Skip to main content
Top
Published in: Journal of Experimental & Clinical Cancer Research 1/2018

Open Access 01-12-2018 | Review

Pancreatic cancer associated with obesity and diabetes: an alternative approach for its targeting

Authors: Ramesh Pothuraju, Satyanarayana Rachagani, Wade M. Junker, Sanjib Chaudhary, Viswanathan Saraswathi, Sukhwinder Kaur, Surinder K. Batra

Published in: Journal of Experimental & Clinical Cancer Research | Issue 1/2018

Login to get access

Abstract

Background

Pancreatic cancer (PC) is among foremost causes of cancer related deaths worldwide due to generic symptoms, lack of effective screening strategies and resistance to chemo- and radiotherapies. The risk factors associated with PC include several metabolic disorders such as obesity, insulin resistance and type 2 diabetes mellitus (T2DM). Studies have shown that obesity and T2DM are associated with PC pathogenesis; however, their role in PC initiation and development remains obscure.

Main body

Several biochemical and physiological factors associated with obesity and/or T2DM including adipokines, inflammatory mediators, and altered microbiome are involved in PC progression and metastasis albeit by different molecular mechanisms. Deep understanding of these factors and causal relationship between factors and altered signaling pathways will facilitate deconvolution of disease complexity as well as lead to development of novel therapies. In the present review, we focuses on the interplay between adipocytokines, gut microbiota, adrenomedullin, hyaluronan, vanin and matrix metalloproteinase affected by metabolic alteration and pancreatic tumor progression.

Conclusions

Metabolic diseases, such as obesity and T2DM, contribute PC development through altered metabolic pathways. Delineating key players in oncogenic development in pancreas due to metabolic disorder could be a beneficial strategy to combat cancers associated with metabolic diseases in particular, PC.
Literature
1.
go back to reference Majumder S, Chari ST, Ahlquist DA. Molecular detection of pancreatic neoplasia: Current status and future promise. World J Gastroenterol. 2015;21(40):11387–95.PubMedPubMedCentralCrossRef Majumder S, Chari ST, Ahlquist DA. Molecular detection of pancreatic neoplasia: Current status and future promise. World J Gastroenterol. 2015;21(40):11387–95.PubMedPubMedCentralCrossRef
2.
3.
go back to reference Hall BR, Cannon A, Atri P, Wichman CS, Smith LM, Ganti AK, et al. Advanced pancreatic cancer: a meta-analysis of clinical trials over thirty years. Oncotarget. 2018;9(27):19396–405.PubMedPubMedCentralCrossRef Hall BR, Cannon A, Atri P, Wichman CS, Smith LM, Ganti AK, et al. Advanced pancreatic cancer: a meta-analysis of clinical trials over thirty years. Oncotarget. 2018;9(27):19396–405.PubMedPubMedCentralCrossRef
6.
go back to reference Conlon KC, Klimstra DS, Brennan MF. Long-term survival after curative resection for pancreatic ductal adenocarcinoma. Clinicopathologic analysis of 5-year survivors. Ann Surg. 1996;223(3):273–9.PubMedPubMedCentralCrossRef Conlon KC, Klimstra DS, Brennan MF. Long-term survival after curative resection for pancreatic ductal adenocarcinoma. Clinicopathologic analysis of 5-year survivors. Ann Surg. 1996;223(3):273–9.PubMedPubMedCentralCrossRef
7.
go back to reference Chakraborty S, Baine MJ, Sasson AR, Batra SK. Current status of molecular markers for early detection of sporadic pancreatic cancer. Biochim Biophys Acta. 2011;1815(1):44–64.PubMed Chakraborty S, Baine MJ, Sasson AR, Batra SK. Current status of molecular markers for early detection of sporadic pancreatic cancer. Biochim Biophys Acta. 2011;1815(1):44–64.PubMed
8.
go back to reference Momi N, Kaur S, Ponnusamy MP, Kumar S, Wittel UA, Batra SK. Interplay between smoking-induced genotoxicity and altered signaling in pancreatic carcinogenesis. Carcinogenesis. 2012;33(9):1617–28.PubMedPubMedCentralCrossRef Momi N, Kaur S, Ponnusamy MP, Kumar S, Wittel UA, Batra SK. Interplay between smoking-induced genotoxicity and altered signaling in pancreatic carcinogenesis. Carcinogenesis. 2012;33(9):1617–28.PubMedPubMedCentralCrossRef
9.
go back to reference Chu GC, Kimmelman AC, Hezel AF, DePinho RA. Stromal biology of pancreatic cancer. J Cell Biochem. 2007;101(4):887–907.PubMedCrossRef Chu GC, Kimmelman AC, Hezel AF, DePinho RA. Stromal biology of pancreatic cancer. J Cell Biochem. 2007;101(4):887–907.PubMedCrossRef
10.
go back to reference Erkan M, Reiser-Erkan C, Michalski CW, Deucker S, Sauliunaite D, Streit S, et al. Cancer-stellate cell interactions perpetuate the hypoxia-fibrosis cycle in pancreatic ductal adenocarcinoma. Neoplasia. 2009;11(5):497–508.PubMedPubMedCentralCrossRef Erkan M, Reiser-Erkan C, Michalski CW, Deucker S, Sauliunaite D, Streit S, et al. Cancer-stellate cell interactions perpetuate the hypoxia-fibrosis cycle in pancreatic ductal adenocarcinoma. Neoplasia. 2009;11(5):497–508.PubMedPubMedCentralCrossRef
11.
go back to reference Mahadevan D, Von Hoff DD. Tumor-stroma interactions in pancreatic ductal adenocarcinoma. Mol Cancer Ther. 2007;6(4):1186–97.PubMedCrossRef Mahadevan D, Von Hoff DD. Tumor-stroma interactions in pancreatic ductal adenocarcinoma. Mol Cancer Ther. 2007;6(4):1186–97.PubMedCrossRef
12.
go back to reference Masamune A, Shimosegawa T. Signal transduction in pancreatic stellate cells. J Gastroenterol. 2009;44(4):249–60.PubMedCrossRef Masamune A, Shimosegawa T. Signal transduction in pancreatic stellate cells. J Gastroenterol. 2009;44(4):249–60.PubMedCrossRef
13.
go back to reference Cannon A, Thompson C, Hall BR, Jain M, Kumar S, Batra SK. Desmoplasia in pancreatic ductal adenocarcinoma: insight into pathological function and therapeutic potential. Genes Cancer. 2018;9(3-4):78–86.PubMedPubMedCentral Cannon A, Thompson C, Hall BR, Jain M, Kumar S, Batra SK. Desmoplasia in pancreatic ductal adenocarcinoma: insight into pathological function and therapeutic potential. Genes Cancer. 2018;9(3-4):78–86.PubMedPubMedCentral
14.
go back to reference Feldmann G, Beaty R, Hruban RH, Maitra A. Molecular genetics of pancreatic intraepithelial neoplasia. J Hepatobiliary Pancreat Surg. 2007;14(3):224–32.PubMedPubMedCentralCrossRef Feldmann G, Beaty R, Hruban RH, Maitra A. Molecular genetics of pancreatic intraepithelial neoplasia. J Hepatobiliary Pancreat Surg. 2007;14(3):224–32.PubMedPubMedCentralCrossRef
15.
go back to reference Guo J, Xie K, Zheng S. Molecular biomarkers of pancreatic intraepithelial neoplasia and their implications in early diagnosis and therapeutic intervention of pancreatic cancer. Int J Biol Sci. 2016;12(3):292–301.PubMedPubMedCentralCrossRef Guo J, Xie K, Zheng S. Molecular biomarkers of pancreatic intraepithelial neoplasia and their implications in early diagnosis and therapeutic intervention of pancreatic cancer. Int J Biol Sci. 2016;12(3):292–301.PubMedPubMedCentralCrossRef
16.
go back to reference Qin R, Smyrk TC, Reed NR, Schmidt RL, Schnelldorfer T, Chari ST, et al. Combining clinicopathological predictors and molecular biomarkers in the oncogenic K-RAS/Ki67/HIF-1alpha pathway to predict survival in resectable pancreatic cancer. Br J Cancer. 2015;112(3):514–22.PubMedPubMedCentralCrossRef Qin R, Smyrk TC, Reed NR, Schmidt RL, Schnelldorfer T, Chari ST, et al. Combining clinicopathological predictors and molecular biomarkers in the oncogenic K-RAS/Ki67/HIF-1alpha pathway to predict survival in resectable pancreatic cancer. Br J Cancer. 2015;112(3):514–22.PubMedPubMedCentralCrossRef
17.
go back to reference Kaur S, Baine MJ, Jain M, Sasson AR, Batra SK. Early diagnosis of pancreatic cancer: challenges and new developments. Biomark Med. 2012;6(5):597–612.PubMedCrossRef Kaur S, Baine MJ, Jain M, Sasson AR, Batra SK. Early diagnosis of pancreatic cancer: challenges and new developments. Biomark Med. 2012;6(5):597–612.PubMedCrossRef
18.
go back to reference Kaur S, Smith LM, Patel A, Menning M, Watley DC, Malik SS, et al. A combination of muc5ac and CA19-9 improves the diagnosis of pancreatic cancer: a multicenter study. Am J Gastroenterol. 2017;112(1):172–83.PubMedCrossRef Kaur S, Smith LM, Patel A, Menning M, Watley DC, Malik SS, et al. A combination of muc5ac and CA19-9 improves the diagnosis of pancreatic cancer: a multicenter study. Am J Gastroenterol. 2017;112(1):172–83.PubMedCrossRef
19.
go back to reference Nix GA, Schmitz PI, Wilson JH, Van BM, Groeneveld CF, Hofwijk R. Carcinoma of the head of the pancreas. Therapeutic implications of endoscopic retrograde cholangiopancreatography findings. Gastroenterology. 1984;87(1):37–43.PubMed Nix GA, Schmitz PI, Wilson JH, Van BM, Groeneveld CF, Hofwijk R. Carcinoma of the head of the pancreas. Therapeutic implications of endoscopic retrograde cholangiopancreatography findings. Gastroenterology. 1984;87(1):37–43.PubMed
20.
go back to reference Yachida S, Jones S, Bozic I, Antal T, Leary R, Fu B, et al. Distant metastasis occurs late during the genetic evolution of pancreatic cancer. Nature. 2010;467(7319):1114–7.PubMedPubMedCentralCrossRef Yachida S, Jones S, Bozic I, Antal T, Leary R, Fu B, et al. Distant metastasis occurs late during the genetic evolution of pancreatic cancer. Nature. 2010;467(7319):1114–7.PubMedPubMedCentralCrossRef
21.
go back to reference Kaur S, Kumar S, Momi N, Sasson AR, Batra SK. Mucins in pancreatic cancer and its microenvironment. Nat Rev Gastroenterol Hepatol. 2013;10(10):607–20.PubMedPubMedCentralCrossRef Kaur S, Kumar S, Momi N, Sasson AR, Batra SK. Mucins in pancreatic cancer and its microenvironment. Nat Rev Gastroenterol Hepatol. 2013;10(10):607–20.PubMedPubMedCentralCrossRef
22.
go back to reference Childs EJ, Mocci E, Campa D, Bracci PM, Gallinger S, Goggins M, et al. Common variation at 2p13.3, 3q29, 7p13 and 17q25.1 associated with susceptibility to pancreatic cancer. Nat Genet. 2015;47(8):911–6.PubMedPubMedCentralCrossRef Childs EJ, Mocci E, Campa D, Bracci PM, Gallinger S, Goggins M, et al. Common variation at 2p13.3, 3q29, 7p13 and 17q25.1 associated with susceptibility to pancreatic cancer. Nat Genet. 2015;47(8):911–6.PubMedPubMedCentralCrossRef
23.
go back to reference Herreros-Villanueva M, Bujanda L. Glypican-1 in exosomes as biomarker for early detection of pancreatic cancer. Ann Transl Med. 2016;4(4):64.PubMedPubMedCentral Herreros-Villanueva M, Bujanda L. Glypican-1 in exosomes as biomarker for early detection of pancreatic cancer. Ann Transl Med. 2016;4(4):64.PubMedPubMedCentral
24.
go back to reference Moore MJ, Goldstein D, Hamm J, Figer A, Hecht JR, Gallinger S, et al. Erlotinib plus gemcitabine compared with gemcitabine alone in patients with advanced pancreatic cancer: a phase III trial of the National Cancer Institute of Canada Clinical Trials Group. J Clin Oncol. 2007;25(15):1960–6.PubMedCrossRef Moore MJ, Goldstein D, Hamm J, Figer A, Hecht JR, Gallinger S, et al. Erlotinib plus gemcitabine compared with gemcitabine alone in patients with advanced pancreatic cancer: a phase III trial of the National Cancer Institute of Canada Clinical Trials Group. J Clin Oncol. 2007;25(15):1960–6.PubMedCrossRef
25.
go back to reference Conroy T, Desseigne F, Ychou M, Bouche O, Guimbaud R, Becouarn Y, et al. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med. 2011;364(19):1817–25.PubMedCrossRef Conroy T, Desseigne F, Ychou M, Bouche O, Guimbaud R, Becouarn Y, et al. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med. 2011;364(19):1817–25.PubMedCrossRef
26.
go back to reference Goldstein D, El-Maraghi RH, Hammel P, Heinemann V, Kunzmann V, Sastre J, et al. Nab-paclitaxel plus gemcitabine for metastatic pancreatic cancer: long-term survival from a phase III trial. J Natl Cancer Inst. 2015;107(2):dju413. https://doi.org/10.1093/jnci/dju413. Goldstein D, El-Maraghi RH, Hammel P, Heinemann V, Kunzmann V, Sastre J, et al. Nab-paclitaxel plus gemcitabine for metastatic pancreatic cancer: long-term survival from a phase III trial. J Natl Cancer Inst. 2015;107(2):dju413. https://​doi.​org/​10.​1093/​jnci/​dju413.
27.
go back to reference Von Hoff DD, Ervin T, Arena FP, Chiorean EG, Infante J, Moore M, et al. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med. 2013;369(18):1691–703.PubMedPubMedCentralCrossRef Von Hoff DD, Ervin T, Arena FP, Chiorean EG, Infante J, Moore M, et al. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med. 2013;369(18):1691–703.PubMedPubMedCentralCrossRef
28.
29.
go back to reference Liu H, Huang D, McArthur DL, Boros LG, Nissen N, Heaney AP. Fructose induces transketolase flux to promote pancreatic cancer growth. Cancer Res. 2010;70(15):6368–76.PubMedCrossRef Liu H, Huang D, McArthur DL, Boros LG, Nissen N, Heaney AP. Fructose induces transketolase flux to promote pancreatic cancer growth. Cancer Res. 2010;70(15):6368–76.PubMedCrossRef
30.
go back to reference Nadler ST, Attie AD. Please pass the chips: genomic insights into obesity and diabetes. J Nutr. 2001;131(8):2078–81.PubMedCrossRef Nadler ST, Attie AD. Please pass the chips: genomic insights into obesity and diabetes. J Nutr. 2001;131(8):2078–81.PubMedCrossRef
31.
go back to reference Pothuraju R, Sharma RK, Chagalamarri J, Jangra S, Kumar KP. A systematic review of gymnema sylvestre in obesity and diabetes management. J Sci Food Agric. 2014;94(5):834–40.PubMedCrossRef Pothuraju R, Sharma RK, Chagalamarri J, Jangra S, Kumar KP. A systematic review of gymnema sylvestre in obesity and diabetes management. J Sci Food Agric. 2014;94(5):834–40.PubMedCrossRef
32.
go back to reference Reaven G. Metabolic syndrome: pathophysiology and implications for management of cardiovascular disease. Circulation. 2002;106(3):286–8.PubMedCrossRef Reaven G. Metabolic syndrome: pathophysiology and implications for management of cardiovascular disease. Circulation. 2002;106(3):286–8.PubMedCrossRef
33.
34.
go back to reference Michaud DS, Giovannucci E, Willett WC, Colditz GA, Stampfer MJ, Fuchs CS. Physical activity, obesity, height, and the risk of pancreatic cancer. JAMA. 2001;286(8):921–9.PubMedCrossRef Michaud DS, Giovannucci E, Willett WC, Colditz GA, Stampfer MJ, Fuchs CS. Physical activity, obesity, height, and the risk of pancreatic cancer. JAMA. 2001;286(8):921–9.PubMedCrossRef
35.
go back to reference Hori M, Takahashi M, Hiraoka N, Yamaji T, Mutoh M, Ishigamori R, et al. Association of pancreatic Fatty infiltration with pancreatic ductal adenocarcinoma. Clin Transl Gastroenterol. 2014;5:e53.PubMedPubMedCentralCrossRef Hori M, Takahashi M, Hiraoka N, Yamaji T, Mutoh M, Ishigamori R, et al. Association of pancreatic Fatty infiltration with pancreatic ductal adenocarcinoma. Clin Transl Gastroenterol. 2014;5:e53.PubMedPubMedCentralCrossRef
36.
go back to reference Zyromski NJ, Mathur A, Pitt HA, Wade TE, Wang S, Nakshatri P, et al. Obesity potentiates the growth and dissemination of pancreatic cancer. Surgery. 2009;146(2):258–63.PubMedCrossRef Zyromski NJ, Mathur A, Pitt HA, Wade TE, Wang S, Nakshatri P, et al. Obesity potentiates the growth and dissemination of pancreatic cancer. Surgery. 2009;146(2):258–63.PubMedCrossRef
37.
38.
go back to reference Smits MM, van Geenen EJ. The clinical significance of pancreatic steatosis. Nat Rev Gastroenterol Hepatol. 2011;8(3):169–77.PubMedCrossRef Smits MM, van Geenen EJ. The clinical significance of pancreatic steatosis. Nat Rev Gastroenterol Hepatol. 2011;8(3):169–77.PubMedCrossRef
40.
go back to reference Renehan AG, Tyson M, Egger M, Heller RF, Zwahlen M. Body-mass index and incidence of cancer: a systematic review and meta-analysis of prospective observational studies. Lancet. 2008;371(9612):569–78.PubMedCrossRef Renehan AG, Tyson M, Egger M, Heller RF, Zwahlen M. Body-mass index and incidence of cancer: a systematic review and meta-analysis of prospective observational studies. Lancet. 2008;371(9612):569–78.PubMedCrossRef
41.
go back to reference Momi N, Kaur S, Krishn SR, Batra SK. Discovering the route from inflammation to pancreatic cancer. Minerva Gastroenterol Dietol. 2012;58(4):283–97.PubMedPubMedCentral Momi N, Kaur S, Krishn SR, Batra SK. Discovering the route from inflammation to pancreatic cancer. Minerva Gastroenterol Dietol. 2012;58(4):283–97.PubMedPubMedCentral
42.
go back to reference Incio J, Liu H, Suboj P, Chin SM, Chen IX, Pinter M, et al. Obesity-induced inflammation and desmoplasia promote pancreatic cancer progression and resistance to chemotherapy. Cancer Discov. 2016;6(8):852–69.PubMedPubMedCentralCrossRef Incio J, Liu H, Suboj P, Chin SM, Chen IX, Pinter M, et al. Obesity-induced inflammation and desmoplasia promote pancreatic cancer progression and resistance to chemotherapy. Cancer Discov. 2016;6(8):852–69.PubMedPubMedCentralCrossRef
44.
go back to reference Khandekar MJ, Cohen P, Spiegelman BM. Molecular mechanisms of cancer development in obesity. Nat Rev Cancer. 2011;11(12):886–95.PubMedCrossRef Khandekar MJ, Cohen P, Spiegelman BM. Molecular mechanisms of cancer development in obesity. Nat Rev Cancer. 2011;11(12):886–95.PubMedCrossRef
45.
go back to reference Hu E, Liang P, Spiegelman BM. AdipoQ is a novel adipose-specific gene dysregulated in obesity. J Biol Chem. 1996;271(18):10697–703.PubMedCrossRef Hu E, Liang P, Spiegelman BM. AdipoQ is a novel adipose-specific gene dysregulated in obesity. J Biol Chem. 1996;271(18):10697–703.PubMedCrossRef
46.
go back to reference Scherer PE, Williams S, Fogliano M, Baldini G, Lodish HF. A novel serum protein similar to C1q, produced exclusively in adipocytes. J Biol Chem. 1995;270(45):26746–9.PubMedCrossRef Scherer PE, Williams S, Fogliano M, Baldini G, Lodish HF. A novel serum protein similar to C1q, produced exclusively in adipocytes. J Biol Chem. 1995;270(45):26746–9.PubMedCrossRef
47.
go back to reference Wijesekara N, Krishnamurthy M, Bhattacharjee A, Suhail A, Sweeney G, Wheeler MB. Adiponectin-induced ERK and Akt phosphorylation protects against pancreatic beta cell apoptosis and increases insulin gene expression and secretion. J Biol Chem. 2010;285(44):33623–31.PubMedPubMedCentralCrossRef Wijesekara N, Krishnamurthy M, Bhattacharjee A, Suhail A, Sweeney G, Wheeler MB. Adiponectin-induced ERK and Akt phosphorylation protects against pancreatic beta cell apoptosis and increases insulin gene expression and secretion. J Biol Chem. 2010;285(44):33623–31.PubMedPubMedCentralCrossRef
48.
go back to reference Huang B, Cheng X, Wang D, Peng M, Xue Z, Da Y, et al. Adiponectin promotes pancreatic cancer progression by inhibiting apoptosis via the activation of AMPK/Sirt1/PGC-1alpha signaling. Oncotarget. 2014;5(13):4732–45.PubMedPubMedCentralCrossRef Huang B, Cheng X, Wang D, Peng M, Xue Z, Da Y, et al. Adiponectin promotes pancreatic cancer progression by inhibiting apoptosis via the activation of AMPK/Sirt1/PGC-1alpha signaling. Oncotarget. 2014;5(13):4732–45.PubMedPubMedCentralCrossRef
49.
go back to reference Kadri Colakoglu M, Bostanci EB, Ozdemir Y, Dalgic T, Aksoy E, Ozer I, et al. Roles of adiponectin and leptin as diagnostic markers in pancreatic cancer. Bratisl Lek Listy. 2017;118(7):394–8.PubMed Kadri Colakoglu M, Bostanci EB, Ozdemir Y, Dalgic T, Aksoy E, Ozer I, et al. Roles of adiponectin and leptin as diagnostic markers in pancreatic cancer. Bratisl Lek Listy. 2017;118(7):394–8.PubMed
50.
go back to reference Pezzilli R, Barassi A, Corsi MM, Morselli-Labate AM, Campana D, Casadei R, et al. Serum leptin, but not adiponectin and receptor for advanced glycation end products, is able to distinguish autoimmune pancreatitis from both chronic pancreatitis and pancreatic neoplasms. Scand J Gastroenterol. 2010;45(1):93–9.PubMedCrossRef Pezzilli R, Barassi A, Corsi MM, Morselli-Labate AM, Campana D, Casadei R, et al. Serum leptin, but not adiponectin and receptor for advanced glycation end products, is able to distinguish autoimmune pancreatitis from both chronic pancreatitis and pancreatic neoplasms. Scand J Gastroenterol. 2010;45(1):93–9.PubMedCrossRef
51.
go back to reference Bao Y, Giovannucci EL, Kraft P, Stampfer MJ, Ogino S, Ma J, et al. A prospective study of plasma adiponectin and pancreatic cancer risk in five US cohorts. J Natl Cancer Inst. 2013;105(2):95–103.PubMedCrossRef Bao Y, Giovannucci EL, Kraft P, Stampfer MJ, Ogino S, Ma J, et al. A prospective study of plasma adiponectin and pancreatic cancer risk in five US cohorts. J Natl Cancer Inst. 2013;105(2):95–103.PubMedCrossRef
52.
go back to reference Grote VA, Rohrmann S, Dossus L, Nieters A, Halkjaer J, Tjonneland A, et al. The association of circulating adiponectin levels with pancreatic cancer risk: a study within the prospective EPIC cohort. Int J Cancer. 2012;130(10):2428–37.PubMedCrossRef Grote VA, Rohrmann S, Dossus L, Nieters A, Halkjaer J, Tjonneland A, et al. The association of circulating adiponectin levels with pancreatic cancer risk: a study within the prospective EPIC cohort. Int J Cancer. 2012;130(10):2428–37.PubMedCrossRef
53.
go back to reference Kuruma S, Egawa N, Kurata M, Honda G, Kamisawa T, Ueda J, et al. Case-control study of diabetes-related genetic variants and pancreatic cancer risk in Japan. World J Gastroenterol. 2014;20(46):17456–62.PubMedPubMedCentralCrossRef Kuruma S, Egawa N, Kurata M, Honda G, Kamisawa T, Ueda J, et al. Case-control study of diabetes-related genetic variants and pancreatic cancer risk in Japan. World J Gastroenterol. 2014;20(46):17456–62.PubMedPubMedCentralCrossRef
54.
go back to reference Yang JP, Li X, Wang F, Gao M, Li SL, Chen KS. Association analysis of genetic variants of adiponectin gene and risk of pancreatic cancer. Int J Clin Exp Med. 2015;8(5):8094–100.PubMedPubMedCentral Yang JP, Li X, Wang F, Gao M, Li SL, Chen KS. Association analysis of genetic variants of adiponectin gene and risk of pancreatic cancer. Int J Clin Exp Med. 2015;8(5):8094–100.PubMedPubMedCentral
55.
go back to reference Peters KE, Beilby J, Cadby G, Warrington NM, Bruce DG, Davis WA, et al. A comprehensive investigation of variants in genes encoding adiponectin (ADIPOQ) and its receptors (ADIPOR1/R2), and their association with serum adiponectin, type 2 diabetes, insulin resistance and the metabolic syndrome. BMC Med Genet. 2013;14:15.PubMedPubMedCentralCrossRef Peters KE, Beilby J, Cadby G, Warrington NM, Bruce DG, Davis WA, et al. A comprehensive investigation of variants in genes encoding adiponectin (ADIPOQ) and its receptors (ADIPOR1/R2), and their association with serum adiponectin, type 2 diabetes, insulin resistance and the metabolic syndrome. BMC Med Genet. 2013;14:15.PubMedPubMedCentralCrossRef
56.
go back to reference Bub JD, Miyazaki T, Iwamoto Y. Adiponectin as a growth inhibitor in prostate cancer cells. Biochem Biophys Res Commun. 2006;340(4):1158–66.PubMedCrossRef Bub JD, Miyazaki T, Iwamoto Y. Adiponectin as a growth inhibitor in prostate cancer cells. Biochem Biophys Res Commun. 2006;340(4):1158–66.PubMedCrossRef
57.
go back to reference Kim AY, Lee YS, Kim KH, Lee JH, Lee HK, Jang SH, et al. Adiponectin represses colon cancer cell proliferation via AdipoR1- and -R2-mediated AMPK activation. Mol Endocrinol. 2010;24(7):1441–52.PubMedPubMedCentralCrossRef Kim AY, Lee YS, Kim KH, Lee JH, Lee HK, Jang SH, et al. Adiponectin represses colon cancer cell proliferation via AdipoR1- and -R2-mediated AMPK activation. Mol Endocrinol. 2010;24(7):1441–52.PubMedPubMedCentralCrossRef
58.
go back to reference Bao B, Wang Z, Li Y, Kong D, Ali S, Banerjee S, et al. The complexities of obesity and diabetes with the development and progression of pancreatic cancer. Biochim Biophys Acta. 2011;1815(2):135–46.PubMed Bao B, Wang Z, Li Y, Kong D, Ali S, Banerjee S, et al. The complexities of obesity and diabetes with the development and progression of pancreatic cancer. Biochim Biophys Acta. 2011;1815(2):135–46.PubMed
61.
go back to reference Lanza-Jacoby S, Yan G, Radice G, LePhong C, Baliff J, Hess R. Calorie restriction delays the progression of lesions to pancreatic cancer in the LSL-KrasG12D; Pdx-1/Cre mouse model of pancreatic cancer. Exp Biol Med (Maywood ). 2013;238(7):787–97.CrossRef Lanza-Jacoby S, Yan G, Radice G, LePhong C, Baliff J, Hess R. Calorie restriction delays the progression of lesions to pancreatic cancer in the LSL-KrasG12D; Pdx-1/Cre mouse model of pancreatic cancer. Exp Biol Med (Maywood ). 2013;238(7):787–97.CrossRef
62.
go back to reference Kato M, Watabe K, Tsujii M, Funahashi T, Shimomura I, Takehara T. Adiponectin inhibits murine pancreatic cancer growth. Dig Dis Sci. 2014;59(6):1192–6.PubMedCrossRef Kato M, Watabe K, Tsujii M, Funahashi T, Shimomura I, Takehara T. Adiponectin inhibits murine pancreatic cancer growth. Dig Dis Sci. 2014;59(6):1192–6.PubMedCrossRef
63.
go back to reference Messaggio F, Mendonsa AM, Castellanos J, Nagathihalli NS, Gorden L, Merchant NB, et al. Adiponectin receptor agonists inhibit leptin induced pSTAT3 and in vivo pancreatic tumor growth. Oncotarget. 2017;8(49):85378–91.PubMedPubMedCentralCrossRef Messaggio F, Mendonsa AM, Castellanos J, Nagathihalli NS, Gorden L, Merchant NB, et al. Adiponectin receptor agonists inhibit leptin induced pSTAT3 and in vivo pancreatic tumor growth. Oncotarget. 2017;8(49):85378–91.PubMedPubMedCentralCrossRef
65.
go back to reference Zhang Y, Guo KY, Diaz PA, Heo M, Leibel RL. Determinants of leptin gene expression in fat depots of lean mice. Am J Physiol Regul Integr Comp Physiol. 2002;282(1):R226–R34.PubMedCrossRef Zhang Y, Guo KY, Diaz PA, Heo M, Leibel RL. Determinants of leptin gene expression in fat depots of lean mice. Am J Physiol Regul Integr Comp Physiol. 2002;282(1):R226–R34.PubMedCrossRef
66.
go back to reference Ozcan L, Ergin AS, Lu A, Chung J, Sarkar S, Nie D, et al. Endoplasmic reticulum stress plays a central role in development of leptin resistance. Cell Metab. 2009;9(1):35–51.PubMedCrossRef Ozcan L, Ergin AS, Lu A, Chung J, Sarkar S, Nie D, et al. Endoplasmic reticulum stress plays a central role in development of leptin resistance. Cell Metab. 2009;9(1):35–51.PubMedCrossRef
67.
go back to reference Ren H, Jia L, Zhao T, Zhang H, Chen J, Yang S, et al. Hypoxia inducible factor (HIF)-1alpha directly activates leptin receptor (Ob-R) in pancreatic cancer cells. Cancer Lett. 2014;354(1):172–80.PubMedCrossRef Ren H, Jia L, Zhao T, Zhang H, Chen J, Yang S, et al. Hypoxia inducible factor (HIF)-1alpha directly activates leptin receptor (Ob-R) in pancreatic cancer cells. Cancer Lett. 2014;354(1):172–80.PubMedCrossRef
68.
go back to reference Fan Y, Gan Y, Shen Y, Cai X, Song Y, Zhao F, et al. Leptin signaling enhances cell invasion and promotes the metastasis of human pancreatic cancer via increasing MMP-13 production. Oncotarget. 2015;6(18):16120–34.PubMedPubMedCentralCrossRef Fan Y, Gan Y, Shen Y, Cai X, Song Y, Zhao F, et al. Leptin signaling enhances cell invasion and promotes the metastasis of human pancreatic cancer via increasing MMP-13 production. Oncotarget. 2015;6(18):16120–34.PubMedPubMedCentralCrossRef
69.
go back to reference Fujiwara Y, Kobayashi T, Chayahara N, Imamura Y, Toyoda M, Kiyota N, et al. Metabolomics evaluation of serum markers for cachexia and their intra-day variation in patients with advanced pancreatic cancer. PLoS One. 2014;9(11):e113259.PubMedPubMedCentralCrossRef Fujiwara Y, Kobayashi T, Chayahara N, Imamura Y, Toyoda M, Kiyota N, et al. Metabolomics evaluation of serum markers for cachexia and their intra-day variation in patients with advanced pancreatic cancer. PLoS One. 2014;9(11):e113259.PubMedPubMedCentralCrossRef
70.
go back to reference Harbuzariu A, Rampoldi A, Daley-Brown DS, Candelaria P, Harmon TL, Lipsey CC, et al. Leptin-notch signaling axis is involved in pancreatic cancer progression. Oncotarget. 2017;8(5):7740–52.PubMedCrossRef Harbuzariu A, Rampoldi A, Daley-Brown DS, Candelaria P, Harmon TL, Lipsey CC, et al. Leptin-notch signaling axis is involved in pancreatic cancer progression. Oncotarget. 2017;8(5):7740–52.PubMedCrossRef
72.
73.
go back to reference Mima K, Nakagawa S, Sawayama H, Ishimoto T, Imai K, Iwatsuki M, et al. The microbiome and hepatobiliary-pancreatic cancers. Cancer Lett. 2017;402:9–15.PubMedCrossRef Mima K, Nakagawa S, Sawayama H, Ishimoto T, Imai K, Iwatsuki M, et al. The microbiome and hepatobiliary-pancreatic cancers. Cancer Lett. 2017;402:9–15.PubMedCrossRef
74.
go back to reference Goyert SM, Ferrero E, Rettig WJ, Yenamandra AK, Obata F, Le Beau MM. The CD14 monocyte differentiation antigen maps to a region encoding growth factors and receptors. Science. 1988;239(4839):497–500.PubMedCrossRef Goyert SM, Ferrero E, Rettig WJ, Yenamandra AK, Obata F, Le Beau MM. The CD14 monocyte differentiation antigen maps to a region encoding growth factors and receptors. Science. 1988;239(4839):497–500.PubMedCrossRef
75.
go back to reference Haziot A, Chen S, Ferrero E, Low MG, Silber R, Goyert SM. The monocyte differentiation antigen, CD14, is anchored to the cell membrane by a phosphatidylinositol linkage. J Immunol. 1988;141(2):547–52.PubMed Haziot A, Chen S, Ferrero E, Low MG, Silber R, Goyert SM. The monocyte differentiation antigen, CD14, is anchored to the cell membrane by a phosphatidylinositol linkage. J Immunol. 1988;141(2):547–52.PubMed
76.
go back to reference Cani PD, Bibiloni R, Knauf C, Waget A, Neyrinck AM, Delzenne NM, et al. Changes in gut microbiota control metabolic endotoxemia-induced inflammation in high-fat diet-induced obesity and diabetes in mice. Diabetes. 2008;57(6):1470–81.PubMedCrossRef Cani PD, Bibiloni R, Knauf C, Waget A, Neyrinck AM, Delzenne NM, et al. Changes in gut microbiota control metabolic endotoxemia-induced inflammation in high-fat diet-induced obesity and diabetes in mice. Diabetes. 2008;57(6):1470–81.PubMedCrossRef
77.
go back to reference Ehses JA, Perren A, Eppler E, Ribaux P, Pospisilik JA, Maor-Cahn R, et al. Increased number of islet-associated macrophages in type 2 diabetes. Diabetes. 2007;56(9):2356–70.PubMedCrossRef Ehses JA, Perren A, Eppler E, Ribaux P, Pospisilik JA, Maor-Cahn R, et al. Increased number of islet-associated macrophages in type 2 diabetes. Diabetes. 2007;56(9):2356–70.PubMedCrossRef
78.
go back to reference Varma V, Yao-Borengasser A, Rasouli N, Nolen GT, Phanavanh B, Starks T, et al. Muscle inflammatory response and insulin resistance: synergistic interaction between macrophages and fatty acids leads to impaired insulin action. Am J Physiol Endocrinol Metab. 2009;296(6):E1300–E10.PubMedPubMedCentralCrossRef Varma V, Yao-Borengasser A, Rasouli N, Nolen GT, Phanavanh B, Starks T, et al. Muscle inflammatory response and insulin resistance: synergistic interaction between macrophages and fatty acids leads to impaired insulin action. Am J Physiol Endocrinol Metab. 2009;296(6):E1300–E10.PubMedPubMedCentralCrossRef
79.
80.
go back to reference Murdoch C, Muthana M, Coffelt SB, Lewis CE. The role of myeloid cells in the promotion of tumour angiogenesis. Nat Rev Cancer. 2008;8(8):618–31.PubMedCrossRef Murdoch C, Muthana M, Coffelt SB, Lewis CE. The role of myeloid cells in the promotion of tumour angiogenesis. Nat Rev Cancer. 2008;8(8):618–31.PubMedCrossRef
81.
go back to reference Pollard JW. Tumour-educated macrophages promote tumour progression and metastasis. Nat Rev Cancer. 2004;4(1):71–8.PubMedCrossRef Pollard JW. Tumour-educated macrophages promote tumour progression and metastasis. Nat Rev Cancer. 2004;4(1):71–8.PubMedCrossRef
83.
go back to reference Li Y, Kundu P, Seow SW, de Matos CT, Aronsson L, Chin KC, et al. Gut microbiota accelerate tumor growth via c-jun and STAT3 phosphorylation in APCMin/+ mice. Carcinogenesis. 2012;33(6):1231–8.PubMedCrossRef Li Y, Kundu P, Seow SW, de Matos CT, Aronsson L, Chin KC, et al. Gut microbiota accelerate tumor growth via c-jun and STAT3 phosphorylation in APCMin/+ mice. Carcinogenesis. 2012;33(6):1231–8.PubMedCrossRef
84.
go back to reference Uronis JM, Muhlbauer M, Herfarth HH, Rubinas TC, Jones GS, Jobin C. Modulation of the intestinal microbiota alters colitis-associated colorectal cancer susceptibility. PLoS One. 2009;4(6):e6026.PubMedPubMedCentralCrossRef Uronis JM, Muhlbauer M, Herfarth HH, Rubinas TC, Jones GS, Jobin C. Modulation of the intestinal microbiota alters colitis-associated colorectal cancer susceptibility. PLoS One. 2009;4(6):e6026.PubMedPubMedCentralCrossRef
85.
go back to reference Tsuji Y, Watanabe T, Kudo M, Arai H, Strober W, Chiba T. Sensing of commensal organisms by the intracellular sensor NOD1 mediates experimental pancreatitis. Immunity. 2012;37(2):326–38.PubMedPubMedCentralCrossRef Tsuji Y, Watanabe T, Kudo M, Arai H, Strober W, Chiba T. Sensing of commensal organisms by the intracellular sensor NOD1 mediates experimental pancreatitis. Immunity. 2012;37(2):326–38.PubMedPubMedCentralCrossRef
86.
go back to reference Frulloni L, Lunardi C, Simone R, Dolcino M, Scattolini C, Falconi M, et al. Identification of a novel antibody associated with autoimmune pancreatitis. N Engl J Med. 2009;361(22):2135–42.PubMedCrossRef Frulloni L, Lunardi C, Simone R, Dolcino M, Scattolini C, Falconi M, et al. Identification of a novel antibody associated with autoimmune pancreatitis. N Engl J Med. 2009;361(22):2135–42.PubMedCrossRef
87.
go back to reference Leal-Lopes C, Velloso FJ, Campopiano JC, Sogayar MC, Correa RG. Roles of commensal microbiota in pancreas homeostasis and pancreatic pathologies. J Diabetes Res. 2015;2015:284680.PubMedPubMedCentralCrossRef Leal-Lopes C, Velloso FJ, Campopiano JC, Sogayar MC, Correa RG. Roles of commensal microbiota in pancreas homeostasis and pancreatic pathologies. J Diabetes Res. 2015;2015:284680.PubMedPubMedCentralCrossRef
88.
go back to reference Sethi V, Kurtom S, Tarique M, Lavania S, Malchiodi Z, Hellmund L, et al. Gut microbiota promotes tumor growth in mice by modulating immune response. Gastroenterology. 2018;155(1):33–7 e6.PubMedCrossRef Sethi V, Kurtom S, Tarique M, Lavania S, Malchiodi Z, Hellmund L, et al. Gut microbiota promotes tumor growth in mice by modulating immune response. Gastroenterology. 2018;155(1):33–7 e6.PubMedCrossRef
89.
go back to reference Nofrarias M, Martinez-Puig D, Pujols J, Majo N, Perez JF. Long-term intake of resistant starch improves colonic mucosal integrity and reduces gut apoptosis and blood immune cells. Nutrition. 2007;23(11-12):861–70.PubMedCrossRef Nofrarias M, Martinez-Puig D, Pujols J, Majo N, Perez JF. Long-term intake of resistant starch improves colonic mucosal integrity and reduces gut apoptosis and blood immune cells. Nutrition. 2007;23(11-12):861–70.PubMedCrossRef
90.
go back to reference Panebianco C, Adamberg K, Adamberg S, Saracino C, Jaagura M, Kolk K, et al. Engineered resistant-starch (ers) diet shapes colon microbiota profile in parallel with the retardation of tumor growth in in vitro and in vivo pancreatic cancer models. Nutrients. 2017;9(4):331.PubMedCentralCrossRef Panebianco C, Adamberg K, Adamberg S, Saracino C, Jaagura M, Kolk K, et al. Engineered resistant-starch (ers) diet shapes colon microbiota profile in parallel with the retardation of tumor growth in in vitro and in vivo pancreatic cancer models. Nutrients. 2017;9(4):331.PubMedCentralCrossRef
91.
go back to reference Panebianco C, Adamberg K, Jaagura M, Copetti M, Fontana A, Adamberg S, et al. Influence of gemcitabine chemotherapy on the microbiota of pancreatic cancer xenografted mice. Cancer Chemother Pharmacol. 2018;81(4):773–82.PubMedCrossRef Panebianco C, Adamberg K, Jaagura M, Copetti M, Fontana A, Adamberg S, et al. Influence of gemcitabine chemotherapy on the microbiota of pancreatic cancer xenografted mice. Cancer Chemother Pharmacol. 2018;81(4):773–82.PubMedCrossRef
92.
go back to reference Pushalkar S, Hundeyin M, Daley D, Zambirinis CP, Kurz E, Mishra A, et al. The pancreatic cancer microbiome promotes oncogenesis by induction of innate and adaptive immune suppression. Cancer Discov. 2018;8(4):403–16.PubMedCrossRefPubMedCentral Pushalkar S, Hundeyin M, Daley D, Zambirinis CP, Kurz E, Mishra A, et al. The pancreatic cancer microbiome promotes oncogenesis by induction of innate and adaptive immune suppression. Cancer Discov. 2018;8(4):403–16.PubMedCrossRefPubMedCentral
93.
go back to reference Hotamisligil GS, Arner P, Caro JF, Atkinson RL, Spiegelman BM. Increased adipose tissue expression of tumor necrosis factor-alpha in human obesity and insulin resistance. J Clin Invest. 1995;95(5):2409–15.PubMedPubMedCentralCrossRef Hotamisligil GS, Arner P, Caro JF, Atkinson RL, Spiegelman BM. Increased adipose tissue expression of tumor necrosis factor-alpha in human obesity and insulin resistance. J Clin Invest. 1995;95(5):2409–15.PubMedPubMedCentralCrossRef
94.
go back to reference Hotamisligil GS, Shargill NS, Spiegelman BM. Adipose expression of tumor necrosis factor-alpha: direct role in obesity-linked insulin resistance. Science. 1993;259(5091):87–91.PubMedCrossRef Hotamisligil GS, Shargill NS, Spiegelman BM. Adipose expression of tumor necrosis factor-alpha: direct role in obesity-linked insulin resistance. Science. 1993;259(5091):87–91.PubMedCrossRef
95.
go back to reference Orosz P, Echtenacher B, Falk W, Ruschoff J, Weber D, Mannel DN. Enhancement of experimental metastasis by tumor necrosis factor. J Exp Med. 1993;177(5):1391–8.PubMedCrossRef Orosz P, Echtenacher B, Falk W, Ruschoff J, Weber D, Mannel DN. Enhancement of experimental metastasis by tumor necrosis factor. J Exp Med. 1993;177(5):1391–8.PubMedCrossRef
96.
go back to reference Bredel M, Scholtens DM, Yadav AK, Alvarez AA, Renfrow JJ, Chandler JP, et al. NFKBIA deletion in glioblastomas. N Engl J Med. 2011;364(7):627–37.PubMedCrossRef Bredel M, Scholtens DM, Yadav AK, Alvarez AA, Renfrow JJ, Chandler JP, et al. NFKBIA deletion in glioblastomas. N Engl J Med. 2011;364(7):627–37.PubMedCrossRef
97.
go back to reference Calado DP, Zhang B, Srinivasan L, Sasaki Y, Seagal J, Unitt C, et al. Constitutive canonical NF-kappaB activation cooperates with disruption of BLIMP1 in the pathogenesis of activated B cell-like diffuse large cell lymphoma. Cancer Cell. 2010;18(6):580–9.PubMedPubMedCentralCrossRef Calado DP, Zhang B, Srinivasan L, Sasaki Y, Seagal J, Unitt C, et al. Constitutive canonical NF-kappaB activation cooperates with disruption of BLIMP1 in the pathogenesis of activated B cell-like diffuse large cell lymphoma. Cancer Cell. 2010;18(6):580–9.PubMedPubMedCentralCrossRef
98.
go back to reference Wang W, Abbruzzese JL, Evans DB, Larry L, Cleary KR, Chiao PJ. The nuclear factor-kappa B RelA transcription factor is constitutively activated in human pancreatic adenocarcinoma cells. Clin Cancer Res. 1999;5(1):119–27.PubMed Wang W, Abbruzzese JL, Evans DB, Larry L, Cleary KR, Chiao PJ. The nuclear factor-kappa B RelA transcription factor is constitutively activated in human pancreatic adenocarcinoma cells. Clin Cancer Res. 1999;5(1):119–27.PubMed
99.
go back to reference De ML, Reni M, Tassi E, Clavenna D, Papa I, Recalde H, et al. Intratumor T helper type 2 cell infiltrate correlates with cancer-associated fibroblast thymic stromal lymphopoietin production and reduced survival in pancreatic cancer. J Exp Med. 2011;208(3):469–78.CrossRef De ML, Reni M, Tassi E, Clavenna D, Papa I, Recalde H, et al. Intratumor T helper type 2 cell infiltrate correlates with cancer-associated fibroblast thymic stromal lymphopoietin production and reduced survival in pancreatic cancer. J Exp Med. 2011;208(3):469–78.CrossRef
100.
go back to reference Demols A, Le MO, Desalle F, Quertinmont E, Van Laethem JL, Deviere J. CD4(+ )T cells play an important role in acute experimental pancreatitis in mice. Gastroenterology. 2000;118(3):582–90.PubMedCrossRef Demols A, Le MO, Desalle F, Quertinmont E, Van Laethem JL, Deviere J. CD4(+ )T cells play an important role in acute experimental pancreatitis in mice. Gastroenterology. 2000;118(3):582–90.PubMedCrossRef
101.
go back to reference Bromberg JF, Wrzeszczynska MH, Devgan G, Zhao Y, Pestell RG, Albanese C, et al. Stat3 as an oncogene. Cell. 1999;98(3):295–303.CrossRefPubMed Bromberg JF, Wrzeszczynska MH, Devgan G, Zhao Y, Pestell RG, Albanese C, et al. Stat3 as an oncogene. Cell. 1999;98(3):295–303.CrossRefPubMed
102.
go back to reference Fukuda A, Wang SC, Morris JP, Folias AE, Liou A, Kim GE, et al. Stat3 and MMP7 contribute to pancreatic ductal adenocarcinoma initiation and progression. Cancer Cell. 2011;19(4):441–55.PubMedPubMedCentralCrossRef Fukuda A, Wang SC, Morris JP, Folias AE, Liou A, Kim GE, et al. Stat3 and MMP7 contribute to pancreatic ductal adenocarcinoma initiation and progression. Cancer Cell. 2011;19(4):441–55.PubMedPubMedCentralCrossRef
103.
go back to reference Lesina M, Kurkowski MU, Ludes K, Rose-John S, Treiber M, Kloppel G, et al. Stat3/Socs3 activation by IL-6 transsignaling promotes progression of pancreatic intraepithelial neoplasia and development of pancreatic cancer. Cancer Cell. 2011;19(4):456–69.PubMedCrossRef Lesina M, Kurkowski MU, Ludes K, Rose-John S, Treiber M, Kloppel G, et al. Stat3/Socs3 activation by IL-6 transsignaling promotes progression of pancreatic intraepithelial neoplasia and development of pancreatic cancer. Cancer Cell. 2011;19(4):456–69.PubMedCrossRef
105.
go back to reference Akakura N, Kobayashi M, Horiuchi I, Suzuki A, Wang J, Chen J, et al. Constitutive expression of hypoxia-inducible factor-1alpha renders pancreatic cancer cells resistant to apoptosis induced by hypoxia and nutrient deprivation. Cancer Res. 2001;61(17):6548–54.PubMed Akakura N, Kobayashi M, Horiuchi I, Suzuki A, Wang J, Chen J, et al. Constitutive expression of hypoxia-inducible factor-1alpha renders pancreatic cancer cells resistant to apoptosis induced by hypoxia and nutrient deprivation. Cancer Res. 2001;61(17):6548–54.PubMed
106.
go back to reference Iqbal MA, Gupta V, Gopinath P, Mazurek S, Bamezai RN. Pyruvate kinase M2 and cancer: an updated assessment. FEBS Lett. 2014;588(16):2685–92.PubMedCrossRef Iqbal MA, Gupta V, Gopinath P, Mazurek S, Bamezai RN. Pyruvate kinase M2 and cancer: an updated assessment. FEBS Lett. 2014;588(16):2685–92.PubMedCrossRef
107.
go back to reference Wang HJ, Hsieh YJ, Cheng WC, Lin CP, Lin YS, Yang SF, et al. JMJD5 regulates PKM2 nuclear translocation and reprograms HIF-1alpha-mediated glucose metabolism. Proc Natl Acad Sci U S A. 2014;111(1):279–84.PubMedCrossRef Wang HJ, Hsieh YJ, Cheng WC, Lin CP, Lin YS, Yang SF, et al. JMJD5 regulates PKM2 nuclear translocation and reprograms HIF-1alpha-mediated glucose metabolism. Proc Natl Acad Sci U S A. 2014;111(1):279–84.PubMedCrossRef
108.
go back to reference Wong N, Ojo D, Yan J, Tang D. PKM2 contributes to cancer metabolism. Cancer Lett. 2015;356(2 Pt A):184–91.PubMedCrossRef Wong N, Ojo D, Yan J, Tang D. PKM2 contributes to cancer metabolism. Cancer Lett. 2015;356(2 Pt A):184–91.PubMedCrossRef
109.
go back to reference Mohammad GH, Olde Damink SW, Malago M, Dhar DK, Pereira SP. Pyruvate kinase m2 and lactate dehydrogenase a are overexpressed in pancreatic cancer and correlate with poor outcome. PLoS One. 2016;11(3):e0151635.PubMedPubMedCentralCrossRef Mohammad GH, Olde Damink SW, Malago M, Dhar DK, Pereira SP. Pyruvate kinase m2 and lactate dehydrogenase a are overexpressed in pancreatic cancer and correlate with poor outcome. PLoS One. 2016;11(3):e0151635.PubMedPubMedCentralCrossRef
110.
go back to reference Li Z, Yang P, Li Z. The multifaceted regulation and functions of PKM2 in tumor progression. Biochim Biophys Acta. 2014;1846(2):285–96.PubMed Li Z, Yang P, Li Z. The multifaceted regulation and functions of PKM2 in tumor progression. Biochim Biophys Acta. 2014;1846(2):285–96.PubMed
111.
go back to reference Yang W, Xia Y, Ji H, Zheng Y, Liang J, Huang W, et al. Nuclear PKM2 regulates beta-catenin transactivation upon EGFR activation. Nature. 2011;480(7375):118–22.PubMedPubMedCentralCrossRef Yang W, Xia Y, Ji H, Zheng Y, Liang J, Huang W, et al. Nuclear PKM2 regulates beta-catenin transactivation upon EGFR activation. Nature. 2011;480(7375):118–22.PubMedPubMedCentralCrossRef
112.
go back to reference Vonlaufen A, Joshi S, Qu C, Phillips PA, Xu Z, Parker NR, et al. Pancreatic stellate cells: partners in crime with pancreatic cancer cells. Cancer Res. 2008;68(7):2085–93.PubMedCrossRef Vonlaufen A, Joshi S, Qu C, Phillips PA, Xu Z, Parker NR, et al. Pancreatic stellate cells: partners in crime with pancreatic cancer cells. Cancer Res. 2008;68(7):2085–93.PubMedCrossRef
113.
go back to reference Vonlaufen A, Phillips PA, Xu Z, Goldstein D, Pirola RC, Wilson JS, et al. Pancreatic stellate cells and pancreatic cancer cells: an unholy alliance. Cancer Res. 2008;68(19):7707–10.PubMedCrossRef Vonlaufen A, Phillips PA, Xu Z, Goldstein D, Pirola RC, Wilson JS, et al. Pancreatic stellate cells and pancreatic cancer cells: an unholy alliance. Cancer Res. 2008;68(19):7707–10.PubMedCrossRef
114.
go back to reference Bell LN, Cai L, Johnstone BH, Traktuev DO, March KL, Considine RV. A central role for hepatocyte growth factor in adipose tissue angiogenesis. Am J Physiol Endocrinol Metab. 2008;294(2):E336–E44.PubMedCrossRef Bell LN, Cai L, Johnstone BH, Traktuev DO, March KL, Considine RV. A central role for hepatocyte growth factor in adipose tissue angiogenesis. Am J Physiol Endocrinol Metab. 2008;294(2):E336–E44.PubMedCrossRef
115.
go back to reference Jiang WG, Martin TA, Parr C, Davies G, Matsumoto K, Nakamura T. Hepatocyte growth factor, its receptor, and their potential value in cancer therapies. Crit Rev Oncol Hematol. 2005;53(1):35–69.PubMedCrossRef Jiang WG, Martin TA, Parr C, Davies G, Matsumoto K, Nakamura T. Hepatocyte growth factor, its receptor, and their potential value in cancer therapies. Crit Rev Oncol Hematol. 2005;53(1):35–69.PubMedCrossRef
116.
go back to reference Bell LN, Ward JL, Degawa-Yamauchi M, Bovenkerk JE, Jones R, Cacucci BM, et al. Adipose tissue production of hepatocyte growth factor contributes to elevated serum HGF in obesity. Am J Physiol Endocrinol Metab. 2006;291(4):E843–E8.PubMedCrossRef Bell LN, Ward JL, Degawa-Yamauchi M, Bovenkerk JE, Jones R, Cacucci BM, et al. Adipose tissue production of hepatocyte growth factor contributes to elevated serum HGF in obesity. Am J Physiol Endocrinol Metab. 2006;291(4):E843–E8.PubMedCrossRef
117.
go back to reference Ziegler KM, Considine RV, True E, Swartz-Basile DA, Pitt HA, Zyromski NJ. Adipocytes enhance murine pancreatic cancer growth via a hepatocyte growth factor (HGF)-mediated mechanism. Int J Surg. 2016;28:179–84.PubMedPubMedCentralCrossRef Ziegler KM, Considine RV, True E, Swartz-Basile DA, Pitt HA, Zyromski NJ. Adipocytes enhance murine pancreatic cancer growth via a hepatocyte growth factor (HGF)-mediated mechanism. Int J Surg. 2016;28:179–84.PubMedPubMedCentralCrossRef
118.
go back to reference Pothula SP, Xu Z, Goldstein D, Biankin AV, Pirola RC, Wilson JS, et al. Hepatocyte growth factor inhibition: a novel therapeutic approach in pancreatic cancer. Br J Cancer. 2016;114(3):269–80.PubMedPubMedCentralCrossRef Pothula SP, Xu Z, Goldstein D, Biankin AV, Pirola RC, Wilson JS, et al. Hepatocyte growth factor inhibition: a novel therapeutic approach in pancreatic cancer. Br J Cancer. 2016;114(3):269–80.PubMedPubMedCentralCrossRef
119.
go back to reference Logan-Collins J, Thomas RM, Yu P, Jaquish D, Mose E, French R, et al. Silencing of RON receptor signaling promotes apoptosis and gemcitabine sensitivity in pancreatic cancers. Cancer Res. 2010;70(3):1130–40.PubMedPubMedCentralCrossRef Logan-Collins J, Thomas RM, Yu P, Jaquish D, Mose E, French R, et al. Silencing of RON receptor signaling promotes apoptosis and gemcitabine sensitivity in pancreatic cancers. Cancer Res. 2010;70(3):1130–40.PubMedPubMedCentralCrossRef
120.
go back to reference Cui J, Xia T, Xie D, Gao Y, Jia Z, Wei D, et al. HGF/Met and FOXM1 form a positive feedback loop and render pancreatic cancer cells resistance to Met inhibition and aggressive phenotypes. Oncogene. 2016;35(36):4708–18.PubMedPubMedCentralCrossRef Cui J, Xia T, Xie D, Gao Y, Jia Z, Wei D, et al. HGF/Met and FOXM1 form a positive feedback loop and render pancreatic cancer cells resistance to Met inhibition and aggressive phenotypes. Oncogene. 2016;35(36):4708–18.PubMedPubMedCentralCrossRef
121.
go back to reference Birchmeier C, Birchmeier W, Gherardi E, Vande Woude GF. Met, metastasis, motility and more. Nat Rev Mol Cell Biol. 2003;4(12):915–25.PubMedCrossRef Birchmeier C, Birchmeier W, Gherardi E, Vande Woude GF. Met, metastasis, motility and more. Nat Rev Mol Cell Biol. 2003;4(12):915–25.PubMedCrossRef
122.
go back to reference Wong KM, Horton KJ, Coveler AL, Hingorani SR, Harris WP. Targeting the tumor stroma: the biology and clinical development of pegylated recombinant human hyaluronidase (PEGPH20). Curr Oncol Rep. 2017;19(7):47.PubMedCrossRef Wong KM, Horton KJ, Coveler AL, Hingorani SR, Harris WP. Targeting the tumor stroma: the biology and clinical development of pegylated recombinant human hyaluronidase (PEGPH20). Curr Oncol Rep. 2017;19(7):47.PubMedCrossRef
123.
go back to reference Chajara A, Raoudi M, Delpech B, Leroy M, Basuyau JP, Levesque H. Increased hyaluronan and hyaluronidase production and hyaluronan degradation in injured aorta of insulin-resistant rats. Arterioscler Thromb Vasc Biol. 2000;20(6):1480–7.PubMedCrossRef Chajara A, Raoudi M, Delpech B, Leroy M, Basuyau JP, Levesque H. Increased hyaluronan and hyaluronidase production and hyaluronan degradation in injured aorta of insulin-resistant rats. Arterioscler Thromb Vasc Biol. 2000;20(6):1480–7.PubMedCrossRef
124.
go back to reference Weiss L, Slavin S, Reich S, Cohen P, Shuster S, Stern R, et al. Induction of resistance to diabetes in non-obese diabetic mice by targeting CD44 with a specific monoclonal antibody. Proc Natl Acad Sci U S A. 2000;97(1):285–90.PubMedPubMedCentralCrossRef Weiss L, Slavin S, Reich S, Cohen P, Shuster S, Stern R, et al. Induction of resistance to diabetes in non-obese diabetic mice by targeting CD44 with a specific monoclonal antibody. Proc Natl Acad Sci U S A. 2000;97(1):285–90.PubMedPubMedCentralCrossRef
125.
go back to reference Kang L, Lantier L, Kennedy A, Bonner JS, Mayes WH, Bracy DP, et al. Hyaluronan accumulates with high-fat feeding and contributes to insulin resistance. Diabetes. 2013;62(6):1888–96.PubMedPubMedCentralCrossRef Kang L, Lantier L, Kennedy A, Bonner JS, Mayes WH, Bracy DP, et al. Hyaluronan accumulates with high-fat feeding and contributes to insulin resistance. Diabetes. 2013;62(6):1888–96.PubMedPubMedCentralCrossRef
126.
go back to reference Sato N, Cheng XB, Kohi S, Koga A, Hirata K. Targeting hyaluronan for the treatment of pancreatic ductal adenocarcinoma. Acta Pharm Sin B. 2016;6(2):101–5.PubMedPubMedCentralCrossRef Sato N, Cheng XB, Kohi S, Koga A, Hirata K. Targeting hyaluronan for the treatment of pancreatic ductal adenocarcinoma. Acta Pharm Sin B. 2016;6(2):101–5.PubMedPubMedCentralCrossRef
127.
go back to reference Itano N, Zhuo L, Kimata K. Impact of the hyaluronan-rich tumor microenvironment on cancer initiation and progression. Cancer Sci. 2008;99(9):1720–5.PubMedCrossRef Itano N, Zhuo L, Kimata K. Impact of the hyaluronan-rich tumor microenvironment on cancer initiation and progression. Cancer Sci. 2008;99(9):1720–5.PubMedCrossRef
128.
go back to reference Sato N, Kohi S, Hirata K, Goggins M. Role of hyaluronan in pancreatic cancer biology and therapy: Once again in the spotlight. Cancer Sci. 2016;107(5):569–75.PubMedPubMedCentralCrossRef Sato N, Kohi S, Hirata K, Goggins M. Role of hyaluronan in pancreatic cancer biology and therapy: Once again in the spotlight. Cancer Sci. 2016;107(5):569–75.PubMedPubMedCentralCrossRef
129.
go back to reference Sironen RK, Tammi M, Tammi R, Auvinen PK, Anttila M, Kosma VM. Hyaluronan in human malignancies. Exp Cell Res. 2011;317(4):383–91.PubMedCrossRef Sironen RK, Tammi M, Tammi R, Auvinen PK, Anttila M, Kosma VM. Hyaluronan in human malignancies. Exp Cell Res. 2011;317(4):383–91.PubMedCrossRef
130.
go back to reference Cheng XB, Sato N, Kohi S, Koga A, Hirata K. Receptor for hyaluronic acid-mediated motility is associated with poor survival in pancreatic ductal adenocarcinoma. J Cancer. 2015;6(11):1093–8.PubMedPubMedCentralCrossRef Cheng XB, Sato N, Kohi S, Koga A, Hirata K. Receptor for hyaluronic acid-mediated motility is associated with poor survival in pancreatic ductal adenocarcinoma. J Cancer. 2015;6(11):1093–8.PubMedPubMedCentralCrossRef
131.
go back to reference Immervoll H, Hoem D, Steffensen OJ, Miletic H, Molven A. Visualization of CD44 and CD133 in normal pancreas and pancreatic ductal adenocarcinomas: non-overlapping membrane expression in cell populations positive for both markers. J Histochem Cytochem. 2011;59(4):441–55.PubMedPubMedCentralCrossRef Immervoll H, Hoem D, Steffensen OJ, Miletic H, Molven A. Visualization of CD44 and CD133 in normal pancreas and pancreatic ductal adenocarcinomas: non-overlapping membrane expression in cell populations positive for both markers. J Histochem Cytochem. 2011;59(4):441–55.PubMedPubMedCentralCrossRef
132.
go back to reference Misra S, Hascall VC, Markwald RR, Ghatak S. Interactions between hyaluronan and Its receptors (CD44, RHAMM) regulate the activities of inflammation and cancer. Front Immunol. 2015;6:201.PubMedPubMedCentralCrossRef Misra S, Hascall VC, Markwald RR, Ghatak S. Interactions between hyaluronan and Its receptors (CD44, RHAMM) regulate the activities of inflammation and cancer. Front Immunol. 2015;6:201.PubMedPubMedCentralCrossRef
133.
go back to reference Theocharis AD, Tsara ME, Papageorgacopoulou N, Karavias DD, Theocharis DA. Pancreatic carcinoma is characterized by elevated content of hyaluronan and chondroitin sulfate with altered disaccharide composition. Biochim Biophys Acta. 2000;1502(2):201–6.PubMedCrossRef Theocharis AD, Tsara ME, Papageorgacopoulou N, Karavias DD, Theocharis DA. Pancreatic carcinoma is characterized by elevated content of hyaluronan and chondroitin sulfate with altered disaccharide composition. Biochim Biophys Acta. 2000;1502(2):201–6.PubMedCrossRef
134.
go back to reference Kohi S, Sato N, Cheng XB, Koga A, Higure A, Hirata K. A novel epigenetic mechanism regulating hyaluronan production in pancreatic cancer cells. Clin Exp Metastasis. 2016;33(3):225–30.PubMedCrossRef Kohi S, Sato N, Cheng XB, Koga A, Higure A, Hirata K. A novel epigenetic mechanism regulating hyaluronan production in pancreatic cancer cells. Clin Exp Metastasis. 2016;33(3):225–30.PubMedCrossRef
135.
go back to reference Cheng XB, Kohi S, Koga A, Hirata K, Sato N. Hyaluronan stimulates pancreatic cancer cell motility. Oncotarget. 2016;7(4):4829–40.PubMedCrossRef Cheng XB, Kohi S, Koga A, Hirata K, Sato N. Hyaluronan stimulates pancreatic cancer cell motility. Oncotarget. 2016;7(4):4829–40.PubMedCrossRef
136.
go back to reference Polvani S, Tarocchi M, Tempesti S, Bencini L, Galli A. Peroxisome proliferator activated receptors at the crossroad of obesity, diabetes, and pancreatic cancer. World J Gastroenterol. 2016;22(8):2441–59.PubMedPubMedCentralCrossRef Polvani S, Tarocchi M, Tempesti S, Bencini L, Galli A. Peroxisome proliferator activated receptors at the crossroad of obesity, diabetes, and pancreatic cancer. World J Gastroenterol. 2016;22(8):2441–59.PubMedPubMedCentralCrossRef
137.
go back to reference Provenzano PP, Cuevas C, Chang AE, Goel VK, Von Hoff DD, Hingorani SR. Enzymatic targeting of the stroma ablates physical barriers to treatment of pancreatic ductal adenocarcinoma. Cancer Cell. 2012;21(3):418–29.PubMedPubMedCentralCrossRef Provenzano PP, Cuevas C, Chang AE, Goel VK, Von Hoff DD, Hingorani SR. Enzymatic targeting of the stroma ablates physical barriers to treatment of pancreatic ductal adenocarcinoma. Cancer Cell. 2012;21(3):418–29.PubMedPubMedCentralCrossRef
138.
go back to reference Hingorani SR, Zheng L, Bullock AJ, Seery TE, Harris WP, Sigal DS, et al. HALO 202: randomized phase II study of PEGPH20 plus nab-paclitaxel/gemcitabine versus nab-paclitaxel/gemcitabine in patients with untreated, metastatic pancreatic ductal adenocarcinoma. J Clin Oncol. 2018;36(4):359–66.PubMedCrossRef Hingorani SR, Zheng L, Bullock AJ, Seery TE, Harris WP, Sigal DS, et al. HALO 202: randomized phase II study of PEGPH20 plus nab-paclitaxel/gemcitabine versus nab-paclitaxel/gemcitabine in patients with untreated, metastatic pancreatic ductal adenocarcinoma. J Clin Oncol. 2018;36(4):359–66.PubMedCrossRef
139.
140.
go back to reference Andersen DK. Diabetes and cancer: placing the association in perspective. Curr Opin Endocrinol Diabetes Obes. 2013;20(2):81–6.PubMedCrossRef Andersen DK. Diabetes and cancer: placing the association in perspective. Curr Opin Endocrinol Diabetes Obes. 2013;20(2):81–6.PubMedCrossRef
141.
go back to reference Setiawan VW, Stram DO, Porcel J, Chari ST, Maskarinec G, Le Marchand L, et al. Pancreatic cancer following incident diabetes in african americans and latinos: the multiethnic cohort. J Natl Cancer Inst. 2019;111(1):djy090. https://doi.org/10.1093/jnci/djy090. Setiawan VW, Stram DO, Porcel J, Chari ST, Maskarinec G, Le Marchand L, et al. Pancreatic cancer following incident diabetes in african americans and latinos: the multiethnic cohort. J Natl Cancer Inst. 2019;111(1):djy090. https://​doi.​org/​10.​1093/​jnci/​djy090.
142.
go back to reference Grote VA, Becker S, Kaaks R. Diabetes mellitus type 2 - an independent risk factor for cancer? Exp Clin Endocrinol Diabetes. 2010;118(1):4–8.PubMedCrossRef Grote VA, Becker S, Kaaks R. Diabetes mellitus type 2 - an independent risk factor for cancer? Exp Clin Endocrinol Diabetes. 2010;118(1):4–8.PubMedCrossRef
143.
go back to reference Mayer D, Shukla A, Enzmann H. Proliferative effects of insulin analogues on mammary epithelial cells. Arch Physiol Biochem. 2008;114(1):38–44.PubMedCrossRef Mayer D, Shukla A, Enzmann H. Proliferative effects of insulin analogues on mammary epithelial cells. Arch Physiol Biochem. 2008;114(1):38–44.PubMedCrossRef
144.
go back to reference Li D, Yeung SC, Hassan MM, Konopleva M, Abbruzzese JL. Antidiabetic therapies affect risk of pancreatic cancer. Gastroenterology. 2009;137(2):482–8.PubMedCrossRef Li D, Yeung SC, Hassan MM, Konopleva M, Abbruzzese JL. Antidiabetic therapies affect risk of pancreatic cancer. Gastroenterology. 2009;137(2):482–8.PubMedCrossRef
145.
go back to reference Huxley R, Ansary-Moghaddam A, de GA B, Barzi F, Woodward M. Type-II diabetes and pancreatic cancer: a meta-analysis of 36 studies. Br J Cancer. 2005;92(11):2076–83.PubMedPubMedCentralCrossRef Huxley R, Ansary-Moghaddam A, de GA B, Barzi F, Woodward M. Type-II diabetes and pancreatic cancer: a meta-analysis of 36 studies. Br J Cancer. 2005;92(11):2076–83.PubMedPubMedCentralCrossRef
146.
go back to reference Chari ST, Leibson CL, Rabe KG, Ransom J, de Andrade M, Petersen GM. Probability of pancreatic cancer following diabetes: a population-based study. Gastroenterology. 2005;129(2):504–11.PubMedCrossRef Chari ST, Leibson CL, Rabe KG, Ransom J, de Andrade M, Petersen GM. Probability of pancreatic cancer following diabetes: a population-based study. Gastroenterology. 2005;129(2):504–11.PubMedCrossRef
147.
go back to reference Ogawa Y, Tanaka M, Inoue K, Yamaguchi K, Chijiiwa K, Mizumoto K, et al. A prospective pancreatographic study of the prevalence of pancreatic carcinoma in patients with diabetes mellitus. Cancer. 2002;94(9):2344–9.PubMedCrossRef Ogawa Y, Tanaka M, Inoue K, Yamaguchi K, Chijiiwa K, Mizumoto K, et al. A prospective pancreatographic study of the prevalence of pancreatic carcinoma in patients with diabetes mellitus. Cancer. 2002;94(9):2344–9.PubMedCrossRef
148.
go back to reference Gupta D, Krueger CB, Lastra G. Over-nutrition, obesity and insulin resistance in the development of beta-cell dysfunction. Curr Diabetes Rev. 2012;8(2):76–83.PubMedCrossRef Gupta D, Krueger CB, Lastra G. Over-nutrition, obesity and insulin resistance in the development of beta-cell dysfunction. Curr Diabetes Rev. 2012;8(2):76–83.PubMedCrossRef
149.
go back to reference Kahn SE, Hull RL, Utzschneider KM. Mechanisms linking obesity to insulin resistance and type 2 diabetes. Nature. 2006;444(7121):840–6.PubMedCrossRef Kahn SE, Hull RL, Utzschneider KM. Mechanisms linking obesity to insulin resistance and type 2 diabetes. Nature. 2006;444(7121):840–6.PubMedCrossRef
151.
go back to reference Hart PA, Baichoo E, Bi Y, Hinton A, Kudva YC, Chari ST. Pancreatic polypeptide response to a mixed meal is blunted in pancreatic head cancer associated with diabetes mellitus. Pancreatology. 2015;15(2):162–6.PubMedCrossRef Hart PA, Baichoo E, Bi Y, Hinton A, Kudva YC, Chari ST. Pancreatic polypeptide response to a mixed meal is blunted in pancreatic head cancer associated with diabetes mellitus. Pancreatology. 2015;15(2):162–6.PubMedCrossRef
152.
go back to reference Aggarwal G, Ramachandran V, Javeed N, Arumugam T, Dutta S, Klee GG, et al. Adrenomedullin is up-regulated in patients with pancreatic cancer and causes insulin resistance in beta cells and mice. Gastroenterology. 2012;143(6):1510–7.PubMedCrossRef Aggarwal G, Ramachandran V, Javeed N, Arumugam T, Dutta S, Klee GG, et al. Adrenomedullin is up-regulated in patients with pancreatic cancer and causes insulin resistance in beta cells and mice. Gastroenterology. 2012;143(6):1510–7.PubMedCrossRef
153.
go back to reference Senna AA, Zedan M, el-Salam GE, el-Mashad AI. Study of plasma adrenomedullin level in normal pregnancy and preclampsia. Medscape J Med. 2008;10(2):29.PubMedPubMedCentral Senna AA, Zedan M, el-Salam GE, el-Mashad AI. Study of plasma adrenomedullin level in normal pregnancy and preclampsia. Medscape J Med. 2008;10(2):29.PubMedPubMedCentral
154.
go back to reference Ramachandran V, Arumugam T, Hwang RF, Greenson JK, Simeone DM, Logsdon CD. Adrenomedullin is expressed in pancreatic cancer and stimulates cell proliferation and invasion in an autocrine manner via the adrenomedullin receptor. ADMR. Cancer Res. 2007;67(6):2666–75.PubMedCrossRef Ramachandran V, Arumugam T, Hwang RF, Greenson JK, Simeone DM, Logsdon CD. Adrenomedullin is expressed in pancreatic cancer and stimulates cell proliferation and invasion in an autocrine manner via the adrenomedullin receptor. ADMR. Cancer Res. 2007;67(6):2666–75.PubMedCrossRef
155.
go back to reference Ramachandran V, Arumugam T, Langley R, Hwang RF, Vivas-Mejia P, Sood AK, et al. The ADMR receptor mediates the effects of adrenomedullin on pancreatic cancer cells and on cells of the tumor microenvironment. PLoS One. 2009;4(10):e7502.PubMedPubMedCentralCrossRef Ramachandran V, Arumugam T, Langley R, Hwang RF, Vivas-Mejia P, Sood AK, et al. The ADMR receptor mediates the effects of adrenomedullin on pancreatic cancer cells and on cells of the tumor microenvironment. PLoS One. 2009;4(10):e7502.PubMedPubMedCentralCrossRef
156.
go back to reference Natsuizaka M, Ozasa M, Darmanin S, Miyamoto M, Kondo S, Kamada S, et al. Synergistic up-regulation of Hexokinase-2, glucose transporters and angiogenic factors in pancreatic cancer cells by glucose deprivation and hypoxia. Exp Cell Res. 2007;313(15):3337–48.PubMedCrossRef Natsuizaka M, Ozasa M, Darmanin S, Miyamoto M, Kondo S, Kamada S, et al. Synergistic up-regulation of Hexokinase-2, glucose transporters and angiogenic factors in pancreatic cancer cells by glucose deprivation and hypoxia. Exp Cell Res. 2007;313(15):3337–48.PubMedCrossRef
157.
go back to reference Melo SA, Luecke LB, Kahlert C, Fernandez AF, Gammon ST, Kaye J, et al. Glypican-1 identifies cancer exosomes and detects early pancreatic cancer. Nature. 2015;523(7559):177–82.PubMedPubMedCentralCrossRef Melo SA, Luecke LB, Kahlert C, Fernandez AF, Gammon ST, Kaye J, et al. Glypican-1 identifies cancer exosomes and detects early pancreatic cancer. Nature. 2015;523(7559):177–82.PubMedPubMedCentralCrossRef
158.
go back to reference AS EL, Mager I, Breakefield XO, Wood MJ. Extracellular vesicles: biology and emerging therapeutic opportunities. Nat Rev Drug Discov. 2013;12(5):347–57.CrossRef AS EL, Mager I, Breakefield XO, Wood MJ. Extracellular vesicles: biology and emerging therapeutic opportunities. Nat Rev Drug Discov. 2013;12(5):347–57.CrossRef
159.
go back to reference Mathivanan S, Ji H, Simpson RJ. Exosomes: extracellular organelles important in intercellular communication. J Proteomics. 2010;73(10):1907–20.PubMedCrossRef Mathivanan S, Ji H, Simpson RJ. Exosomes: extracellular organelles important in intercellular communication. J Proteomics. 2010;73(10):1907–20.PubMedCrossRef
160.
161.
go back to reference Costa-Silva B, Aiello NM, Ocean AJ, Singh S, Zhang H, Thakur BK, et al. Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver. Nat Cell Biol. 2015;17(6):816–26.PubMedPubMedCentralCrossRef Costa-Silva B, Aiello NM, Ocean AJ, Singh S, Zhang H, Thakur BK, et al. Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver. Nat Cell Biol. 2015;17(6):816–26.PubMedPubMedCentralCrossRef
162.
go back to reference Hoshino A, Costa-Silva B, Shen TL, Rodrigues G, Hashimoto A, Tesic MM, et al. Tumour exosome integrins determine organotropic metastasis. Nature. 2015;527(7578):329–35.PubMedPubMedCentralCrossRef Hoshino A, Costa-Silva B, Shen TL, Rodrigues G, Hashimoto A, Tesic MM, et al. Tumour exosome integrins determine organotropic metastasis. Nature. 2015;527(7578):329–35.PubMedPubMedCentralCrossRef
163.
go back to reference Javeed N, Sagar G, Dutta SK, Smyrk TC, Lau JS, Bhattacharya S, et al. Pancreatic cancer-derived exosomes cause paraneoplastic beta-cell dysfunction. Clin Cancer Res. 2015;21(7):1722–33.PubMedCrossRef Javeed N, Sagar G, Dutta SK, Smyrk TC, Lau JS, Bhattacharya S, et al. Pancreatic cancer-derived exosomes cause paraneoplastic beta-cell dysfunction. Clin Cancer Res. 2015;21(7):1722–33.PubMedCrossRef
164.
go back to reference Sagar G, Sah RP, Javeed N, Dutta SK, Smyrk TC, Lau JS, et al. Pathogenesis of pancreatic cancer exosome-induced lipolysis in adipose tissue. Gut. 2016;65(7):1165–74. Sagar G, Sah RP, Javeed N, Dutta SK, Smyrk TC, Lau JS, et al. Pathogenesis of pancreatic cancer exosome-induced lipolysis in adipose tissue. Gut. 2016;65(7):1165–74.
165.
go back to reference Hollander LL, Guo X, Salem RR, Cha CH. The novel tumor angiogenic factor, adrenomedullin-2, predicts survival in pancreatic adenocarcinoma. J Surg Res. 2015;197(2):219–24.PubMedCrossRef Hollander LL, Guo X, Salem RR, Cha CH. The novel tumor angiogenic factor, adrenomedullin-2, predicts survival in pancreatic adenocarcinoma. J Surg Res. 2015;197(2):219–24.PubMedCrossRef
166.
go back to reference Maras B, Barra D, Dupre S, Pitari G. Is pantetheinase the actual identity of mouse and human vanin-1 proteins? FEBS Lett. 1999;461(3):149–52.PubMedCrossRef Maras B, Barra D, Dupre S, Pitari G. Is pantetheinase the actual identity of mouse and human vanin-1 proteins? FEBS Lett. 1999;461(3):149–52.PubMedCrossRef
167.
go back to reference Pitari G, Malergue F, Martin F, Philippe JM, Massucci MT, Chabret C, et al. Pantetheinase activity of membrane-bound vanin-1: lack of free cysteamine in tissues of vanin-1 deficient mice. FEBS Lett. 2000;483(2-3):149–54.PubMedCrossRef Pitari G, Malergue F, Martin F, Philippe JM, Massucci MT, Chabret C, et al. Pantetheinase activity of membrane-bound vanin-1: lack of free cysteamine in tissues of vanin-1 deficient mice. FEBS Lett. 2000;483(2-3):149–54.PubMedCrossRef
168.
go back to reference Dupre S, Graziani MT, Rosei MA, Fabi A, Del GE. The enzymatic breakdown of pantethine to pantothenic acid and cystamine. Eur J Biochem. 1970;16(3):571–8.PubMedCrossRef Dupre S, Graziani MT, Rosei MA, Fabi A, Del GE. The enzymatic breakdown of pantethine to pantothenic acid and cystamine. Eur J Biochem. 1970;16(3):571–8.PubMedCrossRef
169.
go back to reference van Diepen JA, Jansen PA, Ballak DB, Hijmans A, Rutjes FP, Tack CJ, et al. Genetic and pharmacological inhibition of vanin-1 activity in animal models of type 2 diabetes. Sci Rep. 2016;6:21906.PubMedPubMedCentralCrossRef van Diepen JA, Jansen PA, Ballak DB, Hijmans A, Rutjes FP, Tack CJ, et al. Genetic and pharmacological inhibition of vanin-1 activity in animal models of type 2 diabetes. Sci Rep. 2016;6:21906.PubMedPubMedCentralCrossRef
170.
go back to reference Huang H, Dong X, Kang MX, Xu B, Chen Y, Zhang B, et al. Novel blood biomarkers of pancreatic cancer-associated diabetes mellitus identified by peripheral blood-based gene expression profiles. Am J Gastroenterol. 2010;105(7):1661–9.PubMedCrossRef Huang H, Dong X, Kang MX, Xu B, Chen Y, Zhang B, et al. Novel blood biomarkers of pancreatic cancer-associated diabetes mellitus identified by peripheral blood-based gene expression profiles. Am J Gastroenterol. 2010;105(7):1661–9.PubMedCrossRef
171.
go back to reference Roisin-Bouffay C, Castellano R, Valero R, Chasson L, Galland F, Naquet P. Mouse vanin-1 is cytoprotective for islet beta cells and regulates the development of type 1 diabetes. Diabetologia. 2008;51(7):1192–201.PubMedCrossRef Roisin-Bouffay C, Castellano R, Valero R, Chasson L, Galland F, Naquet P. Mouse vanin-1 is cytoprotective for islet beta cells and regulates the development of type 1 diabetes. Diabetologia. 2008;51(7):1192–201.PubMedCrossRef
172.
go back to reference Berruyer C, Martin FM, Castellano R, Macone A, Malergue F, Garrido-Urbani S, et al. Vanin-1-/- mice exhibit a glutathione-mediated tissue resistance to oxidative stress. Mol Cell Biol. 2004;24(16):7214–24.PubMedPubMedCentralCrossRef Berruyer C, Martin FM, Castellano R, Macone A, Malergue F, Garrido-Urbani S, et al. Vanin-1-/- mice exhibit a glutathione-mediated tissue resistance to oxidative stress. Mol Cell Biol. 2004;24(16):7214–24.PubMedPubMedCentralCrossRef
173.
174.
go back to reference Kang M, Qin W, Buya M, Dong X, Zheng W, Lu W, et al. VNN1, a potential biomarker for pancreatic cancer-associated new-onset diabetes, aggravates paraneoplastic islet dysfunction by increasing oxidative stress. Cancer Lett. 2016;373(2):241–50.PubMedCrossRef Kang M, Qin W, Buya M, Dong X, Zheng W, Lu W, et al. VNN1, a potential biomarker for pancreatic cancer-associated new-onset diabetes, aggravates paraneoplastic islet dysfunction by increasing oxidative stress. Cancer Lett. 2016;373(2):241–50.PubMedCrossRef
175.
go back to reference Williams JA. Regulation of normal and adaptive pancreatic growth. Pancreapedia: The Exocrine Pancreas Knowledge Base; 2017. Williams JA. Regulation of normal and adaptive pancreatic growth. Pancreapedia: The Exocrine Pancreas Knowledge Base; 2017.
176.
go back to reference Matsuda A, Makino N, Tozawa T, Shirahata N, Honda T, Ikeda Y, et al. Pancreatic fat accumulation, fibrosis, and acinar cell injury in the Zucker diabetic fatty rat fed a chronic high-fat diet. Pancreas. 2014;43(5):735–43.PubMedPubMedCentralCrossRef Matsuda A, Makino N, Tozawa T, Shirahata N, Honda T, Ikeda Y, et al. Pancreatic fat accumulation, fibrosis, and acinar cell injury in the Zucker diabetic fatty rat fed a chronic high-fat diet. Pancreas. 2014;43(5):735–43.PubMedPubMedCentralCrossRef
177.
go back to reference Dawson DW, Hertzer K, Moro A, Donald G, Chang HH, Go VL, et al. High-fat, high-calorie diet promotes early pancreatic neoplasia in the conditional KrasG12D mouse model. Cancer Prev Res (Phila). 2013;6(10):1064–73.CrossRef Dawson DW, Hertzer K, Moro A, Donald G, Chang HH, Go VL, et al. High-fat, high-calorie diet promotes early pancreatic neoplasia in the conditional KrasG12D mouse model. Cancer Prev Res (Phila). 2013;6(10):1064–73.CrossRef
178.
go back to reference Yang J, Waldron RT, Su HY, Moro A, Chang HH, Eibl G, et al. Insulin promotes proliferation and fibrosing responses in activated pancreatic stellate cells. Am J Physiol Gastrointest Liver Physiol. 2016;311(4):G675–G87.PubMedPubMedCentralCrossRef Yang J, Waldron RT, Su HY, Moro A, Chang HH, Eibl G, et al. Insulin promotes proliferation and fibrosing responses in activated pancreatic stellate cells. Am J Physiol Gastrointest Liver Physiol. 2016;311(4):G675–G87.PubMedPubMedCentralCrossRef
179.
go back to reference Butler AE, Galasso R, Matveyenko A, Rizza RA, Dry S, Butler PC. Pancreatic duct replication is increased with obesity and type 2 diabetes in humans. Diabetologia. 2010;53(1):21–6.PubMedCrossRef Butler AE, Galasso R, Matveyenko A, Rizza RA, Dry S, Butler PC. Pancreatic duct replication is increased with obesity and type 2 diabetes in humans. Diabetologia. 2010;53(1):21–6.PubMedCrossRef
180.
go back to reference Nimmakayala RK, Seshacharyulu P, Lakshmanan I, Rachagani S, Chugh S, Karmakar S, et al. Cigarette smoke induces stem cell features of pancreatic cancer cells via PAF1. Gastroenterology. 2018;155(3):892–908. Nimmakayala RK, Seshacharyulu P, Lakshmanan I, Rachagani S, Chugh S, Karmakar S, et al. Cigarette smoke induces stem cell features of pancreatic cancer cells via PAF1. Gastroenterology. 2018;155(3):892–908.
181.
go back to reference Vaz AP, Deb S, Rachagani S, Dey P, Muniyan S, Lakshmanan I, et al. Overexpression of PD2 leads to increased tumorigenicity and metastasis in pancreatic ductal adenocarcinoma. Oncotarget. 2016;7(3):3317–31.PubMedCrossRef Vaz AP, Deb S, Rachagani S, Dey P, Muniyan S, Lakshmanan I, et al. Overexpression of PD2 leads to increased tumorigenicity and metastasis in pancreatic ductal adenocarcinoma. Oncotarget. 2016;7(3):3317–31.PubMedCrossRef
182.
go back to reference Vaz AP, Ponnusamy MP, Batra SK. Cancer stem cells and therapeutic targets: an emerging field for cancer treatment. Drug Deliv Transl Res. 2013;3(2):113–20.PubMedPubMedCentralCrossRef Vaz AP, Ponnusamy MP, Batra SK. Cancer stem cells and therapeutic targets: an emerging field for cancer treatment. Drug Deliv Transl Res. 2013;3(2):113–20.PubMedPubMedCentralCrossRef
183.
go back to reference Mimeault M, Batra SK. Recent progress on normal and malignant pancreatic stem/progenitor cell research: therapeutic implications for the treatment of type 1 or 2 diabetes mellitus and aggressive pancreatic cancer. Gut. 2008;57(10):1456–68.PubMedCrossRef Mimeault M, Batra SK. Recent progress on normal and malignant pancreatic stem/progenitor cell research: therapeutic implications for the treatment of type 1 or 2 diabetes mellitus and aggressive pancreatic cancer. Gut. 2008;57(10):1456–68.PubMedCrossRef
184.
go back to reference Tchio Mantho CI, Harbuzariu A, Gonzalez-Perez RR. Histone deacetylases, microRNA and leptin crosstalk in pancreatic cancer. World J Clin Oncol. 2017;8(3):178–89.PubMedPubMedCentralCrossRef Tchio Mantho CI, Harbuzariu A, Gonzalez-Perez RR. Histone deacetylases, microRNA and leptin crosstalk in pancreatic cancer. World J Clin Oncol. 2017;8(3):178–89.PubMedPubMedCentralCrossRef
185.
go back to reference Lipsey CC, Harbuzariu A, Daley-Brown D, Gonzalez-Perez RR. Oncogenic role of leptin and Notch interleukin-1 leptin crosstalk outcome in cancer. World J Methodol. 2016;6(1):43–55.PubMedPubMedCentralCrossRef Lipsey CC, Harbuzariu A, Daley-Brown D, Gonzalez-Perez RR. Oncogenic role of leptin and Notch interleukin-1 leptin crosstalk outcome in cancer. World J Methodol. 2016;6(1):43–55.PubMedPubMedCentralCrossRef
186.
go back to reference Rahn S, Zimmermann V, Viol F, Knaack H, Stemmer K, Peters L, et al. Diabetes as risk factor for pancreatic cancer: Hyperglycemia promotes epithelial-mesenchymal-transition and stem cell properties in pancreatic ductal epithelial cells. Cancer Lett. 2018;415:129–50.PubMedCrossRef Rahn S, Zimmermann V, Viol F, Knaack H, Stemmer K, Peters L, et al. Diabetes as risk factor for pancreatic cancer: Hyperglycemia promotes epithelial-mesenchymal-transition and stem cell properties in pancreatic ductal epithelial cells. Cancer Lett. 2018;415:129–50.PubMedCrossRef
Metadata
Title
Pancreatic cancer associated with obesity and diabetes: an alternative approach for its targeting
Authors
Ramesh Pothuraju
Satyanarayana Rachagani
Wade M. Junker
Sanjib Chaudhary
Viswanathan Saraswathi
Sukhwinder Kaur
Surinder K. Batra
Publication date
01-12-2018
Publisher
BioMed Central
Published in
Journal of Experimental & Clinical Cancer Research / Issue 1/2018
Electronic ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-018-0963-4

Other articles of this Issue 1/2018

Journal of Experimental & Clinical Cancer Research 1/2018 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