Skip to main content
Top
Published in: Molecular Cancer 1/2018

Open Access 01-12-2018 | Review

Insulin/IGF-driven cancer cell-stroma crosstalk as a novel therapeutic target in pancreatic cancer

Authors: Ayse Ceren Mutgan, H. Erdinc Besikcioglu, Shenghan Wang, Helmut Friess, Güralp O. Ceyhan, Ihsan Ekin Demir

Published in: Molecular Cancer | Issue 1/2018

Login to get access

Abstract

Pancreatic ductal adenocarcinoma (PDAC) is unrivalled the deadliest gastrointestinal cancer in the western world. There is substantial evidence implying that insulin and insulin-like growth factor (IGF) signaling axis prompt PDAC into an advanced stage by enhancing tumor growth, metastasis and by driving therapy resistance. Numerous efforts have been made to block Insulin/IGF signaling pathway in cancer therapy. However, therapies that target the IGF1 receptor (IGF-1R) and IGF subtypes (IGF-1 and IGF-2) have been repeatedly unsuccessful. This failure may not only be due to the complexity and homology that is shared by Insulin and IGF receptors, but also due to the complex stroma-cancer interactions in the pancreas. Shedding light on the interactions between the endocrine/exocrine pancreas and the stroma in PDAC is likely to steer us toward the development of novel treatments. In this review, we highlight the stroma-derived IGF signaling and IGF-binding proteins as potential novel therapeutic targets in PDAC.
Literature
1.
go back to reference Rahib L, Smith BD, Aizenberg R, Rosenzweig AB, Fleshman JM, Matrisian LM. Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res. 2014;74(11):2913–21.CrossRefPubMed Rahib L, Smith BD, Aizenberg R, Rosenzweig AB, Fleshman JM, Matrisian LM. Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res. 2014;74(11):2913–21.CrossRefPubMed
4.
go back to reference Onitilo AA, Engel JM, Glurich I, Stankowski RV, Williams GM, Doi SA. Diabetes and cancer I: risk, survival, and implications for screening. Cancer Causes Control. 2012;23(6):967–81.CrossRefPubMedPubMedCentral Onitilo AA, Engel JM, Glurich I, Stankowski RV, Williams GM, Doi SA. Diabetes and cancer I: risk, survival, and implications for screening. Cancer Causes Control. 2012;23(6):967–81.CrossRefPubMedPubMedCentral
5.
go back to reference Cotterill AM, Holly JM, Wass JA. The regulation of insulin-like growth factor binding protein (IGFBP)-1 during prolonged fasting. Clin Endocrinol. 1993;39(3):357–62.CrossRef Cotterill AM, Holly JM, Wass JA. The regulation of insulin-like growth factor binding protein (IGFBP)-1 during prolonged fasting. Clin Endocrinol. 1993;39(3):357–62.CrossRef
6.
go back to reference Pollak M. Insulin and insulin-like growth factor signalling in neoplasia. Nat Rev Cancer. 2008;8(12):915–28.CrossRefPubMed Pollak M. Insulin and insulin-like growth factor signalling in neoplasia. Nat Rev Cancer. 2008;8(12):915–28.CrossRefPubMed
7.
go back to reference Bailyes EM, Nave BT, Soos MA, Orr SR, Hayward AC, Siddle K. Insulin receptor/IGF-I receptor hybrids are widely distributed in mammalian tissues: quantification of individual receptor species by selective immunoprecipitation and immunoblotting. Biochem J. 1997;327(Pt 1):209–15.CrossRefPubMedPubMedCentral Bailyes EM, Nave BT, Soos MA, Orr SR, Hayward AC, Siddle K. Insulin receptor/IGF-I receptor hybrids are widely distributed in mammalian tissues: quantification of individual receptor species by selective immunoprecipitation and immunoblotting. Biochem J. 1997;327(Pt 1):209–15.CrossRefPubMedPubMedCentral
8.
go back to reference Federici M, Porzio O, Zucaro L, Fusco A, Borboni P, Lauro D, Sesti G. Distribution of insulin/insulin-like growth factor-I hybrid receptors in human tissues. Mol Cell Endocrinol. 1997;129(2):121–6.CrossRefPubMed Federici M, Porzio O, Zucaro L, Fusco A, Borboni P, Lauro D, Sesti G. Distribution of insulin/insulin-like growth factor-I hybrid receptors in human tissues. Mol Cell Endocrinol. 1997;129(2):121–6.CrossRefPubMed
9.
go back to reference Boucher J, Kleinridders A, Kahn CR. Insulin receptor signaling in normal and insulin-resistant states. Cold Spring Harb Perspect Biol. 2014;6(1). Boucher J, Kleinridders A, Kahn CR. Insulin receptor signaling in normal and insulin-resistant states. Cold Spring Harb Perspect Biol. 2014;6(1).
10.
go back to reference Belfiore A, Frasca F, Pandini G, Sciacca L, Vigneri R. Insulin receptor isoforms and insulin receptor/insulin-like growth factor receptor hybrids in physiology and disease. Endocr Rev. 2009;30(6):586–623.CrossRefPubMed Belfiore A, Frasca F, Pandini G, Sciacca L, Vigneri R. Insulin receptor isoforms and insulin receptor/insulin-like growth factor receptor hybrids in physiology and disease. Endocr Rev. 2009;30(6):586–623.CrossRefPubMed
11.
go back to reference Rajapaksha H, Forbes BE. Ligand-binding affinity at the insulin receptor Isoform-a and subsequent IR-A tyrosine Phosphorylation kinetics are important determinants of Mitogenic biological outcomes. Front Endocrinol (Lausanne). 2015;6:107. Rajapaksha H, Forbes BE. Ligand-binding affinity at the insulin receptor Isoform-a and subsequent IR-A tyrosine Phosphorylation kinetics are important determinants of Mitogenic biological outcomes. Front Endocrinol (Lausanne). 2015;6:107.
12.
go back to reference Siddle K. Signalling by insulin and IGF receptors: supporting acts and new players. J Mol Endocrinol. 2011;47(1):R1–10.CrossRefPubMed Siddle K. Signalling by insulin and IGF receptors: supporting acts and new players. J Mol Endocrinol. 2011;47(1):R1–10.CrossRefPubMed
13.
go back to reference Bergmann U, Funatomi H, Yokoyama M, Beger HG. K M: insulin-like growth factor I Overexpression in human pancreatic cancer: evidence for Autocrine and Paracrine roles. Cancer Res. 1995;55:5. Bergmann U, Funatomi H, Yokoyama M, Beger HG. K M: insulin-like growth factor I Overexpression in human pancreatic cancer: evidence for Autocrine and Paracrine roles. Cancer Res. 1995;55:5.
14.
go back to reference Ohmura E, Okada M, Onoda N, Kamiya Y, Murakami H, Tsushima T, Shizume K. Insulin-like growth factor I and transforming growth factor alpha as autocrine growth factors in human pancreatic cancer cell growth. Cancer Res. 1990;50(1):103–7.PubMed Ohmura E, Okada M, Onoda N, Kamiya Y, Murakami H, Tsushima T, Shizume K. Insulin-like growth factor I and transforming growth factor alpha as autocrine growth factors in human pancreatic cancer cell growth. Cancer Res. 1990;50(1):103–7.PubMed
15.
go back to reference Valsecchi ME, McDonald M, Brody JR, Hyslop T, Freydin B, Yeo CJ, Solomides C, Peiper SC, Witkiewicz AK. Epidermal growth factor receptor and insulinlike growth factor 1 receptor expression predict poor survival in pancreatic ductal adenocarcinoma. Cancer. 2012;118(14):3484–93.CrossRefPubMed Valsecchi ME, McDonald M, Brody JR, Hyslop T, Freydin B, Yeo CJ, Solomides C, Peiper SC, Witkiewicz AK. Epidermal growth factor receptor and insulinlike growth factor 1 receptor expression predict poor survival in pancreatic ductal adenocarcinoma. Cancer. 2012;118(14):3484–93.CrossRefPubMed
16.
go back to reference Ishiwata T, Bergmann U, Kornmann M, Lopez M, Beger HG, Korc M. Altered expression of insulin-like growth factor II receptor in human pancreatic cancer. Pancreas. 1997;15(4):367–73.CrossRefPubMed Ishiwata T, Bergmann U, Kornmann M, Lopez M, Beger HG, Korc M. Altered expression of insulin-like growth factor II receptor in human pancreatic cancer. Pancreas. 1997;15(4):367–73.CrossRefPubMed
17.
18.
go back to reference Ouyang H, Shiwaku HO, Hagiwara H, Miura K, Abe T, Kato Y, Ohtani H, Shiiba K, Souza RF, Meltzer SJ, et al. The insulin-like growth factor II receptor gene is mutated in genetically unstable cancers of the endometrium, stomach, and colorectum. Cancer Res. 1997;57(10):1851–4.PubMed Ouyang H, Shiwaku HO, Hagiwara H, Miura K, Abe T, Kato Y, Ohtani H, Shiiba K, Souza RF, Meltzer SJ, et al. The insulin-like growth factor II receptor gene is mutated in genetically unstable cancers of the endometrium, stomach, and colorectum. Cancer Res. 1997;57(10):1851–4.PubMed
19.
go back to reference Firth SM, Baxter RC. Cellular actions of the insulin-like growth factor binding proteins. Endocr Rev. 2002;23(6):824–54.CrossRefPubMed Firth SM, Baxter RC. Cellular actions of the insulin-like growth factor binding proteins. Endocr Rev. 2002;23(6):824–54.CrossRefPubMed
20.
go back to reference Baxter RC. IGF binding proteins in cancer: mechanistic and clinical insights. Nat Rev Cancer. 2014;14(5):329–41.CrossRefPubMed Baxter RC. IGF binding proteins in cancer: mechanistic and clinical insights. Nat Rev Cancer. 2014;14(5):329–41.CrossRefPubMed
21.
go back to reference Karna E, Surazynski A, Orlowski K, Laszkiewicz J, Puchalski Z, Nawrat P, Palka J. Serum and tissue level of insulin-like growth factor-I (IGF-I) and IGF-I binding proteins as an index of pancreatitis and pancreatic cancer. Int J Exp Pathol. 2002;83(5):239–45.CrossRefPubMedPubMedCentral Karna E, Surazynski A, Orlowski K, Laszkiewicz J, Puchalski Z, Nawrat P, Palka J. Serum and tissue level of insulin-like growth factor-I (IGF-I) and IGF-I binding proteins as an index of pancreatitis and pancreatic cancer. Int J Exp Pathol. 2002;83(5):239–45.CrossRefPubMedPubMedCentral
22.
go back to reference Lin Y, Tamakoshi A, Kikuchi S, Yagyu K, Obata Y, Ishibashi T, Kawamura T, Inaba Y, Kurosawa M, Motohashi Y, et al. Serum insulin-like growth factor-I, insulin-like growth factor binding protein-3, and the risk of pancreatic cancer death. Int J Cancer. 2004;110(4):584–8.CrossRefPubMed Lin Y, Tamakoshi A, Kikuchi S, Yagyu K, Obata Y, Ishibashi T, Kawamura T, Inaba Y, Kurosawa M, Motohashi Y, et al. Serum insulin-like growth factor-I, insulin-like growth factor binding protein-3, and the risk of pancreatic cancer death. Int J Cancer. 2004;110(4):584–8.CrossRefPubMed
23.
go back to reference Rohrmann S, Grote VA, Becker S, Rinaldi S, Tjonneland A, Roswall N, Gronbaek H, Overvad K, Boutron-Ruault MC, Clavel-Chapelon F, et al. Concentrations of IGF-I and IGFBP-3 and pancreatic cancer risk in the European prospective investigation into cancer and nutrition. Br J Cancer. 2012;106(5):1004–10.CrossRefPubMedPubMedCentral Rohrmann S, Grote VA, Becker S, Rinaldi S, Tjonneland A, Roswall N, Gronbaek H, Overvad K, Boutron-Ruault MC, Clavel-Chapelon F, et al. Concentrations of IGF-I and IGFBP-3 and pancreatic cancer risk in the European prospective investigation into cancer and nutrition. Br J Cancer. 2012;106(5):1004–10.CrossRefPubMedPubMedCentral
24.
go back to reference Douglas JB, Silverman DT, Pollak MN, Tao Y, Soliman AS, Stolzenberg-Solomon RZ. Serum IGF-I, IGF-II, IGFBP-3, and IGF-I/IGFBP-3 molar ratio and risk of pancreatic cancer in the prostate, lung, colorectal, and ovarian cancer screening trial. Cancer Epidemiol Biomark Prev. 2010;19(9):2298–306.CrossRef Douglas JB, Silverman DT, Pollak MN, Tao Y, Soliman AS, Stolzenberg-Solomon RZ. Serum IGF-I, IGF-II, IGFBP-3, and IGF-I/IGFBP-3 molar ratio and risk of pancreatic cancer in the prostate, lung, colorectal, and ovarian cancer screening trial. Cancer Epidemiol Biomark Prev. 2010;19(9):2298–306.CrossRef
25.
go back to reference Hirakawa T, Yashiro M, Murata A, Hirata K, Kimura K, Amano R, Yamada N, Nakata B, Hirakawa K. IGF-1 receptor and IGF binding protein-3 might predict prognosis of patients with resectable pancreatic cancer. BMC Cancer. 2013;13:392.CrossRefPubMedPubMedCentral Hirakawa T, Yashiro M, Murata A, Hirata K, Kimura K, Amano R, Yamada N, Nakata B, Hirakawa K. IGF-1 receptor and IGF binding protein-3 might predict prognosis of patients with resectable pancreatic cancer. BMC Cancer. 2013;13:392.CrossRefPubMedPubMedCentral
26.
go back to reference Grimberg A, Liu B, Bannerman P, El-Deiry WS, Cohen P. IGFBP-3 mediates p53-induced apoptosis during serum starvation. Int J Oncol. 2002;21(2):327–35.PubMedPubMedCentral Grimberg A, Liu B, Bannerman P, El-Deiry WS, Cohen P. IGFBP-3 mediates p53-induced apoptosis during serum starvation. Int J Oncol. 2002;21(2):327–35.PubMedPubMedCentral
27.
go back to reference Werner H, Karnieli E, Rauscher FJ, LeRoith D. Wild-type and mutant p53 differentially regulate transcription of the insulin-like growth factor I receptor gene. Proc Natl Acad Sci U S A. 1996;93(16):8318–23.CrossRefPubMedPubMedCentral Werner H, Karnieli E, Rauscher FJ, LeRoith D. Wild-type and mutant p53 differentially regulate transcription of the insulin-like growth factor I receptor gene. Proc Natl Acad Sci U S A. 1996;93(16):8318–23.CrossRefPubMedPubMedCentral
28.
go back to reference Ozdemir BC, Pentcheva-Hoang T, Carstens JL, Zheng X, Wu CC, Simpson TR, Laklai H, Sugimoto H, Kahlert C, Novitskiy SV, et al. Depletion of carcinoma-associated fibroblasts and fibrosis induces immunosuppression and accelerates pancreas cancer with reduced survival. Cancer Cell. 2014;25(6):719–34.CrossRefPubMedPubMedCentral Ozdemir BC, Pentcheva-Hoang T, Carstens JL, Zheng X, Wu CC, Simpson TR, Laklai H, Sugimoto H, Kahlert C, Novitskiy SV, et al. Depletion of carcinoma-associated fibroblasts and fibrosis induces immunosuppression and accelerates pancreas cancer with reduced survival. Cancer Cell. 2014;25(6):719–34.CrossRefPubMedPubMedCentral
29.
go back to reference Rhim AD, Oberstein PE, Thomas DH, Mirek ET, Palermo CF, Sastra SA, Dekleva EN, Saunders T, Becerra CP, Tattersall IW, et al. Stromal elements act to restrain, rather than support, pancreatic ductal adenocarcinoma. Cancer Cell. 2014;25(6):735–47.CrossRefPubMedPubMedCentral Rhim AD, Oberstein PE, Thomas DH, Mirek ET, Palermo CF, Sastra SA, Dekleva EN, Saunders T, Becerra CP, Tattersall IW, et al. Stromal elements act to restrain, rather than support, pancreatic ductal adenocarcinoma. Cancer Cell. 2014;25(6):735–47.CrossRefPubMedPubMedCentral
31.
go back to reference Tape CJ, Ling S, Dimitriadi M, McMahon KM, Worboys JD, Leong HS, Norrie IC, Miller CJ, Poulogiannis G, Lauffenburger DA, et al. Oncogenic KRAS regulates tumor cell signaling via Stromal reciprocation. Cell. 2016;165(4):910–20.CrossRefPubMedPubMedCentral Tape CJ, Ling S, Dimitriadi M, McMahon KM, Worboys JD, Leong HS, Norrie IC, Miller CJ, Poulogiannis G, Lauffenburger DA, et al. Oncogenic KRAS regulates tumor cell signaling via Stromal reciprocation. Cell. 2016;165(4):910–20.CrossRefPubMedPubMedCentral
32.
go back to reference Scales SJ, de Sauvage FJ. Mechanisms of hedgehog pathway activation in cancer and implications for therapy. Trends Pharmacol Sci. 2009;30(6):303–12.CrossRefPubMed Scales SJ, de Sauvage FJ. Mechanisms of hedgehog pathway activation in cancer and implications for therapy. Trends Pharmacol Sci. 2009;30(6):303–12.CrossRefPubMed
33.
go back to reference Rucki AA, Foley K, Zhang P, Xiao Q, Kleponis J, Wu AA, Sharma R, Mo G, Liu A, Van Eyk J, et al. Heterogeneous Stromal signaling within the tumor microenvironment controls the metastasis of pancreatic cancer. Cancer Res. 2017;77(1):41–52.CrossRefPubMed Rucki AA, Foley K, Zhang P, Xiao Q, Kleponis J, Wu AA, Sharma R, Mo G, Liu A, Van Eyk J, et al. Heterogeneous Stromal signaling within the tumor microenvironment controls the metastasis of pancreatic cancer. Cancer Res. 2017;77(1):41–52.CrossRefPubMed
34.
go back to reference Hirakawa T, Yashiro M, Doi Y, Kinoshita H, Morisaki T, Fukuoka T, Hasegawa T, Kimura K, Amano R, Hirakawa K. Pancreatic fibroblasts stimulate the motility of pancreatic cancer cells through IGF1/IGF1R signaling under hypoxia. PLoS One. 2016;11(8):e0159912.CrossRefPubMedPubMedCentral Hirakawa T, Yashiro M, Doi Y, Kinoshita H, Morisaki T, Fukuoka T, Hasegawa T, Kimura K, Amano R, Hirakawa K. Pancreatic fibroblasts stimulate the motility of pancreatic cancer cells through IGF1/IGF1R signaling under hypoxia. PLoS One. 2016;11(8):e0159912.CrossRefPubMedPubMedCentral
35.
go back to reference Rosendahl AH, Gundewar C, Said Hilmersson K, Ni L, Saleem MA, Andersson R. Conditionally immortalized human pancreatic stellate cell lines demonstrate enhanced proliferation and migration in response to IGF-I. Exp Cell Res. 2015;330(2):300–10.CrossRefPubMed Rosendahl AH, Gundewar C, Said Hilmersson K, Ni L, Saleem MA, Andersson R. Conditionally immortalized human pancreatic stellate cell lines demonstrate enhanced proliferation and migration in response to IGF-I. Exp Cell Res. 2015;330(2):300–10.CrossRefPubMed
36.
go back to reference Rajah R, Katz L, Nunn S, Solberg P, Beers T, Cohen P. Insulin-like growth factor binding protein (IGFBP) proteases: functional regulators of cell growth. Prog Growth Factor Res. 1995;6(2–4):273–84.CrossRefPubMed Rajah R, Katz L, Nunn S, Solberg P, Beers T, Cohen P. Insulin-like growth factor binding protein (IGFBP) proteases: functional regulators of cell growth. Prog Growth Factor Res. 1995;6(2–4):273–84.CrossRefPubMed
37.
go back to reference Ceyhan GO, Schafer KH, Kerscher AG, Rauch U, Demir IE, Kadihasanoglu M, Bohm C, Muller MW, Buchler MW, Giese NA, et al. Nerve growth factor and artemin are paracrine mediators of pancreatic neuropathy in pancreatic adenocarcinoma. Ann Surg. 2010;251(5):923–31.CrossRefPubMed Ceyhan GO, Schafer KH, Kerscher AG, Rauch U, Demir IE, Kadihasanoglu M, Bohm C, Muller MW, Buchler MW, Giese NA, et al. Nerve growth factor and artemin are paracrine mediators of pancreatic neuropathy in pancreatic adenocarcinoma. Ann Surg. 2010;251(5):923–31.CrossRefPubMed
38.
go back to reference Rajah R, Bhala A, Nunn SE, Peehl DM, Cohen P. 7S nerve growth factor is an insulin-like growth factor-binding protein protease. Endocrinology. 1996;137(7):2676–82.CrossRefPubMed Rajah R, Bhala A, Nunn SE, Peehl DM, Cohen P. 7S nerve growth factor is an insulin-like growth factor-binding protein protease. Endocrinology. 1996;137(7):2676–82.CrossRefPubMed
39.
go back to reference Braulke T, Claussen M, Saftig P, Wendland M, Neifer K, Schmidt B, Zapf J, von Figura K, Peters C. Proteolysis of IGFBPs by cathepsin D in vitro and in cathepsin D-deficient mice. Prog Growth Factor Res. 1995;6(2–4):265–71.CrossRefPubMed Braulke T, Claussen M, Saftig P, Wendland M, Neifer K, Schmidt B, Zapf J, von Figura K, Peters C. Proteolysis of IGFBPs by cathepsin D in vitro and in cathepsin D-deficient mice. Prog Growth Factor Res. 1995;6(2–4):265–71.CrossRefPubMed
40.
go back to reference Park HD, Kang ES, Kim JW, Lee KT, Lee KH, Park YS, Park JO, Lee J, Heo JS, Choi SH, et al. Serum CA19-9, cathepsin D, and matrix metalloproteinase-7 as a diagnostic panel for pancreatic ductal adenocarcinoma. Proteomics. 2012;12(23–24):3590–7.CrossRefPubMed Park HD, Kang ES, Kim JW, Lee KT, Lee KH, Park YS, Park JO, Lee J, Heo JS, Choi SH, et al. Serum CA19-9, cathepsin D, and matrix metalloproteinase-7 as a diagnostic panel for pancreatic ductal adenocarcinoma. Proteomics. 2012;12(23–24):3590–7.CrossRefPubMed
41.
go back to reference Jakubowska K, Pryczynicz A, Januszewska J, Sidorkiewicz I, Kemona A, Niewinski A, Lewczuk L, Kedra B, Guzinska-Ustymowicz K. Expressions of matrix Metalloproteinases 2, 7, and 9 in carcinogenesis of pancreatic Ductal Adenocarcinoma. Dis Markers. 2016;2016:9895721.CrossRefPubMedPubMedCentral Jakubowska K, Pryczynicz A, Januszewska J, Sidorkiewicz I, Kemona A, Niewinski A, Lewczuk L, Kedra B, Guzinska-Ustymowicz K. Expressions of matrix Metalloproteinases 2, 7, and 9 in carcinogenesis of pancreatic Ductal Adenocarcinoma. Dis Markers. 2016;2016:9895721.CrossRefPubMedPubMedCentral
42.
go back to reference Miyamoto S, Yano K, Sugimoto S, Ishii G, Hasebe T, Endoh Y, Kodama K, Goya M, Chiba T, Ochiai A. Matrix metalloproteinase-7 facilitates insulin-like growth factor bioavailability through its proteinase activity on insulin-like growth factor binding protein 3. Cancer Res. 2004;64(2):665–71.CrossRefPubMed Miyamoto S, Yano K, Sugimoto S, Ishii G, Hasebe T, Endoh Y, Kodama K, Goya M, Chiba T, Ochiai A. Matrix metalloproteinase-7 facilitates insulin-like growth factor bioavailability through its proteinase activity on insulin-like growth factor binding protein 3. Cancer Res. 2004;64(2):665–71.CrossRefPubMed
43.
go back to reference Miyamoto S, Nakamura M, Yano K, Ishii G, Hasebe T, Endoh Y, Sangai T, Maeda H, Shi-Chuang Z, Chiba T, et al. Matrix metalloproteinase-7 triggers the matricrine action of insulin-like growth factor-II via proteinase activity on insulin-like growth factor binding protein 2 in the extracellular matrix. Cancer Sci. 2007;98(5):685–91.CrossRefPubMed Miyamoto S, Nakamura M, Yano K, Ishii G, Hasebe T, Endoh Y, Sangai T, Maeda H, Shi-Chuang Z, Chiba T, et al. Matrix metalloproteinase-7 triggers the matricrine action of insulin-like growth factor-II via proteinase activity on insulin-like growth factor binding protein 2 in the extracellular matrix. Cancer Sci. 2007;98(5):685–91.CrossRefPubMed
44.
go back to reference Prudova A, auf dem Keller U, Butler GS, Overall CM. Multiplex N-terminome analysis of MMP-2 and MMP-9 substrate degradomes by iTRAQ-TAILS quantitative proteomics. Mol Cell Proteomics. 2010;9(5):894–911.CrossRefPubMedPubMedCentral Prudova A, auf dem Keller U, Butler GS, Overall CM. Multiplex N-terminome analysis of MMP-2 and MMP-9 substrate degradomes by iTRAQ-TAILS quantitative proteomics. Mol Cell Proteomics. 2010;9(5):894–911.CrossRefPubMedPubMedCentral
45.
46.
go back to reference Aldinucci D, Colombatti A. The inflammatory chemokine CCL5 and cancer progression. Mediat Inflamm. 2014;2014:292376.CrossRef Aldinucci D, Colombatti A. The inflammatory chemokine CCL5 and cancer progression. Mediat Inflamm. 2014;2014:292376.CrossRef
47.
go back to reference Makinoshima H, Dezawa M. Pancreatic cancer cells activate CCL5 expression in mesenchymal stromal cells through the insulin-like growth factor-I pathway. FEBS Lett. 2009;583(22):3697–703.CrossRefPubMed Makinoshima H, Dezawa M. Pancreatic cancer cells activate CCL5 expression in mesenchymal stromal cells through the insulin-like growth factor-I pathway. FEBS Lett. 2009;583(22):3697–703.CrossRefPubMed
48.
go back to reference Ireland L, Santos A, Ahmed MS, Rainer C, Nielsen SR, Quaranta V, Weyer-Czernilofsky U, Engle DD, Perez-Mancera PA, Coupland SE, et al. Chemoresistance in pancreatic cancer is driven by Stroma-derived insulin-like growth factors. Cancer Res. 2016;76(23):6851–63.CrossRefPubMedPubMedCentral Ireland L, Santos A, Ahmed MS, Rainer C, Nielsen SR, Quaranta V, Weyer-Czernilofsky U, Engle DD, Perez-Mancera PA, Coupland SE, et al. Chemoresistance in pancreatic cancer is driven by Stroma-derived insulin-like growth factors. Cancer Res. 2016;76(23):6851–63.CrossRefPubMedPubMedCentral
49.
go back to reference Yang J, Waldron RT, Su HY, Moro A, Chang HH, Eibl G, Ferreri K, Kandeel FR, Lugea A, Li L, et al. Insulin promotes proliferation and fibrosing responses in activated pancreatic stellate cells. Am J Physiol Gastrointest Liver Physiol. 2016;311(4):G675–87.CrossRefPubMedPubMedCentral Yang J, Waldron RT, Su HY, Moro A, Chang HH, Eibl G, Ferreri K, Kandeel FR, Lugea A, Li L, et al. Insulin promotes proliferation and fibrosing responses in activated pancreatic stellate cells. Am J Physiol Gastrointest Liver Physiol. 2016;311(4):G675–87.CrossRefPubMedPubMedCentral
50.
go back to reference Kikuta K, Masamune A, Hamada S, Takikawa T, Nakano E, Shimosegawa T. Pancreatic stellate cells reduce insulin expression and induce apoptosis in pancreatic beta-cells. Biochem Biophys Res Commun. 2013;433(3):292–7.CrossRefPubMed Kikuta K, Masamune A, Hamada S, Takikawa T, Nakano E, Shimosegawa T. Pancreatic stellate cells reduce insulin expression and induce apoptosis in pancreatic beta-cells. Biochem Biophys Res Commun. 2013;433(3):292–7.CrossRefPubMed
51.
go back to reference Zang G, Sandberg M, Carlsson PO, Welsh N, Jansson L, Barbu A. Activated pancreatic stellate cells can impair pancreatic islet function in mice. Ups J Med Sci. 2015;120(3):169–80.CrossRefPubMedPubMedCentral Zang G, Sandberg M, Carlsson PO, Welsh N, Jansson L, Barbu A. Activated pancreatic stellate cells can impair pancreatic islet function in mice. Ups J Med Sci. 2015;120(3):169–80.CrossRefPubMedPubMedCentral
52.
go back to reference Mossner J, Logsdon CD, Goldfine ID, Williams JA. Do insulin and the insulin like growth factors (IGFs) stimulate growth of the exocrine pancreas? Gut. 1987;28(Suppl):51–5.CrossRefPubMedPubMedCentral Mossner J, Logsdon CD, Goldfine ID, Williams JA. Do insulin and the insulin like growth factors (IGFs) stimulate growth of the exocrine pancreas? Gut. 1987;28(Suppl):51–5.CrossRefPubMedPubMedCentral
53.
go back to reference Ludwig CU, Menke A, Adler G, Lutz MP. Fibroblasts stimulate acinar cell proliferation through IGF-I during regeneration from acute pancreatitis. Am J Phys. 1999;276(1 Pt 1):G193–8. Ludwig CU, Menke A, Adler G, Lutz MP. Fibroblasts stimulate acinar cell proliferation through IGF-I during regeneration from acute pancreatitis. Am J Phys. 1999;276(1 Pt 1):G193–8.
54.
go back to reference Warzecha Z, Dembinski A, Ceranowicz P, Konturek SJ, Tomaszewska R, Stachura J, Konturek PC. IGF-1 stimulates production of interleukin-10 and inhibits development of caerulein-induced pancreatitis. J Physiol Pharmacol. 2003;54(4):575–90.PubMed Warzecha Z, Dembinski A, Ceranowicz P, Konturek SJ, Tomaszewska R, Stachura J, Konturek PC. IGF-1 stimulates production of interleukin-10 and inhibits development of caerulein-induced pancreatitis. J Physiol Pharmacol. 2003;54(4):575–90.PubMed
55.
go back to reference Watanabe H, Saito H, Rychahou PG, Uchida T, Evers BM. Aging is associated with decreased pancreatic acinar cell regeneration and phosphatidylinositol 3-kinase/Akt activation. Gastroenterology. 2005;128(5):1391–404.CrossRefPubMed Watanabe H, Saito H, Rychahou PG, Uchida T, Evers BM. Aging is associated with decreased pancreatic acinar cell regeneration and phosphatidylinositol 3-kinase/Akt activation. Gastroenterology. 2005;128(5):1391–404.CrossRefPubMed
56.
go back to reference Takahashi H, Okamura D, Starr ME, Saito H, Evers BM. Age-dependent reduction of the PI3K regulatory subunit p85alpha suppresses pancreatic acinar cell proliferation. Aging Cell. 2012;11(2):305–14.CrossRefPubMedPubMedCentral Takahashi H, Okamura D, Starr ME, Saito H, Evers BM. Age-dependent reduction of the PI3K regulatory subunit p85alpha suppresses pancreatic acinar cell proliferation. Aging Cell. 2012;11(2):305–14.CrossRefPubMedPubMedCentral
57.
go back to reference Kulkarni RN. Receptors for insulin and insulin-like growth factor-1 and insulin receptor substrate-1 mediate pathways that regulate islet function. Biochem Soc Trans. 2002;30(2):317–22.CrossRefPubMed Kulkarni RN. Receptors for insulin and insulin-like growth factor-1 and insulin receptor substrate-1 mediate pathways that regulate islet function. Biochem Soc Trans. 2002;30(2):317–22.CrossRefPubMed
59.
go back to reference Kulkarni RN. New insights into the roles of insulin/IGF-I in the development and maintenance of beta-cell mass. Rev Endocr Metab Disord. 2005;6(3):199–210.CrossRefPubMed Kulkarni RN. New insights into the roles of insulin/IGF-I in the development and maintenance of beta-cell mass. Rev Endocr Metab Disord. 2005;6(3):199–210.CrossRefPubMed
60.
go back to reference Cordoba-Chacon J, Majumdar N, Pokala NK, Gahete MD, Kineman RD. Islet insulin content and release are increased in male mice with elevated endogenous GH and IGF-I, without evidence of systemic insulin resistance or alterations in beta-cell mass. Growth Hormon IGF Res. 2015;25(4):189–95.CrossRef Cordoba-Chacon J, Majumdar N, Pokala NK, Gahete MD, Kineman RD. Islet insulin content and release are increased in male mice with elevated endogenous GH and IGF-I, without evidence of systemic insulin resistance or alterations in beta-cell mass. Growth Hormon IGF Res. 2015;25(4):189–95.CrossRef
61.
go back to reference Agudo J, Ayuso E, Jimenez V, Salavert A, Casellas A, Tafuro S, Haurigot V, Ruberte J, Segovia JC, Bueren J, et al. IGF-I mediates regeneration of endocrine pancreas by increasing beta cell replication through cell cycle protein modulation in mice. Diabetologia. 2008;51(10):1862–72.CrossRefPubMed Agudo J, Ayuso E, Jimenez V, Salavert A, Casellas A, Tafuro S, Haurigot V, Ruberte J, Segovia JC, Bueren J, et al. IGF-I mediates regeneration of endocrine pancreas by increasing beta cell replication through cell cycle protein modulation in mice. Diabetologia. 2008;51(10):1862–72.CrossRefPubMed
62.
go back to reference Ma F, Wei Z, Shi C, Gan Y, Lu J, Frank SJ, Balducci J, Huang Y. Signaling cross talk between growth hormone (GH) and insulin-like growth factor-I (IGF-I) in pancreatic islet beta-cells. Mol Endocrinol. 2011;25(12):2119–33.CrossRefPubMedPubMedCentral Ma F, Wei Z, Shi C, Gan Y, Lu J, Frank SJ, Balducci J, Huang Y. Signaling cross talk between growth hormone (GH) and insulin-like growth factor-I (IGF-I) in pancreatic islet beta-cells. Mol Endocrinol. 2011;25(12):2119–33.CrossRefPubMedPubMedCentral
63.
go back to reference Escribano O, Gomez-Hernandez A, Diaz-Castroverde S, Nevado C, Garcia G, Otero YF, Perdomo L, Beneit N, Benito M. Insulin receptor isoform a confers a higher proliferative capability to pancreatic beta cells enabling glucose availability and IGF-I signaling. Mol Cell Endocrinol. 2015;409:82–91.CrossRefPubMed Escribano O, Gomez-Hernandez A, Diaz-Castroverde S, Nevado C, Garcia G, Otero YF, Perdomo L, Beneit N, Benito M. Insulin receptor isoform a confers a higher proliferative capability to pancreatic beta cells enabling glucose availability and IGF-I signaling. Mol Cell Endocrinol. 2015;409:82–91.CrossRefPubMed
64.
go back to reference Thankamony A, Capalbo D, Marcovecchio ML, Sleigh A, Jorgensen SW, Hill NR, Mooslehner K, Yeo GS, Bluck L, Juul A, et al. Low circulating levels of IGF-1 in healthy adults are associated with reduced beta-cell function, increased intramyocellular lipid, and enhanced fat utilization during fasting. J Clin Endocrinol Metab. 2014;99(6):2198–207.CrossRefPubMedPubMedCentral Thankamony A, Capalbo D, Marcovecchio ML, Sleigh A, Jorgensen SW, Hill NR, Mooslehner K, Yeo GS, Bluck L, Juul A, et al. Low circulating levels of IGF-1 in healthy adults are associated with reduced beta-cell function, increased intramyocellular lipid, and enhanced fat utilization during fasting. J Clin Endocrinol Metab. 2014;99(6):2198–207.CrossRefPubMedPubMedCentral
65.
go back to reference Casellas A, Mallol C, Salavert A, Jimenez V, Garcia M, Agudo J, Obach M, Haurigot V, Vila L, Molas M, et al. Insulin-like growth factor 2 Overexpression induces Beta-cell dysfunction and increases beta-cell susceptibility to damage. J Biol Chem. 2015;290(27):16772–85.CrossRefPubMedPubMedCentral Casellas A, Mallol C, Salavert A, Jimenez V, Garcia M, Agudo J, Obach M, Haurigot V, Vila L, Molas M, et al. Insulin-like growth factor 2 Overexpression induces Beta-cell dysfunction and increases beta-cell susceptibility to damage. J Biol Chem. 2015;290(27):16772–85.CrossRefPubMedPubMedCentral
66.
go back to reference Shim ML, Levitt Katz LE, Davis J, Dotzler WC, Cohen P, Ferry RJ Jr. Insulin-like growth factor binding protein-3 is a novel mediator of apoptosis in insulin-secreting cells. Growth Hormon IGF Res. 2004;14(3):216–25.CrossRef Shim ML, Levitt Katz LE, Davis J, Dotzler WC, Cohen P, Ferry RJ Jr. Insulin-like growth factor binding protein-3 is a novel mediator of apoptosis in insulin-secreting cells. Growth Hormon IGF Res. 2004;14(3):216–25.CrossRef
67.
go back to reference Nguyen KH, Yao XH, Moulik S, Mishra S, Nyomba BL. Human IGF binding protein-3 overexpression impairs glucose regulation in mice via an inhibition of insulin secretion. Endocrinology. 2011;152(6):2184–96.CrossRefPubMed Nguyen KH, Yao XH, Moulik S, Mishra S, Nyomba BL. Human IGF binding protein-3 overexpression impairs glucose regulation in mice via an inhibition of insulin secretion. Endocrinology. 2011;152(6):2184–96.CrossRefPubMed
68.
go back to reference Trajkovic-Arsic M, Kalideris E, Siveke JT. The role of insulin and IGF system in pancreatic cancer. J Mol Endocrinol. 2013;50(3):R67–74.CrossRefPubMed Trajkovic-Arsic M, Kalideris E, Siveke JT. The role of insulin and IGF system in pancreatic cancer. J Mol Endocrinol. 2013;50(3):R67–74.CrossRefPubMed
69.
go back to reference Barreto SG, Carati CJ, Toouli J, Saccone GT. The islet-acinar axis of the pancreas: more than just insulin. Am J Physiol Gastrointest Liver Physiol. 2010;299(1):G10–22.CrossRefPubMed Barreto SG, Carati CJ, Toouli J, Saccone GT. The islet-acinar axis of the pancreas: more than just insulin. Am J Physiol Gastrointest Liver Physiol. 2010;299(1):G10–22.CrossRefPubMed
70.
go back to reference Lankisch PG, Manthey G, Otto J, Koop H, Talaulicar M, Willms B, Creutzfeldt W. Exocrine pancreatic function in insulin-dependent diabetes mellitus. Digestion. 1982;25(3):211–6.CrossRefPubMed Lankisch PG, Manthey G, Otto J, Koop H, Talaulicar M, Willms B, Creutzfeldt W. Exocrine pancreatic function in insulin-dependent diabetes mellitus. Digestion. 1982;25(3):211–6.CrossRefPubMed
71.
72.
go back to reference Hayden MR, Patel K, Habibi J, Gupta D, Tekwani SS, Whaley-Connell A, Sowers JR. Attenuation of endocrine-exocrine pancreatic communication in type 2 diabetes: pancreatic extracellular matrix ultrastructural abnormalities. J Cardiometab Syndr. 2008;3(4):234–43.CrossRefPubMedPubMedCentral Hayden MR, Patel K, Habibi J, Gupta D, Tekwani SS, Whaley-Connell A, Sowers JR. Attenuation of endocrine-exocrine pancreatic communication in type 2 diabetes: pancreatic extracellular matrix ultrastructural abnormalities. J Cardiometab Syndr. 2008;3(4):234–43.CrossRefPubMedPubMedCentral
73.
go back to reference Gidekel Friedlander SY, Chu GC, Snyder EL, Girnius N, Dibelius G, Crowley D, Vasile E, DePinho RA, Jacks T. Context-dependent transformation of adult pancreatic cells by oncogenic K-Ras. Cancer Cell. 2009;16(5):379–89.CrossRefPubMedPubMedCentral Gidekel Friedlander SY, Chu GC, Snyder EL, Girnius N, Dibelius G, Crowley D, Vasile E, DePinho RA, Jacks T. Context-dependent transformation of adult pancreatic cells by oncogenic K-Ras. Cancer Cell. 2009;16(5):379–89.CrossRefPubMedPubMedCentral
74.
go back to reference Azzarelli R, Hurley C, Sznurkowska MK, Rulands S, Hardwick L, Gamper I, Ali F, McCracken L, Hindley C, McDuff F, et al. Multi-site Neurogenin3 Phosphorylation controls pancreatic endocrine differentiation. Dev Cell. 2017;41(3):274–86. e275CrossRefPubMedPubMedCentral Azzarelli R, Hurley C, Sznurkowska MK, Rulands S, Hardwick L, Gamper I, Ali F, McCracken L, Hindley C, McDuff F, et al. Multi-site Neurogenin3 Phosphorylation controls pancreatic endocrine differentiation. Dev Cell. 2017;41(3):274–86. e275CrossRefPubMedPubMedCentral
75.
go back to reference Lee J, Sugiyama T, Liu Y, Wang J, Gu X, Lei J, Markmann JF, Miyazaki S, Miyazaki J, Szot GL, et al. Expansion and conversion of human pancreatic ductal cells into insulin-secreting endocrine cells. elife. 2013;2:e00940.PubMedPubMedCentral Lee J, Sugiyama T, Liu Y, Wang J, Gu X, Lei J, Markmann JF, Miyazaki S, Miyazaki J, Szot GL, et al. Expansion and conversion of human pancreatic ductal cells into insulin-secreting endocrine cells. elife. 2013;2:e00940.PubMedPubMedCentral
76.
go back to reference Smith SB, Watada H, German MS. Neurogenin3 activates the islet differentiation program while repressing its own expression. Mol Endocrinol. 2004;18(1):142–9.CrossRefPubMed Smith SB, Watada H, German MS. Neurogenin3 activates the islet differentiation program while repressing its own expression. Mol Endocrinol. 2004;18(1):142–9.CrossRefPubMed
77.
go back to reference Valdez IA, Dirice E, Gupta MK, Shirakawa J, Teo AKK, Kulkarni RN. Proinflammatory cytokines induce endocrine differentiation in pancreatic Ductal cells via STAT3-dependent NGN3 activation. Cell Rep. 2016;15(3):460–70.CrossRefPubMedPubMedCentral Valdez IA, Dirice E, Gupta MK, Shirakawa J, Teo AKK, Kulkarni RN. Proinflammatory cytokines induce endocrine differentiation in pancreatic Ductal cells via STAT3-dependent NGN3 activation. Cell Rep. 2016;15(3):460–70.CrossRefPubMedPubMedCentral
78.
go back to reference Carpino G, Renzi A, Cardinale V, Franchitto A, Onori P, Overi D, Rossi M, Berloco PB, Alvaro D, Reid LM, et al. Progenitor cell niches in the human pancreatic duct system and associated pancreatic duct glands: an anatomical and immunophenotyping study. J Anat. 2016;228(3):474–86.CrossRefPubMed Carpino G, Renzi A, Cardinale V, Franchitto A, Onori P, Overi D, Rossi M, Berloco PB, Alvaro D, Reid LM, et al. Progenitor cell niches in the human pancreatic duct system and associated pancreatic duct glands: an anatomical and immunophenotyping study. J Anat. 2016;228(3):474–86.CrossRefPubMed
79.
go back to reference Phillips JM, O'Reilly L, Bland C, Foulis AK, Cooke A. Patients with chronic pancreatitis have islet progenitor cells in their ducts, but reversal of overt diabetes in NOD mice by anti-CD3 shows no evidence for islet regeneration. Diabetes. 2007;56(3):634–40.CrossRefPubMed Phillips JM, O'Reilly L, Bland C, Foulis AK, Cooke A. Patients with chronic pancreatitis have islet progenitor cells in their ducts, but reversal of overt diabetes in NOD mice by anti-CD3 shows no evidence for islet regeneration. Diabetes. 2007;56(3):634–40.CrossRefPubMed
80.
go back to reference Conroy T, Desseigne F, Ychou M, Bouche O, Guimbaud R, Becouarn Y, Adenis A, Raoul JL, Gourgou-Bourgade S, de la Fouchardiere C, et al. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med. 2011;364(19):1817–25.CrossRefPubMed Conroy T, Desseigne F, Ychou M, Bouche O, Guimbaud R, Becouarn Y, Adenis A, Raoul JL, Gourgou-Bourgade S, de la Fouchardiere C, et al. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med. 2011;364(19):1817–25.CrossRefPubMed
81.
go back to reference Tian X, Hao K, Qin C, Xie K, Xie X, Yang Y. Insulin-like growth factor 1 receptor promotes the growth and chemoresistance of pancreatic cancer. Dig Dis Sci. 2013;58(9):2705–12.CrossRefPubMed Tian X, Hao K, Qin C, Xie K, Xie X, Yang Y. Insulin-like growth factor 1 receptor promotes the growth and chemoresistance of pancreatic cancer. Dig Dis Sci. 2013;58(9):2705–12.CrossRefPubMed
82.
go back to reference Fuchs CS, Azevedo S, Okusaka T, Van Laethem JL, Lipton LR, Riess H, Szczylik C, Moore MJ, Peeters M, Bodoky G, et al. A phase 3 randomized, double-blind, placebo-controlled trial of ganitumab or placebo in combination with gemcitabine as first-line therapy for metastatic adenocarcinoma of the pancreas: the GAMMA trial. Ann Oncol. 2015;26(5):921–7.CrossRefPubMedPubMedCentral Fuchs CS, Azevedo S, Okusaka T, Van Laethem JL, Lipton LR, Riess H, Szczylik C, Moore MJ, Peeters M, Bodoky G, et al. A phase 3 randomized, double-blind, placebo-controlled trial of ganitumab or placebo in combination with gemcitabine as first-line therapy for metastatic adenocarcinoma of the pancreas: the GAMMA trial. Ann Oncol. 2015;26(5):921–7.CrossRefPubMedPubMedCentral
83.
go back to reference Philip PA, Benedetti J, Corless CL, Wong R, O’Reilly EM, Flynn PJ, Rowland KM, Atkins JN, Mirtsching BC, Rivkin SE, et al. Phase III study comparing gemcitabine plus cetuximab versus gemcitabine in patients with advanced pancreatic adenocarcinoma: southwest oncology group-directed intergroup trial S0205. J Clin Oncol. 2010;28(22):3605–10.CrossRefPubMedPubMedCentral Philip PA, Benedetti J, Corless CL, Wong R, O’Reilly EM, Flynn PJ, Rowland KM, Atkins JN, Mirtsching BC, Rivkin SE, et al. Phase III study comparing gemcitabine plus cetuximab versus gemcitabine in patients with advanced pancreatic adenocarcinoma: southwest oncology group-directed intergroup trial S0205. J Clin Oncol. 2010;28(22):3605–10.CrossRefPubMedPubMedCentral
84.
go back to reference Fitzgerald JB, Johnson BW, Baum J, Adams S, Iadevaia S, Tang J, Rimkunas V, Xu L, Kohli N, Rennard R, et al. MM-141, an IGF-IR- and ErbB3-directed bispecific antibody, overcomes network adaptations that limit activity of IGF-IR inhibitors. Mol Cancer Ther. 2014;13(2):410–25.CrossRefPubMed Fitzgerald JB, Johnson BW, Baum J, Adams S, Iadevaia S, Tang J, Rimkunas V, Xu L, Kohli N, Rennard R, et al. MM-141, an IGF-IR- and ErbB3-directed bispecific antibody, overcomes network adaptations that limit activity of IGF-IR inhibitors. Mol Cancer Ther. 2014;13(2):410–25.CrossRefPubMed
85.
go back to reference Friedbichler K, Hofmann MH, Kroez M, Ostermann E, Lamche HR, Koessl C, Borges E, Pollak MN, Adolf G, Adam PJ. Pharmacodynamic and Antineoplastic activity of BI 836845, a fully human IGF Ligand-neutralizing antibody, and mechanistic rationale for combination with Rapamycin. Mol Cancer Ther. 2014;13(2):399–409.CrossRefPubMed Friedbichler K, Hofmann MH, Kroez M, Ostermann E, Lamche HR, Koessl C, Borges E, Pollak MN, Adolf G, Adam PJ. Pharmacodynamic and Antineoplastic activity of BI 836845, a fully human IGF Ligand-neutralizing antibody, and mechanistic rationale for combination with Rapamycin. Mol Cancer Ther. 2014;13(2):399–409.CrossRefPubMed
86.
go back to reference Kindler HL, Richards DA, Garbo LE, Garon EB, Stephenson JJ Jr, Rocha-Lima CM, Safran H, Chan D, Kocs DM, Galimi F, et al. A randomized, placebo-controlled phase 2 study of ganitumab (AMG 479) or conatumumab (AMG 655) in combination with gemcitabine in patients with metastatic pancreatic cancer. Ann Oncol. 2012;23(11):2834–42.CrossRefPubMed Kindler HL, Richards DA, Garbo LE, Garon EB, Stephenson JJ Jr, Rocha-Lima CM, Safran H, Chan D, Kocs DM, Galimi F, et al. A randomized, placebo-controlled phase 2 study of ganitumab (AMG 479) or conatumumab (AMG 655) in combination with gemcitabine in patients with metastatic pancreatic cancer. Ann Oncol. 2012;23(11):2834–42.CrossRefPubMed
87.
go back to reference Tabernero J, Chawla SP, Kindler H, Reckamp K, Chiorean EG, Azad NS, Lockhart AC, Hsu CP, Baker NF, Galimi F, et al. Anticancer activity of the type I insulin-like growth factor receptor antagonist, ganitumab, in combination with the death receptor 5 agonist, conatumumab. Target Oncol. 2015;10(1):65–76.CrossRefPubMed Tabernero J, Chawla SP, Kindler H, Reckamp K, Chiorean EG, Azad NS, Lockhart AC, Hsu CP, Baker NF, Galimi F, et al. Anticancer activity of the type I insulin-like growth factor receptor antagonist, ganitumab, in combination with the death receptor 5 agonist, conatumumab. Target Oncol. 2015;10(1):65–76.CrossRefPubMed
88.
go back to reference Philip PA, Goldman B, Ramanathan RK, Lenz HJ, Lowy AM, Whitehead RP, Wakatsuki T, Iqbal S, Gaur R, Benedetti JK, et al. Dual blockade of epidermal growth factor receptor and insulin-like growth factor receptor-1 signaling in metastatic pancreatic cancer: phase Ib and randomized phase II trial of gemcitabine, erlotinib, and cixutumumab versus gemcitabine plus erlotinib (SWOG S0727). Cancer. 2014;120(19):2980–5.CrossRefPubMedPubMedCentral Philip PA, Goldman B, Ramanathan RK, Lenz HJ, Lowy AM, Whitehead RP, Wakatsuki T, Iqbal S, Gaur R, Benedetti JK, et al. Dual blockade of epidermal growth factor receptor and insulin-like growth factor receptor-1 signaling in metastatic pancreatic cancer: phase Ib and randomized phase II trial of gemcitabine, erlotinib, and cixutumumab versus gemcitabine plus erlotinib (SWOG S0727). Cancer. 2014;120(19):2980–5.CrossRefPubMedPubMedCentral
89.
go back to reference Braghiroli MI, de Celis Ferrari AC, Pfiffer TE, Alex AK, Nebuloni D, Carneiro AS, Caparelli F, Senna L, Lobo J, Hoff PM, et al. Phase II trial of metformin and paclitaxel for patients with gemcitabine-refractory advanced adenocarcinoma of the pancreas. Ecancermedicalscience. 2015;9:563.CrossRefPubMedPubMedCentral Braghiroli MI, de Celis Ferrari AC, Pfiffer TE, Alex AK, Nebuloni D, Carneiro AS, Caparelli F, Senna L, Lobo J, Hoff PM, et al. Phase II trial of metformin and paclitaxel for patients with gemcitabine-refractory advanced adenocarcinoma of the pancreas. Ecancermedicalscience. 2015;9:563.CrossRefPubMedPubMedCentral
90.
go back to reference Suleiman Y, Mahipal A, Shibata D, Siegel EM, Jump H, Fulp WJ, Springett GM, Kim R. Phase I study of combination of pasireotide LAR + gemcitabine in locally advanced or metastatic pancreatic cancer. Cancer Chemother Pharmacol. 2015;76(3):481–7.CrossRefPubMed Suleiman Y, Mahipal A, Shibata D, Siegel EM, Jump H, Fulp WJ, Springett GM, Kim R. Phase I study of combination of pasireotide LAR + gemcitabine in locally advanced or metastatic pancreatic cancer. Cancer Chemother Pharmacol. 2015;76(3):481–7.CrossRefPubMed
Metadata
Title
Insulin/IGF-driven cancer cell-stroma crosstalk as a novel therapeutic target in pancreatic cancer
Authors
Ayse Ceren Mutgan
H. Erdinc Besikcioglu
Shenghan Wang
Helmut Friess
Güralp O. Ceyhan
Ihsan Ekin Demir
Publication date
01-12-2018
Publisher
BioMed Central
Published in
Molecular Cancer / Issue 1/2018
Electronic ISSN: 1476-4598
DOI
https://doi.org/10.1186/s12943-018-0806-0

Other articles of this Issue 1/2018

Molecular Cancer 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