubMedPubMedCentralCrossRef Barretina J, Caponigro G, Stransky N, Venkatesan K, Margolin AA, Kim S, et al. The Cancer Cell Line Encyclopedia enables predictive modelling of anticancer drug sensitivity. Nature. 2012;483:603–7.PubMedPubMedCentralCrossRef
23.
go back to reference Li Y, Ge D, Lu C. The SMART App: an interactive web application for comprehensive DNA methylation analysis and visualization. Epigenet Chromatin. 2019;12:71.CrossRef Li Y, Ge D, Lu C. The SMART App: an interactive web application for comprehensive DNA methylation analysis and visualization. Epigenet Chromatin. 2019;12:71.CrossRef
24.
go back to reference Chandrashekar DS, Bashel B, Balasubramanya SAH, Creighton CJ, Ponce-Rodriguez I, Chakravarthi BVSK, et al. UALCAN: a portal for facilitating tumor subgroup gene expression and survival analyses. Neoplasia. 2017;19:649–58.PubMedPubMedCentralCrossRef Chandrashekar DS, Bashel B, Balasubramanya SAH, Creighton CJ, Ponce-Rodriguez I, Chakravarthi BVSK, et al. UALCAN: a portal for facilitating tumor subgroup gene expression and survival analyses. Neoplasia. 2017;19:649–58.PubMedPubMedCentralCrossRef
25.
go back to reference R Core Team. R: a language and environment for statistical computing. R Found Stat Comput. 2021. R Core Team. R: a language and environment for statistical computing. R Found Stat Comput. 2021.
27.
go back to reference Medvedeva YA, Khamis AM, Kulakovskiy IV, Ba-Alawi W, Bhuyan MSI, Kawaji H, et al. Effects of cytosine methylation on transcription factor binding sites. BMC Genomics. 2014;15:119.PubMedPubMedCentralCrossRef Medvedeva YA, Khamis AM, Kulakovskiy IV, Ba-Alawi W, Bhuyan MSI, Kawaji H, et al. Effects of cytosine methylation on transcription factor binding sites. BMC Genomics. 2014;15:119.PubMedPubMedCentralCrossRef
28.
go back to reference Overs A, Flammang M, Hervouet E, Bermont L, Pretet JL, Christophe B, et al. The detection of specific hypermethylated WIF1 and NPY genes in circulating DNA by crystal digital PCRTM is a powerful new tool for colorectal cancer diagnosis and screening. BMC Cancer. 2021;21:1092.PubMedPubMedCentralCrossRef Overs A, Flammang M, Hervouet E, Bermont L, Pretet JL, Christophe B, et al. The detection of specific hypermethylated WIF1 and NPY genes in circulating DNA by crystal digital PCRTM is a powerful new tool for colorectal cancer diagnosis and screening. BMC Cancer. 2021;21:1092.PubMedPubMedCentralCrossRef
29.
go back to reference Jensen LH, Olesen R, Petersen LN, Boysen AK, Andersen RF, Lindebjerg J, et al. NPY gene methylation as a universal, longitudinal plasma marker for evaluating the clinical benefit from last-line treatment with regorafenib in metastatic colorectal cancer. Cancers (Basel). 2019;11:1649.CrossRef Jensen LH, Olesen R, Petersen LN, Boysen AK, Andersen RF, Lindebjerg J, et al. NPY gene methylation as a universal, longitudinal plasma marker for evaluating the clinical benefit from last-line treatment with regorafenib in metastatic colorectal cancer. Cancers (Basel). 2019;11:1649.CrossRef
30.
go back to reference Bai Y, Wei C, Zhong Y, Zhang Y, Long J, Huang S, et al. Development and validation of a prognostic nomogram for gastric cancer based on DNA methylation-driven differentially expressed genes. Int J Biol Sci. 2020;16:1153–65.PubMedPubMedCentralCrossRef Bai Y, Wei C, Zhong Y, Zhang Y, Long J, Huang S, et al. Development and validation of a prognostic nomogram for gastric cancer based on DNA methylation-driven differentially expressed genes. Int J Biol Sci. 2020;16:1153–65.PubMedPubMedCentralCrossRef
32.
go back to reference Hong CJ, Yeon J, Yeo BK, Woo H, An HK, Heo W, et al. Fas-apoptotic inhibitory molecule 2 localizes to the lysosome and facilitates autophagosome-lysosome fusion through the LC3 interaction region motif-dependent interaction with LC3. FASEB J. 2020;34:161–79.PubMedCrossRef Hong CJ, Yeon J, Yeo BK, Woo H, An HK, Heo W, et al. Fas-apoptotic inhibitory molecule 2 localizes to the lysosome and facilitates autophagosome-lysosome fusion through the LC3 interaction region motif-dependent interaction with LC3. FASEB J. 2020;34:161–79.PubMedCrossRef
33.
go back to reference Kader F, Ghai M. DNA methylation-based variation between human populations. Mol Genet Genomics. 2017;292:5–35.PubMedCrossRef Kader F, Ghai M. DNA methylation-based variation between human populations. Mol Genet Genomics. 2017;292:5–35.PubMedCrossRef
34.
go back to reference Wang H, Guan Q, Nan Y, Ma Q, Zhong Y. Overexpression of human MX2 gene suppresses cell proliferation, migration, and invasion via ERK/P38/NF-κB pathway in glioblastoma cells. J Cell Biochem. 2019;120:18762–70.PubMedCrossRef Wang H, Guan Q, Nan Y, Ma Q, Zhong Y. Overexpression of human MX2 gene suppresses cell proliferation, migration, and invasion via ERK/P38/NF-κB pathway in glioblastoma cells. J Cell Biochem. 2019;120:18762–70.PubMedCrossRef
35.
go back to reference Kim JC, Ha YJ, Tak KH, Roh SA, Kwon YH, Kim CW, et al. Opposite functions of GSN and OAS2 on colorectal cancer metastasis, mediating perineural and lymphovascular invasion, respectively. PLoS ONE. 2018;13:e0202856.PubMedPubMedCentralCrossRef Kim JC, Ha YJ, Tak KH, Roh SA, Kwon YH, Kim CW, et al. Opposite functions of GSN and OAS2 on colorectal cancer metastasis, mediating perineural and lymphovascular invasion, respectively. PLoS ONE. 2018;13:e0202856.PubMedPubMedCentralCrossRef
36.
go back to reference Ribas A, Wolchok JD. Cancer immunotherapy using checkpoint blockade. Science (80−). 2018;359:1350–5.CrossRef Ribas A, Wolchok JD. Cancer immunotherapy using checkpoint blockade. Science (80−). 2018;359:1350–5.CrossRef
37.
go back to reference Fujisawa M, Kanda T, Shibata T, Sasaki R, Masuzaki R, Matsumoto N, et al. Involvement of the interferon signaling pathways in pancreatic cancer cells. Anticancer Res. 2020;40:4445–55.PubMedCrossRef Fujisawa M, Kanda T, Shibata T, Sasaki R, Masuzaki R, Matsumoto N, et al. Involvement of the interferon signaling pathways in pancreatic cancer cells. Anticancer Res. 2020;40:4445–55.PubMedCrossRef
38.
go back to reference Hansen KD, Timp W, Bravo HC, Sabunciyan S, Langmead B, McDonald OG, et al. Increased methylation variation in epigenetic domains across cancer types. Nat Genet. 2011;43:768–75.PubMedPubMedCentralCrossRef Hansen KD, Timp W, Bravo HC, Sabunciyan S, Langmead B, McDonald OG, et al. Increased methylation variation in epigenetic domains across cancer types. Nat Genet. 2011;43:768–75.PubMedPubMedCentralCrossRef
39.
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.PubMedPubMedCentralCrossRef 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.PubMedPubMedCentralCrossRef
40.
go back to reference Kinugawa Y, Uehara T, Sano K, Matsuda K, Maruyama Y, Kobayashi Y, et al. Methylation of tumor suppressor genes in autoimmune pancreatitis. Pancreas. 2017;46:614–8.PubMedCrossRef Kinugawa Y, Uehara T, Sano K, Matsuda K, Maruyama Y, Kobayashi Y, et al. Methylation of tumor suppressor genes in autoimmune pancreatitis. Pancreas. 2017;46:614–8.PubMedCrossRef
42.
go back to reference Munaron L. Systems biology of ion channels and transporters in tumor angiogenesis: an omics view. Biochim Biophys Acta Biomembr. 2015;1848:2647–56.CrossRef Munaron L. Systems biology of ion channels and transporters in tumor angiogenesis: an omics view. Biochim Biophys Acta Biomembr. 2015;1848:2647–56.CrossRef
43.
go back to reference Kang JA, Park SH, Jeong SP, Han MH, Lee CR, Lee KM, et al. Epigenetic regulation of Kcna3-encoding Kv1.3 potassium channel by cereblon contributes to regulation of CD4+ T-cell activation. Proc Natl Acad Sci USA. 2016;113:8771–6.PubMedPubMedCentralCrossRef Kang JA, Park SH, Jeong SP, Han MH, Lee CR, Lee KM, et al. Epigenetic regulation of Kcna3-encoding Kv1.3 potassium channel by cereblon contributes to regulation of CD4+ T-cell activation. Proc Natl Acad Sci USA. 2016;113:8771–6.PubMedPubMedCentralCrossRef
44.
go back to reference Patel SH, Edwards MJ, Ahmad SA. Intracellular ion channels in pancreas cancer. Cell Physiol Biochem. 2019;53:44–51.PubMedCrossRef Patel SH, Edwards MJ, Ahmad SA. Intracellular ion channels in pancreas cancer. Cell Physiol Biochem. 2019;53:44–51.PubMedCrossRef
45.
go back to reference Gregório C, Soares-Lima SC, Alemar B, Recamonde-Mendoza M, Camuzi D, de Souza-Santos PT, et al. Calcium signaling alterations caused by epigenetic mechanisms in pancreatic cancer: From early markers to prognostic impact. Cancers (Basel). 2020;12:1–22.CrossRef Gregório C, Soares-Lima SC, Alemar B, Recamonde-Mendoza M, Camuzi D, de Souza-Santos PT, et al. Calcium signaling alterations caused by epigenetic mechanisms in pancreatic cancer: From early markers to prognostic impact. Cancers (Basel). 2020;12:1–22.CrossRef
46.
go back to reference Johnson KC, Koestler DC, Fleischer T, Chen P, Jenson EG, Marotti JD, et al. DNA methylation in ductal carcinoma in situ related with future development of invasive breast cancer. Clin Epigenet. 2015;7:75.CrossRef Johnson KC, Koestler DC, Fleischer T, Chen P, Jenson EG, Marotti JD, et al. DNA methylation in ductal carcinoma in situ related with future development of invasive breast cancer. Clin Epigenet. 2015;7:75.CrossRef
47.
go back to reference Wu L, Zhao X, Shen Y, Zhang MX, Yan Y, Hou D, et al. Promoter methylation of fas apoptotic inhibitory molecule 2 gene is associated with obesity and dyslipidaemia in Chinese children. Diabetes Vasc Dis Res. 2015;12:217–20.CrossRef Wu L, Zhao X, Shen Y, Zhang MX, Yan Y, Hou D, et al. Promoter methylation of fas apoptotic inhibitory molecule 2 gene is associated with obesity and dyslipidaemia in Chinese children. Diabetes Vasc Dis Res. 2015;12:217–20.CrossRef
48.
go back to reference Kang HC, Kim JI, Chang HK, Woodard G, Choi YS, Ku JL, et al. FAIM2, as a novel diagnostic maker and a potential therapeutic target for small-cell lung cancer and atypical carcinoid. Sci Rep. 2016;6:34022.PubMedPubMedCentralCrossRef Kang HC, Kim JI, Chang HK, Woodard G, Choi YS, Ku JL, et al. FAIM2, as a novel diagnostic maker and a potential therapeutic target for small-cell lung cancer and atypical carcinoid. Sci Rep. 2016;6:34022.PubMedPubMedCentralCrossRef
49.
go back to reference Planells-Ferrer L, Urresti J, Coccia E, Galenkamp KMO, Calleja-Yagüe I, López-Soriano J, et al. Fas apoptosis inhibitory molecules: more than death-receptor antagonists in the nervous system. J Neurochem. 2016;139:11–21.PubMedCrossRef Planells-Ferrer L, Urresti J, Coccia E, Galenkamp KMO, Calleja-Yagüe I, López-Soriano J, et al. Fas apoptosis inhibitory molecules: more than death-receptor antagonists in the nervous system. J Neurochem. 2016;139:11–21.PubMedCrossRef
50.
go back to reference She K, Yang W, Li M, Xiong W, Zhou M. FAIM2 promotes non-small cell lung cancer cell growth and bone metastasis by activating the Wnt/β-catenin pathway. Front Oncol. 2021;11:3373.CrossRef She K, Yang W, Li M, Xiong W, Zhou M. FAIM2 promotes non-small cell lung cancer cell growth and bone metastasis by activating the Wnt/β-catenin pathway. Front Oncol. 2021;11:3373.CrossRef
51.
go back to reference DeMorrow S, Onori P, Venter J, Invernizzi P, Frampton G, White M, et al. Neuropeptide Y inhibits cholangiocarcinoma cell growth and invasion. Am J Physiol Cell Physiol. 2011;300:C1078–89.PubMedPubMedCentralCrossRef DeMorrow S, Onori P, Venter J, Invernizzi P, Frampton G, White M, et al. Neuropeptide Y inhibits cholangiocarcinoma cell growth and invasion. Am J Physiol Cell Physiol. 2011;300:C1078–89.PubMedPubMedCentralCrossRef
52.
go back to reference Alshalalfa M, Nguyen PL, Beltran H, Chen WS, Davicioni E, Zhao SG, et al. Transcriptomic and clinical characterization of neuropeptide Y expression in localized and metastatic prostate cancer: identification of novel prostate cancer subtype with clinical implications. Eur Urol Oncol. 2019;2:405–12.PubMedPubMedCentralCrossRef Alshalalfa M, Nguyen PL, Beltran H, Chen WS, Davicioni E, Zhao SG, et al. Transcriptomic and clinical characterization of neuropeptide Y expression in localized and metastatic prostate cancer: identification of novel prostate cancer subtype with clinical implications. Eur Urol Oncol. 2019;2:405–12.PubMedPubMedCentralCrossRef
53.
go back to reference Zhang L, Bijker MS, Herzog H. The neuropeptide Y system: pathophysiological and therapeutic implications in obesity and cancer. Pharmacol Ther. 2011;131:91–113.PubMedCrossRef Zhang L, Bijker MS, Herzog H. The neuropeptide Y system: pathophysiological and therapeutic implications in obesity and cancer. Pharmacol Ther. 2011;131:91–113.PubMedCrossRef
54.
go back to reference Sikdar N, Saha G, Dutta A, Ghosh S, Shrikhande SV, Banerjee S. Genetic alterations of periampullary and pancreatic ductal adenocarcinoma: an overview. Curr Genomics. 2018;19:444–63.PubMedPubMedCentralCrossRef Sikdar N, Saha G, Dutta A, Ghosh S, Shrikhande SV, Banerjee S. Genetic alterations of periampullary and pancreatic ductal adenocarcinoma: an overview. Curr Genomics. 2018;19:444–63.PubMedPubMedCentralCrossRef
55.
57.
go back to reference Saha G, Singh R, Mandal A, Das S, Chattopadhyay E, Panja P, et al. A novel hotspot and rare somatic mutation p.A138V, at TP53 is associated with poor survival of pancreatic ductal and periampullary adenocarcinoma patients. Mol Med. 2020;26:59.PubMedPubMedCentralCrossRef Saha G, Singh R, Mandal A, Das S, Chattopadhyay E, Panja P, et al. A novel hotspot and rare somatic mutation p.A138V, at TP53 is associated with poor survival of pancreatic ductal and periampullary adenocarcinoma patients. Mol Med. 2020;26:59.PubMedPubMedCentralCrossRef
Metadata
Title
DNA methylome in pancreatic cancer identified novel promoter hyper-methylation in NPY and FAIM2 genes associated with poor prognosis in Indian patient cohort
Authors
Ankita Chatterjee
Akash Bararia
Debopriyo Ganguly
Pronoy Kanti Mondal
Paromita Roy
Sudeep Banerjee
Shibajyoti Ghosh
Sumit Gulati
Supriyo Ghatak
Bitan Kumar Chattopadhay
Priyadarshi Basu
Aniruddha Chatterjee
Nilabja Sikdar
Publication date
01-12-2022
Publisher
BioMed Central
Published in
Cancer Cell International / Issue 1/2022
Electronic ISSN: 1475-2867
DOI
https://doi.org/10.1186/s12935-022-02737-1

Other articles of this Issue 1/2022

Cancer Cell International 1/2022 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