Skip to main content
Top
Published in: Cancer and Metastasis Reviews 1-2/2013

01-06-2013

Challenges and advances in mouse modeling for human pancreatic tumorigenesis and metastasis

Authors: Wanglong Qiu, Gloria H. Su

Published in: Cancer and Metastasis Reviews | Issue 1-2/2013

Login to get access

Abstract

Pancreatic cancer is critical for developed countries, where its rate of diagnosis has been increasing steadily annually. In the past decade, the advances of pancreatic cancer research have not contributed to the decline in mortality rates from pancreatic cancer—the overall 5-year survival rate remains about 5% low. This number only underscores an obvious urgency for us to better understand the biological features of pancreatic carcinogenesis, to develop early detection methods, and to improve novel therapeutic treatments. To achieve these goals, animal modeling that faithfully recapitulates the whole process of human pancreatic cancer is central to making the advancements. In this review, we summarize the currently available animal models for pancreatic cancer and the advances in pancreatic cancer animal modeling. We compare and contrast the advantages and disadvantages of three major categories of these models: (1) carcinogen-induced; (2) xenograft and allograft; and (3) genetically engineered mouse models. We focus more on the genetically engineered mouse models, a category which has been rapidly expanded recently for their capacities to mimic human pancreatic cancer and metastasis, and highlight the combinations of these models with various newly developed strategies and cell-lineage labeling systems.
Literature
1.
go back to reference Hruban, R. H., et al. (2001). Pancreatic intraepithelial neoplasia: A new nomenclature and classification system for pancreatic duct lesions. The American Journal of Surgical Pathology, 25(5), 579–586.PubMed Hruban, R. H., et al. (2001). Pancreatic intraepithelial neoplasia: A new nomenclature and classification system for pancreatic duct lesions. The American Journal of Surgical Pathology, 25(5), 579–586.PubMed
2.
go back to reference Takaori, K., et al. (2006). Current topics on precursors to pancreatic cancer. Advances in Medical Sciences, 51, 23–30.PubMed Takaori, K., et al. (2006). Current topics on precursors to pancreatic cancer. Advances in Medical Sciences, 51, 23–30.PubMed
3.
go back to reference Adsay, N. V. (2003). The “new kid on the block”: Intraductal papillary mucinous neoplasms of the pancreas: Current concepts and controversies. Surgery, 133(5), 459–463.PubMed Adsay, N. V. (2003). The “new kid on the block”: Intraductal papillary mucinous neoplasms of the pancreas: Current concepts and controversies. Surgery, 133(5), 459–463.PubMed
4.
go back to reference Hruban, R. H., Wilentz, R. E., & Kern, S. E. (2000). Genetic progression in the pancreatic ducts. American Journal of Pathology, 156(6), 1821–1825.PubMed Hruban, R. H., Wilentz, R. E., & Kern, S. E. (2000). Genetic progression in the pancreatic ducts. American Journal of Pathology, 156(6), 1821–1825.PubMed
5.
go back to reference Wilentz, R. E., Albores-Saavedra, J., & Hruban, R. H. (2000). Mucinous cystic neoplasms of the pancreas. Seminars in Diagnostic Pathology, 17(1), 31–42.PubMed Wilentz, R. E., Albores-Saavedra, J., & Hruban, R. H. (2000). Mucinous cystic neoplasms of the pancreas. Seminars in Diagnostic Pathology, 17(1), 31–42.PubMed
6.
go back to reference Yachida, S., et al. (2010). Distant metastasis occurs late during the genetic evolution of pancreatic cancer. Nature, 467(7319), 1114–1117.PubMed Yachida, S., et al. (2010). Distant metastasis occurs late during the genetic evolution of pancreatic cancer. Nature, 467(7319), 1114–1117.PubMed
7.
go back to reference Almoguera, C., et al. (1988). Most human carcinomas of the exocrine pancreas contain mutant c-K-ras genes. Cell, 53(4), 549–554.PubMed Almoguera, C., et al. (1988). Most human carcinomas of the exocrine pancreas contain mutant c-K-ras genes. Cell, 53(4), 549–554.PubMed
8.
go back to reference Caldas, C., et al. (1994). Frequent somatic mutations and homozygous deletions of the p16 (MTS1) gene in pancreatic adenocarcinoma. Nature Genetics, 8, 27–31.PubMed Caldas, C., et al. (1994). Frequent somatic mutations and homozygous deletions of the p16 (MTS1) gene in pancreatic adenocarcinoma. Nature Genetics, 8, 27–31.PubMed
9.
go back to reference Schutte, M., et al. (1997). Abrogation of the Rb/p16 tumor-suppressive pathway in virtually all pancreatic carcinomas. Cancer Research, 57, 3126–3130.PubMed Schutte, M., et al. (1997). Abrogation of the Rb/p16 tumor-suppressive pathway in virtually all pancreatic carcinomas. Cancer Research, 57, 3126–3130.PubMed
10.
go back to reference Redston, M. S., et al. (1994). p53 mutations in pancreatic carcinoma and evidence of common involvement of homocopolymer tracts in DNA microdeletions. Cancer Research, 54, 3025–3033.PubMed Redston, M. S., et al. (1994). p53 mutations in pancreatic carcinoma and evidence of common involvement of homocopolymer tracts in DNA microdeletions. Cancer Research, 54, 3025–3033.PubMed
11.
go back to reference Pellegata, S., et al. (1994). K-ras and p53 gene mutations in pancreatic cancer: Ductal and nonductal tumors progress through different genetic lesions. Cancer Research, 54, 1556–1560.PubMed Pellegata, S., et al. (1994). K-ras and p53 gene mutations in pancreatic cancer: Ductal and nonductal tumors progress through different genetic lesions. Cancer Research, 54, 1556–1560.PubMed
12.
go back to reference Hahn, S. A., et al. (1996). DPC4, a candidate tumor suppressor gene at human chromosome 18q21.1. Science, 271(5247), 350–353.PubMed Hahn, S. A., et al. (1996). DPC4, a candidate tumor suppressor gene at human chromosome 18q21.1. Science, 271(5247), 350–353.PubMed
13.
go back to reference Wilentz, R. E., et al. (2000). Loss of expression of Dpc4 in pancreatic intraepithelial neoplasia (PanIN): Evidence that DPC4 inactivation occurs late in neoplastic progression. Cancer Research, 60, 2002–2006.PubMed Wilentz, R. E., et al. (2000). Loss of expression of Dpc4 in pancreatic intraepithelial neoplasia (PanIN): Evidence that DPC4 inactivation occurs late in neoplastic progression. Cancer Research, 60, 2002–2006.PubMed
14.
go back to reference Su, G. H., & Kern, S. E. (2000). Molecular genetics of ductal pancreatic neoplasia. Curr Opin Gastroenterology, 16, 419–425. Su, G. H., & Kern, S. E. (2000). Molecular genetics of ductal pancreatic neoplasia. Curr Opin Gastroenterology, 16, 419–425.
15.
go back to reference Klimstra, D. S., & Longnecker, D. S. (1994). K-ras mutations in pancreatic ductal proliferative lesions. American Journal of Pathology, 145(6), 1547–1550.PubMed Klimstra, D. S., & Longnecker, D. S. (1994). K-ras mutations in pancreatic ductal proliferative lesions. American Journal of Pathology, 145(6), 1547–1550.PubMed
16.
go back to reference DiGiuseppe, J. A., et al. (1994). Overexpression of p53 protein in adenocarcinoma of the pancreas. American Journal of Clinical Pathology, 101(6), 684–688.PubMed DiGiuseppe, J. A., et al. (1994). Overexpression of p53 protein in adenocarcinoma of the pancreas. American Journal of Clinical Pathology, 101(6), 684–688.PubMed
17.
go back to reference Sato, N., et al. (2001). STK11/LKB1 Peutz-Jeghers gene inactivation in intraductal papillary–mucinous neoplasms of the pancreas. American Journal of Pathology, 159(6), 2017–2022.PubMed Sato, N., et al. (2001). STK11/LKB1 Peutz-Jeghers gene inactivation in intraductal papillary–mucinous neoplasms of the pancreas. American Journal of Pathology, 159(6), 2017–2022.PubMed
18.
go back to reference Sahin, F., et al. (2003). Loss of Stk11/Lkb1 expression in pancreatic and biliary neoplasms. Modern Pathology, 16(7), 686–691.PubMed Sahin, F., et al. (2003). Loss of Stk11/Lkb1 expression in pancreatic and biliary neoplasms. Modern Pathology, 16(7), 686–691.PubMed
19.
go back to reference Schonleben, F., et al. (2006). PIK3CA mutations in intraductal papillary mucinous neoplasm/carcinoma of the pancreas. Clinical Cancer Research, 12(12), 3851–3855.PubMed Schonleben, F., et al. (2006). PIK3CA mutations in intraductal papillary mucinous neoplasm/carcinoma of the pancreas. Clinical Cancer Research, 12(12), 3851–3855.PubMed
20.
go back to reference Schonleben, F., et al. (2007). BRAF and KRAS gene mutations in intraductal papillary mucinous neoplasm/carcinoma (IPMN/IPMC) of the pancreas. Cancer Letters, 249(2), 242–248.PubMed Schonleben, F., et al. (2007). BRAF and KRAS gene mutations in intraductal papillary mucinous neoplasm/carcinoma (IPMN/IPMC) of the pancreas. Cancer Letters, 249(2), 242–248.PubMed
21.
go back to reference Corbett, T. H., et al. (1984). Induction and chemotherapeutic response of two transplantable ductal adenocarcinomas of the pancreas in C57BL/6 mice. Cancer Research, 44(2), 717–726.PubMed Corbett, T. H., et al. (1984). Induction and chemotherapeutic response of two transplantable ductal adenocarcinomas of the pancreas in C57BL/6 mice. Cancer Research, 44(2), 717–726.PubMed
22.
go back to reference Zimmerman, J. A., et al. (1982). Pancreatic carcinoma induced by N-methyl-N′-nitrosourea in aged mice. Gerontology, 28(2), 114–120.PubMed Zimmerman, J. A., et al. (1982). Pancreatic carcinoma induced by N-methyl-N′-nitrosourea in aged mice. Gerontology, 28(2), 114–120.PubMed
23.
go back to reference Gingell, R., et al. (1976). Metabolism of the pancreatic carcinogens N-nitroso-bis(2-oxopropyl)amine and N-nitroso-bis(2-hydroxypropyl)amine in the Syrian hamster. Journal of the National Cancer Institute, 57(5), 1175–1178.PubMed Gingell, R., et al. (1976). Metabolism of the pancreatic carcinogens N-nitroso-bis(2-oxopropyl)amine and N-nitroso-bis(2-hydroxypropyl)amine in the Syrian hamster. Journal of the National Cancer Institute, 57(5), 1175–1178.PubMed
24.
go back to reference Pour, P., et al. (1976). A further pancreatic carcinogen in Syrian golden hamsters: N-nitroso-bis(2-acetoxypropyl)amine. Cancer Letters, 1(4), 197–202.PubMed Pour, P., et al. (1976). A further pancreatic carcinogen in Syrian golden hamsters: N-nitroso-bis(2-acetoxypropyl)amine. Cancer Letters, 1(4), 197–202.PubMed
25.
go back to reference Pour, P., et al. (1975). Pancreatic neoplasms in an animal model: Morphological, biological, and comparative studies. Cancer, 36(2), 379–389.PubMed Pour, P., et al. (1975). Pancreatic neoplasms in an animal model: Morphological, biological, and comparative studies. Cancer, 36(2), 379–389.PubMed
26.
go back to reference Roebuck, B. D., Baumgartner, K. J., & Thron, C. D. (1984). Characterization of two populations of pancreatic atypical acinar cell foci induced by azaserine in the rat. Laboratory Investigation, 50(2), 141–146.PubMed Roebuck, B. D., Baumgartner, K. J., & Thron, C. D. (1984). Characterization of two populations of pancreatic atypical acinar cell foci induced by azaserine in the rat. Laboratory Investigation, 50(2), 141–146.PubMed
27.
go back to reference Roebuck, B. D., & Longnecker, D. S. (1977). Species and rat strain variation in pancreatic nodule induction by azaserine. Journal of the National Cancer Institute, 59(4), 1273–1277.PubMed Roebuck, B. D., & Longnecker, D. S. (1977). Species and rat strain variation in pancreatic nodule induction by azaserine. Journal of the National Cancer Institute, 59(4), 1273–1277.PubMed
28.
go back to reference Longnecker, D. S., & Curphey, T. J. (1975). Adenocarcinoma of the pancreas in azaserine-treated rats. Cancer Research, 35(8), 2249–2258.PubMed Longnecker, D. S., & Curphey, T. J. (1975). Adenocarcinoma of the pancreas in azaserine-treated rats. Cancer Research, 35(8), 2249–2258.PubMed
29.
go back to reference Osvaldt, A. B., et al. (2006). Pancreatic intraepithelial neoplasia and ductal adenocarcinoma induced by DMBA in mice. Surgery, 140(5), 803–809.PubMed Osvaldt, A. B., et al. (2006). Pancreatic intraepithelial neoplasia and ductal adenocarcinoma induced by DMBA in mice. Surgery, 140(5), 803–809.PubMed
30.
go back to reference Bax, J., et al. (1986). Adenosine triphosphatase, a new marker for the differentiation of putative precancerous foci induced in rat pancreas by azaserine. Carcinogenesis, 7(3), 457–462.PubMed Bax, J., et al. (1986). Adenosine triphosphatase, a new marker for the differentiation of putative precancerous foci induced in rat pancreas by azaserine. Carcinogenesis, 7(3), 457–462.PubMed
31.
go back to reference Roebuck, B. D., Baumgartner, K. J., & Longnecker, D. S. (1987). Growth of pancreatic foci and development of pancreatic cancer with a single dose of azaserine in the rat. Carcinogenesis, 8(12), 1831–1835.PubMed Roebuck, B. D., Baumgartner, K. J., & Longnecker, D. S. (1987). Growth of pancreatic foci and development of pancreatic cancer with a single dose of azaserine in the rat. Carcinogenesis, 8(12), 1831–1835.PubMed
32.
go back to reference Woutersen, R. A., van Garderen-Hoetmer, A., & Longnecker, D. S. (1987). Characterization of a 4-month protocol for the quantitation of BOP-induced lesions in hamster pancreas and its application in studying the effect of dietary fat. Carcinogenesis, 8(6), 833–837.PubMed Woutersen, R. A., van Garderen-Hoetmer, A., & Longnecker, D. S. (1987). Characterization of a 4-month protocol for the quantitation of BOP-induced lesions in hamster pancreas and its application in studying the effect of dietary fat. Carcinogenesis, 8(6), 833–837.PubMed
33.
go back to reference Mizumoto, K., et al. (1989). Cycles of repeated augmentation pressure in rapid production of pancreatic and cholangiocellular carcinomas in hamsters initiated with N-nitrosobis(2-oxopropyl)amine. Carcinogenesis, 10(8), 1457–1459.PubMed Mizumoto, K., et al. (1989). Cycles of repeated augmentation pressure in rapid production of pancreatic and cholangiocellular carcinomas in hamsters initiated with N-nitrosobis(2-oxopropyl)amine. Carcinogenesis, 10(8), 1457–1459.PubMed
34.
go back to reference Meijers, M., et al. (1989). Histogenesis of early preneoplastic lesions induced by N-nitrosobis(2-oxopropyl)amine in exocrine pancreas of hamsters. International Journal of Pancreatology, 4(2), 127–137.PubMed Meijers, M., et al. (1989). Histogenesis of early preneoplastic lesions induced by N-nitrosobis(2-oxopropyl)amine in exocrine pancreas of hamsters. International Journal of Pancreatology, 4(2), 127–137.PubMed
35.
go back to reference Kilian, M., et al. (2006). Matrix metalloproteinase inhibitor RO 28–2653 decreases liver metastasis by reduction of MMP-2 and MMP-9 concentration in BOP-induced ductal pancreatic cancer in Syrian hamsters: Inhibition of matrix metalloproteinases in pancreatic cancer. Prostaglandins, Leukotrienes, and Essential Fatty Acids, 75(6), 429–434.PubMed Kilian, M., et al. (2006). Matrix metalloproteinase inhibitor RO 28–2653 decreases liver metastasis by reduction of MMP-2 and MMP-9 concentration in BOP-induced ductal pancreatic cancer in Syrian hamsters: Inhibition of matrix metalloproteinases in pancreatic cancer. Prostaglandins, Leukotrienes, and Essential Fatty Acids, 75(6), 429–434.PubMed
36.
go back to reference Kubozoe, T., et al. (2001). Absence of beta-catenin gene mutations in pancreatic duct lesions induced by N-nitrosobis(2-oxopropyl)amine in hamsters. Cancer Letters, 168(1), 1–6.PubMed Kubozoe, T., et al. (2001). Absence of beta-catenin gene mutations in pancreatic duct lesions induced by N-nitrosobis(2-oxopropyl)amine in hamsters. Cancer Letters, 168(1), 1–6.PubMed
37.
go back to reference Kokkinakis, D. M., & Scarpelli, D. G. (1989). DNA alkylation in the hamster induced by two pancreatic carcinogens. Cancer Research, 49(12), 3184–3189.PubMed Kokkinakis, D. M., & Scarpelli, D. G. (1989). DNA alkylation in the hamster induced by two pancreatic carcinogens. Cancer Research, 49(12), 3184–3189.PubMed
38.
go back to reference Mangino, M. M., Scarpelli, D. G., & Kokkinakis, D. M. (1990). Metabolism and activation of the pancreatic carcinogen N-nitrosobis(2-oxopropyl)amine by isolated hepatocytes and pancreatic cells of the Syrian hamster. Carcinogenesis, 11(4), 625–631.PubMed Mangino, M. M., Scarpelli, D. G., & Kokkinakis, D. M. (1990). Metabolism and activation of the pancreatic carcinogen N-nitrosobis(2-oxopropyl)amine by isolated hepatocytes and pancreatic cells of the Syrian hamster. Carcinogenesis, 11(4), 625–631.PubMed
39.
go back to reference Sugio, K., et al. (1996). High yields of K-ras mutations in intraductal papillary mucinous tumors and invasive adenocarcinomas induced by N-nitroso(2-hydroxypropyl)(2-oxopropyl)amine in the pancreas of female Syrian hamsters. Carcinogenesis, 17(2), 303–309.PubMed Sugio, K., et al. (1996). High yields of K-ras mutations in intraductal papillary mucinous tumors and invasive adenocarcinomas induced by N-nitroso(2-hydroxypropyl)(2-oxopropyl)amine in the pancreas of female Syrian hamsters. Carcinogenesis, 17(2), 303–309.PubMed
40.
go back to reference Longnecker, D. S., et al. (1985). Induction of pancreatic carcinomas in rats with N-nitroso(2-hydroxypropyl)(2-oxopropyl)amine: Histopathology. Journal of the National Cancer Institute, 74(1), 209–217.PubMed Longnecker, D. S., et al. (1985). Induction of pancreatic carcinomas in rats with N-nitroso(2-hydroxypropyl)(2-oxopropyl)amine: Histopathology. Journal of the National Cancer Institute, 74(1), 209–217.PubMed
41.
go back to reference Bockman, D. E., et al. (1978). Origin of tubular complexes developing during induction of pancreatic adenocarcinoma by 7,12-dimethylbenz(a)anthracene. American Journal of Pathology, 90(3), 645–658.PubMed Bockman, D. E., et al. (1978). Origin of tubular complexes developing during induction of pancreatic adenocarcinoma by 7,12-dimethylbenz(a)anthracene. American Journal of Pathology, 90(3), 645–658.PubMed
42.
go back to reference Rivera, J. A., et al. (1997). A rat model of pancreatic ductal adenocarcinoma: Targeting chemical carcinogens. Surgery, 122(1), 82–90.PubMed Rivera, J. A., et al. (1997). A rat model of pancreatic ductal adenocarcinoma: Targeting chemical carcinogens. Surgery, 122(1), 82–90.PubMed
43.
go back to reference Z’Graggen, K., et al. (2001). Promoting effect of a high-fat/high-protein diet in DMBA-induced ductal pancreatic cancer in rats. Annals of Surgery, 233(5), 688–695.PubMed Z’Graggen, K., et al. (2001). Promoting effect of a high-fat/high-protein diet in DMBA-induced ductal pancreatic cancer in rats. Annals of Surgery, 233(5), 688–695.PubMed
44.
go back to reference Wendt, L. R., et al. (2007). Pancreatic intraepithelial neoplasia and ductal adenocarcinoma induced by DMBA in mice: Effects of alcohol and caffeine. Acta Cirúrgica Brasileira, 22(3), 202–209.PubMed Wendt, L. R., et al. (2007). Pancreatic intraepithelial neoplasia and ductal adenocarcinoma induced by DMBA in mice: Effects of alcohol and caffeine. Acta Cirúrgica Brasileira, 22(3), 202–209.PubMed
45.
go back to reference Kimura, K., et al. (2007). Activation of Notch signaling in tumorigenesis of experimental pancreatic cancer induced by dimethylbenzanthracene in mice. Cancer Science, 98(2), 155–162.PubMed Kimura, K., et al. (2007). Activation of Notch signaling in tumorigenesis of experimental pancreatic cancer induced by dimethylbenzanthracene in mice. Cancer Science, 98(2), 155–162.PubMed
46.
go back to reference Pour, P. M., & Rivenson, A. (1989). Induction of a mixed ductal-squamous-islet cell carcinoma in a rat treated with a tobacco-specific carcinogen. American Journal of Pathology, 134(3), 627–631.PubMed Pour, P. M., & Rivenson, A. (1989). Induction of a mixed ductal-squamous-islet cell carcinoma in a rat treated with a tobacco-specific carcinogen. American Journal of Pathology, 134(3), 627–631.PubMed
47.
go back to reference Rivenson, A., et al. (1988). Induction of lung and exocrine pancreas tumors in F344 rats by tobacco-specific and Areca-derived N-nitrosamines. Cancer Research, 48(23), 6912–6917.PubMed Rivenson, A., et al. (1988). Induction of lung and exocrine pancreas tumors in F344 rats by tobacco-specific and Areca-derived N-nitrosamines. Cancer Research, 48(23), 6912–6917.PubMed
48.
go back to reference Schuller, H. M., et al. (1993). Transplacental induction of pancreas tumors in hamsters by ethanol and the tobacco-specific nitrosamine 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone. Cancer Research, 53(11), 2498–2501.PubMed Schuller, H. M., et al. (1993). Transplacental induction of pancreas tumors in hamsters by ethanol and the tobacco-specific nitrosamine 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone. Cancer Research, 53(11), 2498–2501.PubMed
49.
go back to reference Van Benthem, J., et al. (1994). Immunocytochemical identification of DNA adducts, O6-methylguanine and 7-methylguanine, in respiratory and other tissues of rat, mouse and Syrian hamster exposed to 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone. Carcinogenesis, 15(9), 2023–2029.PubMed Van Benthem, J., et al. (1994). Immunocytochemical identification of DNA adducts, O6-methylguanine and 7-methylguanine, in respiratory and other tissues of rat, mouse and Syrian hamster exposed to 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone. Carcinogenesis, 15(9), 2023–2029.PubMed
50.
go back to reference Hayashi, Y., et al. (1987). Induction of cells with acinar cell phenotype including presence of intracellular amylase. Treatment with 12-O-tetradecanoyl-phorbol-13-acetate in a neoplastic human salivary intercalated duct cell line grown in athymic nude mice. Cancer, 60(5), 1000–1008.PubMed Hayashi, Y., et al. (1987). Induction of cells with acinar cell phenotype including presence of intracellular amylase. Treatment with 12-O-tetradecanoyl-phorbol-13-acetate in a neoplastic human salivary intercalated duct cell line grown in athymic nude mice. Cancer, 60(5), 1000–1008.PubMed
51.
go back to reference Reddy, J. K., & Rao, M. S. (1975). Pancreatic adenocarcinoma in inbred guinea pigs induced by n-methyl-N-nitrosourea. Cancer Research, 35(8), 2269–2277.PubMed Reddy, J. K., & Rao, M. S. (1975). Pancreatic adenocarcinoma in inbred guinea pigs induced by n-methyl-N-nitrosourea. Cancer Research, 35(8), 2269–2277.PubMed
52.
go back to reference Rao, M. S., & Reddy, J. K. (1980). Histogenesis of pseudo-ductular changes induced in the pancreas of guinea pigs treated with N-methyl-N-nitrosourea. Carcinogenesis, 1(12), 1027–1037.PubMed Rao, M. S., & Reddy, J. K. (1980). Histogenesis of pseudo-ductular changes induced in the pancreas of guinea pigs treated with N-methyl-N-nitrosourea. Carcinogenesis, 1(12), 1027–1037.PubMed
53.
go back to reference Chang, K. W., et al. (1994). Genomic p53 mutation in a chemically induced hamster pancreatic ductal adenocarcinoma. Cancer Research, 54(14), 3878–3883.PubMed Chang, K. W., et al. (1994). Genomic p53 mutation in a chemically induced hamster pancreatic ductal adenocarcinoma. Cancer Research, 54(14), 3878–3883.PubMed
54.
go back to reference Al-Wadei, H. A., & Schuller, H. M. (2009). Nicotinic receptor-associated modulation of stimulatory and inhibitory neurotransmitters in NNK-induced adenocarcinoma of the lungs and pancreas. The Journal of Pathology, 218(4), 437–445.PubMed Al-Wadei, H. A., & Schuller, H. M. (2009). Nicotinic receptor-associated modulation of stimulatory and inhibitory neurotransmitters in NNK-induced adenocarcinoma of the lungs and pancreas. The Journal of Pathology, 218(4), 437–445.PubMed
55.
go back to reference Longnecker, D. S. (1991). Hormones and pancreatic cancer. International Journal of Pancreatology, 9, 81–86.PubMed Longnecker, D. S. (1991). Hormones and pancreatic cancer. International Journal of Pancreatology, 9, 81–86.PubMed
56.
go back to reference Lhoste, E. F., & Longnecker, D. S. (1987). Effect of bombesin and caerulein on early stages of carcinogenesis induced by azaserine in the rat pancreas. Cancer Research, 47(12), 3273–3277.PubMed Lhoste, E. F., & Longnecker, D. S. (1987). Effect of bombesin and caerulein on early stages of carcinogenesis induced by azaserine in the rat pancreas. Cancer Research, 47(12), 3273–3277.PubMed
57.
go back to reference McGuinness, E. E., Morgan, R. G., & Wormsley, K. G. (1987). Fate of pancreatic nodules induced by raw soya flour in rats. Gut, 28(Suppl), 207–212.PubMed McGuinness, E. E., Morgan, R. G., & Wormsley, K. G. (1987). Fate of pancreatic nodules induced by raw soya flour in rats. Gut, 28(Suppl), 207–212.PubMed
58.
go back to reference Al-Wadei, H. A., Al-Wadei, M. H., & Schuller, H. M. (2009). Prevention of pancreatic cancer by the beta-blocker propranolol. Anti-Cancer Drugs, 20(6), 477–482.PubMed Al-Wadei, H. A., Al-Wadei, M. H., & Schuller, H. M. (2009). Prevention of pancreatic cancer by the beta-blocker propranolol. Anti-Cancer Drugs, 20(6), 477–482.PubMed
59.
go back to reference Valentich, M. A., et al. (2006). Effect of the co-administration of phenobarbital, quercetin and mancozeb on nitrosomethylurea-induced pancreatic tumors in rats. Food and Chemical Toxicology, 44(12), 2101–2105.PubMed Valentich, M. A., et al. (2006). Effect of the co-administration of phenobarbital, quercetin and mancozeb on nitrosomethylurea-induced pancreatic tumors in rats. Food and Chemical Toxicology, 44(12), 2101–2105.PubMed
60.
go back to reference Ouyang, N., et al. (2006). Nitric oxide-donating aspirin prevents pancreatic cancer in a hamster tumor model. Cancer Research, 66(8), 4503–4511.PubMed Ouyang, N., et al. (2006). Nitric oxide-donating aspirin prevents pancreatic cancer in a hamster tumor model. Cancer Research, 66(8), 4503–4511.PubMed
61.
go back to reference Kuroiwa, Y., et al. (2006). Protective effects of benzyl isothiocyanate and sulforaphane but not resveratrol against initiation of pancreatic carcinogenesis in hamsters. Cancer Letters, 241(2), 275–280.PubMed Kuroiwa, Y., et al. (2006). Protective effects of benzyl isothiocyanate and sulforaphane but not resveratrol against initiation of pancreatic carcinogenesis in hamsters. Cancer Letters, 241(2), 275–280.PubMed
62.
go back to reference Heukamp, I., et al. (2005). Effects of the antioxidative vitamins A, C and E on liver metastasis and intrametastatic lipid peroxidation in BOP-induced pancreatic cancer in Syrian hamsters. Pancreatology, 5(4–5), 403–409.PubMed Heukamp, I., et al. (2005). Effects of the antioxidative vitamins A, C and E on liver metastasis and intrametastatic lipid peroxidation in BOP-induced pancreatic cancer in Syrian hamsters. Pancreatology, 5(4–5), 403–409.PubMed
63.
go back to reference Schuller, H. M., et al. (2002). The cyclooxygenase inhibitor ibuprofen and the FLAP inhibitor MK886 inhibit pancreatic carcinogenesis induced in hamsters by transplacental exposure to ethanol and the tobacco carcinogen NNK. Journal of Cancer Research and Clinical Oncology, 128(10), 525–532.PubMed Schuller, H. M., et al. (2002). The cyclooxygenase inhibitor ibuprofen and the FLAP inhibitor MK886 inhibit pancreatic carcinogenesis induced in hamsters by transplacental exposure to ethanol and the tobacco carcinogen NNK. Journal of Cancer Research and Clinical Oncology, 128(10), 525–532.PubMed
64.
go back to reference Strouch, M. J., et al. (2011). A high omega-3 fatty acid diet mitigates murine pancreatic precancer development. Journal of Surgical Research, 165(1), 75–81.PubMed Strouch, M. J., et al. (2011). A high omega-3 fatty acid diet mitigates murine pancreatic precancer development. Journal of Surgical Research, 165(1), 75–81.PubMed
65.
go back to reference Gray, M. J., et al. (2005). Neuropilin-1 suppresses tumorigenic properties in a human pancreatic adenocarcinoma cell line lacking neuropilin-1 coreceptors. Cancer Research, 65(9), 3664–3670.PubMed Gray, M. J., et al. (2005). Neuropilin-1 suppresses tumorigenic properties in a human pancreatic adenocarcinoma cell line lacking neuropilin-1 coreceptors. Cancer Research, 65(9), 3664–3670.PubMed
66.
go back to reference Bosma, M. J., & Carroll, A. M. (1991). The SCID mouse mutant: Definition, characterization, and potential uses. Annual Review of Immunology, 9, 323–350.PubMed Bosma, M. J., & Carroll, A. M. (1991). The SCID mouse mutant: Definition, characterization, and potential uses. Annual Review of Immunology, 9, 323–350.PubMed
67.
go back to reference Ito, M., et al. (2002). NOD/SCID/gamma(c)(null) mouse: An excellent recipient mouse model for engraftment of human cells. Blood, 100(9), 3175–3182.PubMed Ito, M., et al. (2002). NOD/SCID/gamma(c)(null) mouse: An excellent recipient mouse model for engraftment of human cells. Blood, 100(9), 3175–3182.PubMed
68.
go back to reference Quintana, E., et al. (2008). Efficient tumour formation by single human melanoma cells. Nature, 456(7222), 593–598.PubMed Quintana, E., et al. (2008). Efficient tumour formation by single human melanoma cells. Nature, 456(7222), 593–598.PubMed
69.
go back to reference Hafeez, B.B., et al., Plumbagin, a plant derived natural agent inhibits the growth of pancreatic cancer cells in in vitro and in vivo via targeting EGFR, Stat3 and NF-kappaB signaling pathways. Int J Cancer, 2012; 131:2175-2186. Hafeez, B.B., et al., Plumbagin, a plant derived natural agent inhibits the growth of pancreatic cancer cells in in vitro and in vivo via targeting EGFR, Stat3 and NF-kappaB signaling pathways. Int J Cancer, 2012; 131:2175-2186.
70.
go back to reference Girgis, M. D., et al. (2011). Targeting CEA in pancreas cancer xenografts with a mutated scFv-Fc antibody fragment. EJNMMI Research, 1(1), 24.PubMed Girgis, M. D., et al. (2011). Targeting CEA in pancreas cancer xenografts with a mutated scFv-Fc antibody fragment. EJNMMI Research, 1(1), 24.PubMed
71.
go back to reference Aurisicchio, L., et al., Novel anti-ErbB3 monoclonal antibodies show therapeutic efficacy in xenografted and spontaneous mouse tumors. J Cell Physiol, 2012;227:3381-3388. Aurisicchio, L., et al., Novel anti-ErbB3 monoclonal antibodies show therapeutic efficacy in xenografted and spontaneous mouse tumors. J Cell Physiol, 2012;227:3381-3388.
72.
go back to reference Nakamura, M., et al., Targeting the hedgehog signaling pathway with interacting peptides to Patched-1. J Gastroenterol, 2012; 47:452-460. Nakamura, M., et al., Targeting the hedgehog signaling pathway with interacting peptides to Patched-1. J Gastroenterol, 2012; 47:452-460.
73.
go back to reference Li, H., et al. (2011). STAT3 knockdown reduces pancreatic cancer cell invasiveness and matrix metalloproteinase-7 expression in nude mice. PLoS One, 6(10), e25941.PubMed Li, H., et al. (2011). STAT3 knockdown reduces pancreatic cancer cell invasiveness and matrix metalloproteinase-7 expression in nude mice. PLoS One, 6(10), e25941.PubMed
74.
go back to reference Fidler, I. J. (1986). Rationale and methods for the use of nude mice to study the biology and therapy of human cancer metastasis. Cancer Metastasis Reviews, 5(1), 29–49.PubMed Fidler, I. J. (1986). Rationale and methods for the use of nude mice to study the biology and therapy of human cancer metastasis. Cancer Metastasis Reviews, 5(1), 29–49.PubMed
75.
go back to reference DeRosier, L. C., et al. (2006). Treatment with gemcitabine and TRA-8 anti-death receptor-5 mAb reduces pancreatic adenocarcinoma cell viability in vitro and growth in vivo. Journal of Gastrointestinal Surgery, 10(9), 1291–1300. discussion 1300.PubMed DeRosier, L. C., et al. (2006). Treatment with gemcitabine and TRA-8 anti-death receptor-5 mAb reduces pancreatic adenocarcinoma cell viability in vitro and growth in vivo. Journal of Gastrointestinal Surgery, 10(9), 1291–1300. discussion 1300.PubMed
76.
go back to reference Capella, G., et al. (1999). Orthotopic models of human pancreatic cancer. Annals of the New York Academy of Sciences, 880, 103–109.PubMed Capella, G., et al. (1999). Orthotopic models of human pancreatic cancer. Annals of the New York Academy of Sciences, 880, 103–109.PubMed
77.
go back to reference Rubio-Viqueira, B., et al. (2006). An in vivo platform for translational drug development in pancreatic cancer. Clinical Cancer Research, 12(15), 4652–4661.PubMed Rubio-Viqueira, B., et al. (2006). An in vivo platform for translational drug development in pancreatic cancer. Clinical Cancer Research, 12(15), 4652–4661.PubMed
78.
go back to reference Trevino, J. G., et al. (2006). Inhibition of SRC expression and activity inhibits tumor progression and metastasis of human pancreatic adenocarcinoma cells in an orthotopic nude mouse model. American Journal of Pathology, 168(3), 962–972.PubMed Trevino, J. G., et al. (2006). Inhibition of SRC expression and activity inhibits tumor progression and metastasis of human pancreatic adenocarcinoma cells in an orthotopic nude mouse model. American Journal of Pathology, 168(3), 962–972.PubMed
79.
go back to reference Li, C., et al. (2007). Identification of pancreatic cancer stem cells. Cancer Research, 67(3), 1030–1037.PubMed Li, C., et al. (2007). Identification of pancreatic cancer stem cells. Cancer Research, 67(3), 1030–1037.PubMed
80.
go back to reference Derosier, L. C., et al. (2007). TRA-8 anti-DR5 monoclonal antibody and gemcitabine induce apoptosis and inhibit radiologically validated orthotopic pancreatic tumor growth. Molecular Cancer Therapeutics, 6(12 Pt 1), 3198–3207.PubMed Derosier, L. C., et al. (2007). TRA-8 anti-DR5 monoclonal antibody and gemcitabine induce apoptosis and inhibit radiologically validated orthotopic pancreatic tumor growth. Molecular Cancer Therapeutics, 6(12 Pt 1), 3198–3207.PubMed
81.
go back to reference Jimeno, A., et al. (2009). A direct pancreatic cancer xenograft model as a platform for cancer stem cell therapeutic development. Molecular Cancer Therapeutics, 8(2), 310–314.PubMed Jimeno, A., et al. (2009). A direct pancreatic cancer xenograft model as a platform for cancer stem cell therapeutic development. Molecular Cancer Therapeutics, 8(2), 310–314.PubMed
82.
go back to reference Fogh, J., et al. (1980). Thirty-four lines of six human tumor categories established in nude mice. Journal of the National Cancer Institute, 64(4), 745–751.PubMed Fogh, J., et al. (1980). Thirty-four lines of six human tumor categories established in nude mice. Journal of the National Cancer Institute, 64(4), 745–751.PubMed
83.
go back to reference Alisauskus, R., Wong, G. Y., & Gold, D. V. (1995). Initial studies of monoclonal antibody PAM4 targeting to xenografted orthotopic pancreatic cancer. Cancer Research, 55(23 Suppl), 5743s–5748s.PubMed Alisauskus, R., Wong, G. Y., & Gold, D. V. (1995). Initial studies of monoclonal antibody PAM4 targeting to xenografted orthotopic pancreatic cancer. Cancer Research, 55(23 Suppl), 5743s–5748s.PubMed
84.
go back to reference Killion, J. J., Radinsky, R., & Fidler, I. J. (1998). Orthotopic models are necessary to predict therapy of transplantable tumors in mice. Cancer Metastasis Reviews, 17(3), 279–284.PubMed Killion, J. J., Radinsky, R., & Fidler, I. J. (1998). Orthotopic models are necessary to predict therapy of transplantable tumors in mice. Cancer Metastasis Reviews, 17(3), 279–284.PubMed
85.
go back to reference Loukopoulos, P., et al. (2004). Orthotopic transplantation models of pancreatic adenocarcinoma derived from cell lines and primary tumors and displaying varying metastatic activity. Pancreas, 29(3), 193–203.PubMed Loukopoulos, P., et al. (2004). Orthotopic transplantation models of pancreatic adenocarcinoma derived from cell lines and primary tumors and displaying varying metastatic activity. Pancreas, 29(3), 193–203.PubMed
86.
go back to reference Huynh, A. S., et al. (2011). Development of an orthotopic human pancreatic cancer xenograft model using ultrasound guided injection of cells. PLoS One, 6(5), e20330.PubMed Huynh, A. S., et al. (2011). Development of an orthotopic human pancreatic cancer xenograft model using ultrasound guided injection of cells. PLoS One, 6(5), e20330.PubMed
87.
go back to reference DeRosier, L. C., et al. (2007). Combination treatment with TRA-8 anti death receptor 5 antibody and CPT-11 induces tumor regression in an orthotopic model of pancreatic cancer. Clinical Cancer Research, 13(18 Pt 2), 5535s–5543s.PubMed DeRosier, L. C., et al. (2007). Combination treatment with TRA-8 anti death receptor 5 antibody and CPT-11 induces tumor regression in an orthotopic model of pancreatic cancer. Clinical Cancer Research, 13(18 Pt 2), 5535s–5543s.PubMed
88.
go back to reference Partecke, I. L., et al. (2011). In vivo imaging of pancreatic tumours and liver metastases using 7 Tesla MRI in a murine orthotopic pancreatic cancer model and a liver metastases model. BMC Cancer, 11, 40.PubMed Partecke, I. L., et al. (2011). In vivo imaging of pancreatic tumours and liver metastases using 7 Tesla MRI in a murine orthotopic pancreatic cancer model and a liver metastases model. BMC Cancer, 11, 40.PubMed
89.
go back to reference Cheng, L., et al. (1996). Use of green fluorescent protein variants to monitor gene transfer and expression in mammalian cells. Nature Biotechnology, 14(5), 606–609.PubMed Cheng, L., et al. (1996). Use of green fluorescent protein variants to monitor gene transfer and expression in mammalian cells. Nature Biotechnology, 14(5), 606–609.PubMed
90.
go back to reference Chalfie, M., et al. (1994). Green fluorescent protein as a marker for gene expression. Science, 263(5148), 802–805.PubMed Chalfie, M., et al. (1994). Green fluorescent protein as a marker for gene expression. Science, 263(5148), 802–805.PubMed
91.
go back to reference Astoul, P., et al. (1994). A “patient-like” nude mouse model of parietal pleural human lung adenocarcinoma. Anticancer Research, 14(1A), 85–91.PubMed Astoul, P., et al. (1994). A “patient-like” nude mouse model of parietal pleural human lung adenocarcinoma. Anticancer Research, 14(1A), 85–91.PubMed
92.
go back to reference Budinger, T. F., Benaron, D. A., & Koretsky, A. P. (1999). Imaging transgenic animals. Annual Review of Biomedical Engineering, 1, 611–648.PubMed Budinger, T. F., Benaron, D. A., & Koretsky, A. P. (1999). Imaging transgenic animals. Annual Review of Biomedical Engineering, 1, 611–648.PubMed
93.
go back to reference Hoffman, R. M. (2005). The multiple uses of fluorescent proteins to visualize cancer in vivo. Nature Reviews. Cancer, 5(10), 796–806.PubMed Hoffman, R. M. (2005). The multiple uses of fluorescent proteins to visualize cancer in vivo. Nature Reviews. Cancer, 5(10), 796–806.PubMed
94.
go back to reference Bouvet, M., et al. (2000). Chronologically-specific metastatic targeting of human pancreatic tumors in orthotopic models. Clinical & Experimental Metastasis, 18(3), 213–218. Bouvet, M., et al. (2000). Chronologically-specific metastatic targeting of human pancreatic tumors in orthotopic models. Clinical & Experimental Metastasis, 18(3), 213–218.
95.
go back to reference Bouvet, M., et al. (2002). Real-time optical imaging of primary tumor growth and multiple metastatic events in a pancreatic cancer orthotopic model. Cancer Research, 62(5), 1534–1540.PubMed Bouvet, M., et al. (2002). Real-time optical imaging of primary tumor growth and multiple metastatic events in a pancreatic cancer orthotopic model. Cancer Research, 62(5), 1534–1540.PubMed
96.
go back to reference Katz, M. H., et al. (2003). A novel red fluorescent protein orthotopic pancreatic cancer model for the preclinical evaluation of chemotherapeutics. Journal of Surgical Research, 113(1), 151–160.PubMed Katz, M. H., et al. (2003). A novel red fluorescent protein orthotopic pancreatic cancer model for the preclinical evaluation of chemotherapeutics. Journal of Surgical Research, 113(1), 151–160.PubMed
97.
go back to reference Katz, M. H., et al. (2004). An imageable highly metastatic orthotopic red fluorescent protein model of pancreatic cancer. Clinical & Experimental Metastasis, 21(1), 7–12. Katz, M. H., et al. (2004). An imageable highly metastatic orthotopic red fluorescent protein model of pancreatic cancer. Clinical & Experimental Metastasis, 21(1), 7–12.
98.
go back to reference Bouvet, M., et al. (2005). High correlation of whole-body red fluorescent protein imaging and magnetic resonance imaging on an orthotopic model of pancreatic cancer. Cancer Research, 65(21), 9829–9833.PubMed Bouvet, M., et al. (2005). High correlation of whole-body red fluorescent protein imaging and magnetic resonance imaging on an orthotopic model of pancreatic cancer. Cancer Research, 65(21), 9829–9833.PubMed
99.
go back to reference Katz, M. H., et al. (2003). Selective antimetastatic activity of cytosine analog CS-682 in a red fluorescent protein orthotopic model of pancreatic cancer. Cancer Research, 63(17), 5521–5525.PubMed Katz, M. H., et al. (2003). Selective antimetastatic activity of cytosine analog CS-682 in a red fluorescent protein orthotopic model of pancreatic cancer. Cancer Research, 63(17), 5521–5525.PubMed
100.
go back to reference Katz, M. H., et al. (2004). Survival efficacy of adjuvant cytosine-analogue CS-682 in a fluorescent orthotopic model of human pancreatic cancer. Cancer Research, 64(5), 1828–1833.PubMed Katz, M. H., et al. (2004). Survival efficacy of adjuvant cytosine-analogue CS-682 in a fluorescent orthotopic model of human pancreatic cancer. Cancer Research, 64(5), 1828–1833.PubMed
101.
go back to reference Amoh, Y., et al. (2006). Dual-color imaging of nascent blood vessels vascularizing pancreatic cancer in an orthotopic model demonstrates antiangiogenesis efficacy of gemcitabine. Journal of Surgical Research, 132(2), 164–169.PubMed Amoh, Y., et al. (2006). Dual-color imaging of nascent blood vessels vascularizing pancreatic cancer in an orthotopic model demonstrates antiangiogenesis efficacy of gemcitabine. Journal of Surgical Research, 132(2), 164–169.PubMed
102.
go back to reference Amoh, Y., et al. (2006). Visualization of nascent tumor angiogenesis in lung and liver metastasis by differential dual-color fluorescence imaging in nestin-linked-GFP mice. Clinical & Experimental Metastasis, 23(7–8), 315–322. Amoh, Y., et al. (2006). Visualization of nascent tumor angiogenesis in lung and liver metastasis by differential dual-color fluorescence imaging in nestin-linked-GFP mice. Clinical & Experimental Metastasis, 23(7–8), 315–322.
103.
go back to reference Amoh, Y., et al. (2006). Dual-color imaging of nascent angiogenesis and its inhibition in liver metastases of pancreatic cancer. Anticancer Research, 26(5A), 3237–3242.PubMed Amoh, Y., et al. (2006). Dual-color imaging of nascent angiogenesis and its inhibition in liver metastases of pancreatic cancer. Anticancer Research, 26(5A), 3237–3242.PubMed
104.
go back to reference Sweeney, T. J., et al. (1999). Visualizing the kinetics of tumor-cell clearance in living animals. Proc Natl Acad Sci USA, 96(21), 12044–12049.PubMed Sweeney, T. J., et al. (1999). Visualizing the kinetics of tumor-cell clearance in living animals. Proc Natl Acad Sci USA, 96(21), 12044–12049.PubMed
105.
go back to reference Kunnumakkara, A.B., et al., Zyflamend suppresses growth and sensitizes human pancreatic tumors to gemcitabine in an orthotopic mouse model through modulation of multiple targets. Int J Cancer 2012; 131:E292-E303. Kunnumakkara, A.B., et al., Zyflamend suppresses growth and sensitizes human pancreatic tumors to gemcitabine in an orthotopic mouse model through modulation of multiple targets. Int J Cancer 2012; 131:E292-E303.
106.
go back to reference Hingorani, S. R., et al. (2003). Preinvasive and invasive ductal pancreatic cancer and its early detection in the mouse. Cancer Cell, 4(6), 437–450.PubMed Hingorani, S. R., et al. (2003). Preinvasive and invasive ductal pancreatic cancer and its early detection in the mouse. Cancer Cell, 4(6), 437–450.PubMed
107.
go back to reference Hingorani, S. R., et al. (2005). Trp53R172H and KrasG12D cooperate to promote chromosomal instability and widely metastatic pancreatic ductal adenocarcinoma in mice. Cancer Cell, 7(5), 469–483.PubMed Hingorani, S. R., et al. (2005). Trp53R172H and KrasG12D cooperate to promote chromosomal instability and widely metastatic pancreatic ductal adenocarcinoma in mice. Cancer Cell, 7(5), 469–483.PubMed
108.
go back to reference Tuveson, D. A., & Hingorani, S. R. (2005). Ductal pancreatic cancer in humans and mice. Cold Spring Harbor Symposia on Quantitative Biology, 70, 65–72.PubMed Tuveson, D. A., & Hingorani, S. R. (2005). Ductal pancreatic cancer in humans and mice. Cold Spring Harbor Symposia on Quantitative Biology, 70, 65–72.PubMed
109.
go back to reference Tseng, W.W., et al., Development of an orthotopic model of invasive pancreatic cancer in an immunocompetent murine host. Clin Cancer Res. 16(14): p. 3684–95. Tseng, W.W., et al., Development of an orthotopic model of invasive pancreatic cancer in an immunocompetent murine host. Clin Cancer Res. 16(14): p. 3684–95.
110.
go back to reference Partecke, L. I., et al. (2011). A syngeneic orthotopic murine model of pancreatic adenocarcinoma in the C57/BL6 mouse using the Panc02 and 6606PDA cell lines. European Surgical Research, 47(2), 98–107.PubMed Partecke, L. I., et al. (2011). A syngeneic orthotopic murine model of pancreatic adenocarcinoma in the C57/BL6 mouse using the Panc02 and 6606PDA cell lines. European Surgical Research, 47(2), 98–107.PubMed
111.
go back to reference Wang, B., et al. (2001). A novel, clinically relevant animal model of metastatic pancreatic adenocarcinoma biology and therapy. International Journal of Gastrointestinal Cancer, 29(1), 37–46.PubMed Wang, B., et al. (2001). A novel, clinically relevant animal model of metastatic pancreatic adenocarcinoma biology and therapy. International Journal of Gastrointestinal Cancer, 29(1), 37–46.PubMed
112.
go back to reference Tseng, W. W., et al. (2010). Development of an orthotopic model of invasive pancreatic cancer in an immunocompetent murine host. Clinical Cancer Research, 16(14), 3684–3695.PubMed Tseng, W. W., et al. (2010). Development of an orthotopic model of invasive pancreatic cancer in an immunocompetent murine host. Clinical Cancer Research, 16(14), 3684–3695.PubMed
113.
go back to reference Qiu, W., et al. (2011). Disruption of p16 and activation of Kras in pancreas increase ductal adenocarcinoma formation and metastasis in vivo. Oncotarget, 2(11), 862–873.PubMed Qiu, W., et al. (2011). Disruption of p16 and activation of Kras in pancreas increase ductal adenocarcinoma formation and metastasis in vivo. Oncotarget, 2(11), 862–873.PubMed
114.
go back to reference Chu, Q.D. and R.H. Kim, Immunocompetent orthotopic pancreatic cancer murine model: A step in the right direction. J Surg Res, 2011 [Epub ahead of print] Chu, Q.D. and R.H. Kim, Immunocompetent orthotopic pancreatic cancer murine model: A step in the right direction. J Surg Res, 2011 [Epub ahead of print]
115.
go back to reference Brembeck, F. H., et al. (2003). The mutant K-ras oncogene causes pancreatic periductal lymphocytic infiltration and gastric mucous neck cell hyperplasia in transgenic mice. Cancer Research, 63(9), 2005–2009.PubMed Brembeck, F. H., et al. (2003). The mutant K-ras oncogene causes pancreatic periductal lymphocytic infiltration and gastric mucous neck cell hyperplasia in transgenic mice. Cancer Research, 63(9), 2005–2009.PubMed
116.
go back to reference Jhappan, C., et al. (1990). TGF alpha overexpression in transgenic mice induces liver neoplasia and abnormal development of the mammary gland and pancreas. Cell, 61(6), 1137–1146.PubMed Jhappan, C., et al. (1990). TGF alpha overexpression in transgenic mice induces liver neoplasia and abnormal development of the mammary gland and pancreas. Cell, 61(6), 1137–1146.PubMed
117.
go back to reference Pujal, J., et al. (2009). Keratin 7 promoter selectively targets transgene expression to normal and neoplastic pancreatic ductal cells in vitro and in vivo. The FASEB Journal, 23(5), 1366–1375. Pujal, J., et al. (2009). Keratin 7 promoter selectively targets transgene expression to normal and neoplastic pancreatic ductal cells in vitro and in vivo. The FASEB Journal, 23(5), 1366–1375.
118.
go back to reference Offield, M. F., et al. (1996). PDX-1 is required for pancreatic outgrowth and differentiation of the rostral duodenum. Development, 122(3), 983–995.PubMed Offield, M. F., et al. (1996). PDX-1 is required for pancreatic outgrowth and differentiation of the rostral duodenum. Development, 122(3), 983–995.PubMed
119.
go back to reference Aichler, M., et al. (2012). Origin of pancreatic ductal adenocarcinoma from atypical flat lesions: A comparative study in transgenic mice and human tissues. The Journal of Pathology, 226(5), 723–734. Aichler, M., et al. (2012). Origin of pancreatic ductal adenocarcinoma from atypical flat lesions: A comparative study in transgenic mice and human tissues. The Journal of Pathology, 226(5), 723–734.
120.
go back to reference Zhu, L., et al. (2007). Acinar cells contribute to the molecular heterogeneity of pancreatic intraepithelial neoplasia. American Journal of Pathology, 171(1), 263–273.PubMed Zhu, L., et al. (2007). Acinar cells contribute to the molecular heterogeneity of pancreatic intraepithelial neoplasia. American Journal of Pathology, 171(1), 263–273.PubMed
121.
go back to reference Means, A. L., et al. (2005). Pancreatic epithelial plasticity mediated by acinar cell transdifferentiation and generation of nestin-positive intermediates. Development, 132(16), 3767–3776.PubMed Means, A. L., et al. (2005). Pancreatic epithelial plasticity mediated by acinar cell transdifferentiation and generation of nestin-positive intermediates. Development, 132(16), 3767–3776.PubMed
122.
go back to reference Hruban, R. H., et al. (2006). Pathology of genetically engineered mouse models of pancreatic exocrine cancer: Consensus report and recommendations. Cancer Research, 66(1), 95–106.PubMed Hruban, R. H., et al. (2006). Pathology of genetically engineered mouse models of pancreatic exocrine cancer: Consensus report and recommendations. Cancer Research, 66(1), 95–106.PubMed
123.
go back to reference Ornitz, D. M., et al. (1987). Pancreatic neoplasia induced by SV40 T-antigen expression in acinar cells of transgenic mice. Science, 238(4824), 188–193.PubMed Ornitz, D. M., et al. (1987). Pancreatic neoplasia induced by SV40 T-antigen expression in acinar cells of transgenic mice. Science, 238(4824), 188–193.PubMed
124.
go back to reference Quaife, C. J., et al. (1987). Pancreatic neoplasia induced by ras expression in acinar cells of transgenic mice. Cell, 48(6), 1023–1034.PubMed Quaife, C. J., et al. (1987). Pancreatic neoplasia induced by ras expression in acinar cells of transgenic mice. Cell, 48(6), 1023–1034.PubMed
125.
go back to reference Lewis, B. C., Klimstra, D. S., & Varmus, H. E. (2003). The c-myc and PyMT oncogenes induce different tumor types in a somatic mouse model for pancreatic cancer. Genes & Development, 17(24), 3127–3138. Lewis, B. C., Klimstra, D. S., & Varmus, H. E. (2003). The c-myc and PyMT oncogenes induce different tumor types in a somatic mouse model for pancreatic cancer. Genes & Development, 17(24), 3127–3138.
126.
go back to reference Sandgren, E. P., et al. (1991). Pancreatic tumor pathogenesis reflects the causative genetic lesion. Proc Natl Acad Sci USA, 88(1), 93–97.PubMed Sandgren, E. P., et al. (1991). Pancreatic tumor pathogenesis reflects the causative genetic lesion. Proc Natl Acad Sci USA, 88(1), 93–97.PubMed
127.
go back to reference Liao, D. J., et al. (2006). Characterization of pancreatic lesions from MT-tgf alpha, Ela-myc and MT-tgf alpha/Ela-myc single and double transgenic mice. J Carcinog, 5, 19.PubMed Liao, D. J., et al. (2006). Characterization of pancreatic lesions from MT-tgf alpha, Ela-myc and MT-tgf alpha/Ela-myc single and double transgenic mice. J Carcinog, 5, 19.PubMed
128.
go back to reference Grippo, P.J. and E.P. Sandgren. Acinar-to-ductal metaplasia accompanies c-myc-induced exocrine pancreatic cancer progression in transgenic rodents. Int J Cancer, 2012;131:1243-1248. Grippo, P.J. and E.P. Sandgren. Acinar-to-ductal metaplasia accompanies c-myc-induced exocrine pancreatic cancer progression in transgenic rodents. Int J Cancer, 2012;131:1243-1248.
129.
go back to reference Grippo, P. J., et al. (2003). Preinvasive pancreatic neoplasia of ductal phenotype induced by acinar cell targeting of mutant Kras in transgenic mice. Cancer Research, 63(9), 2016–2019.PubMed Grippo, P. J., et al. (2003). Preinvasive pancreatic neoplasia of ductal phenotype induced by acinar cell targeting of mutant Kras in transgenic mice. Cancer Research, 63(9), 2016–2019.PubMed
130.
go back to reference Jones, S., et al. (2008). Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science, 321(5897), 1801–1806.PubMed Jones, S., et al. (2008). Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science, 321(5897), 1801–1806.PubMed
131.
go back to reference Thayer, S. P., et al. (2003). Hedgehog is an early and late mediator of pancreatic cancer tumorigenesis. Nature, 425(6960), 851–856.PubMed Thayer, S. P., et al. (2003). Hedgehog is an early and late mediator of pancreatic cancer tumorigenesis. Nature, 425(6960), 851–856.PubMed
132.
go back to reference Pasca di Magliano, M. (2006). Hedgehog/Ras interactions regulate early stages of pancreatic cancer. Genes & Development, 20(22), 3161–3173. Pasca di Magliano, M. (2006). Hedgehog/Ras interactions regulate early stages of pancreatic cancer. Genes & Development, 20(22), 3161–3173.
133.
go back to reference Sandgren, E. P., et al. (1990). Overexpression of TGF alpha in transgenic mice: Induction of epithelial hyperplasia, pancreatic metaplasia, and carcinoma of the breast. Cell, 61(6), 1121–1135.PubMed Sandgren, E. P., et al. (1990). Overexpression of TGF alpha in transgenic mice: Induction of epithelial hyperplasia, pancreatic metaplasia, and carcinoma of the breast. Cell, 61(6), 1121–1135.PubMed
134.
go back to reference Schmid, R. M., et al. (1999). Acinar-ductal-carcinoma sequence in transforming growth factor-alpha transgenic mice. Annals of the New York Academy of Sciences, 880, 219–230.PubMed Schmid, R. M., et al. (1999). Acinar-ductal-carcinoma sequence in transforming growth factor-alpha transgenic mice. Annals of the New York Academy of Sciences, 880, 219–230.PubMed
135.
go back to reference Liao, D. J., et al. (2000). Cell cycle basis for the onset and progression of c-Myc-induced, TGFalpha-enhanced mouse mammary gland carcinogenesis. Oncogene, 19(10), 1307–1317.PubMed Liao, D. J., et al. (2000). Cell cycle basis for the onset and progression of c-Myc-induced, TGFalpha-enhanced mouse mammary gland carcinogenesis. Oncogene, 19(10), 1307–1317.PubMed
136.
go back to reference Thorgeirsson, S. S., Factor, V. M., & Snyderwine, E. G. (2000). Transgenic mouse models in carcinogenesis research and testing. Toxicology Letters, 112–113, 553–555.PubMed Thorgeirsson, S. S., Factor, V. M., & Snyderwine, E. G. (2000). Transgenic mouse models in carcinogenesis research and testing. Toxicology Letters, 112–113, 553–555.PubMed
137.
go back to reference Malka, D., et al. (2002). Risk of pancreatic adenocarcinoma in chronic pancreatitis. Gut, 51(6), 849–852.PubMed Malka, D., et al. (2002). Risk of pancreatic adenocarcinoma in chronic pancreatitis. Gut, 51(6), 849–852.PubMed
138.
go back to reference Norman, J. G., et al. (1996). Transgenic animals demonstrate a role for the IL-1 receptor in regulating IL-1beta gene expression at steady-state and during the systemic stress induced by acute pancreatitis. Journal of Surgical Research, 63(1), 231–236.PubMed Norman, J. G., et al. (1996). Transgenic animals demonstrate a role for the IL-1 receptor in regulating IL-1beta gene expression at steady-state and during the systemic stress induced by acute pancreatitis. Journal of Surgical Research, 63(1), 231–236.PubMed
139.
go back to reference Marrache, F., et al. (2008). Overexpression of interleukin-1beta in the murine pancreas results in chronic pancreatitis. Gastroenterology, 135(4), 1277–1287.PubMed Marrache, F., et al. (2008). Overexpression of interleukin-1beta in the murine pancreas results in chronic pancreatitis. Gastroenterology, 135(4), 1277–1287.PubMed
140.
go back to reference Yip-Schneider, M. T., et al. (2000). Cyclooxygenase-2 expression in human pancreatic adenocarcinomas. Carcinogenesis, 21(2), 139–146.PubMed Yip-Schneider, M. T., et al. (2000). Cyclooxygenase-2 expression in human pancreatic adenocarcinomas. Carcinogenesis, 21(2), 139–146.PubMed
141.
go back to reference Muller-Decker, K., et al. (2006). Preinvasive duct-derived neoplasms in pancreas of keratin 5-promoter cyclooxygenase-2 transgenic mice. Gastroenterology, 130(7), 2165–2178.PubMed Muller-Decker, K., et al. (2006). Preinvasive duct-derived neoplasms in pancreas of keratin 5-promoter cyclooxygenase-2 transgenic mice. Gastroenterology, 130(7), 2165–2178.PubMed
142.
go back to reference Neufang, G., et al. (2001). Abnormal differentiation of epidermis in transgenic mice constitutively expressing cyclooxygenase-2 in skin. Proc Natl Acad Sci USA, 98(13), 7629–7634.PubMed Neufang, G., et al. (2001). Abnormal differentiation of epidermis in transgenic mice constitutively expressing cyclooxygenase-2 in skin. Proc Natl Acad Sci USA, 98(13), 7629–7634.PubMed
143.
go back to reference Colby, J. K., et al. (2008). Progressive metaplastic and dysplastic changes in mouse pancreas induced by cyclooxygenase-2 overexpression. Neoplasia, 10(8), 782–796.PubMed Colby, J. K., et al. (2008). Progressive metaplastic and dysplastic changes in mouse pancreas induced by cyclooxygenase-2 overexpression. Neoplasia, 10(8), 782–796.PubMed
144.
go back to reference Tang, C., Biemond, I., & Lamers, C. B. (1996). Cholecystokinin receptors in human pancreas and gallbladder muscle: A comparative study. Gastroenterology, 111(6), 1621–1626.PubMed Tang, C., Biemond, I., & Lamers, C. B. (1996). Cholecystokinin receptors in human pancreas and gallbladder muscle: A comparative study. Gastroenterology, 111(6), 1621–1626.PubMed
145.
go back to reference Saillan-Barreau, C., et al. (1998). Transgenic CCK-B/gastrin receptor mediates murine exocrine pancreatic secretion. Gastroenterology, 115(4), 988–996.PubMed Saillan-Barreau, C., et al. (1998). Transgenic CCK-B/gastrin receptor mediates murine exocrine pancreatic secretion. Gastroenterology, 115(4), 988–996.PubMed
146.
go back to reference Mathieu, A., et al. (2005). Transgenic expression of CCK2 receptors sensitizes murine pancreatic acinar cells to carcinogen-induced preneoplastic lesions formation. International Journal of Cancer, 115(1), 46–54. Mathieu, A., et al. (2005). Transgenic expression of CCK2 receptors sensitizes murine pancreatic acinar cells to carcinogen-induced preneoplastic lesions formation. International Journal of Cancer, 115(1), 46–54.
147.
go back to reference Goggins, M., et al. (1998). Genetic alterations of the transforming growth factor beta receptor genes in pancreatic and biliary adenocarcinomas. Cancer Research, 58, 5329–5332.PubMed Goggins, M., et al. (1998). Genetic alterations of the transforming growth factor beta receptor genes in pancreatic and biliary adenocarcinomas. Cancer Research, 58, 5329–5332.PubMed
148.
go back to reference Kuang, C., et al. (2006). In vivo disruption of TGF-beta signaling by Smad7 leads to premalignant ductal lesions in the pancreas. Proc Natl Acad Sci USA, 103(6), 1858–1863.PubMed Kuang, C., et al. (2006). In vivo disruption of TGF-beta signaling by Smad7 leads to premalignant ductal lesions in the pancreas. Proc Natl Acad Sci USA, 103(6), 1858–1863.PubMed
149.
go back to reference Izeradjene, K., et al. (2007). Kras(G12D) and Smad4/Dpc4 haploinsufficiency cooperate to induce mucinous cystic neoplasms and invasive adenocarcinoma of the pancreas. Cancer Cell, 11(3), 229–243.PubMed Izeradjene, K., et al. (2007). Kras(G12D) and Smad4/Dpc4 haploinsufficiency cooperate to induce mucinous cystic neoplasms and invasive adenocarcinoma of the pancreas. Cancer Cell, 11(3), 229–243.PubMed
150.
go back to reference Wagner, M., et al. (1998). Malignant transformation of duct-like cells originating from acini in transforming growth factor transgenic mice. Gastroenterology, 115(5), 1254–1262.PubMed Wagner, M., et al. (1998). Malignant transformation of duct-like cells originating from acini in transforming growth factor transgenic mice. Gastroenterology, 115(5), 1254–1262.PubMed
151.
go back to reference Glasner, S., Memoli, V., & Longnecker, D. S. (1992). Characterization of the ELSV transgenic mouse model of pancreatic carcinoma. Histologic type of large and small tumors. American Journal of Pathology, 140(5), 1237–1245.PubMed Glasner, S., Memoli, V., & Longnecker, D. S. (1992). Characterization of the ELSV transgenic mouse model of pancreatic carcinoma. Histologic type of large and small tumors. American Journal of Pathology, 140(5), 1237–1245.PubMed
152.
go back to reference Bell, R. H., Jr., Memol, V. A., & Longnecker, D. S. (1990). Hyperplasia and tumors of the islets of Langerhans in mice bearing an elastase I-SV40 T-antigen fusion gene. Carcinogenesis, 11(8), 1393–1398.PubMed Bell, R. H., Jr., Memol, V. A., & Longnecker, D. S. (1990). Hyperplasia and tumors of the islets of Langerhans in mice bearing an elastase I-SV40 T-antigen fusion gene. Carcinogenesis, 11(8), 1393–1398.PubMed
153.
go back to reference Schaeffer, B. K., Terhune, P. G., & Longnecker, D. S. (1994). Pancreatic carcinomas of acinar and mixed acinar/ductal phenotypes in Ela-1-myc transgenic mice do not contain c-K-ras mutations. American Journal of Pathology, 145(3), 696–701.PubMed Schaeffer, B. K., Terhune, P. G., & Longnecker, D. S. (1994). Pancreatic carcinomas of acinar and mixed acinar/ductal phenotypes in Ela-1-myc transgenic mice do not contain c-K-ras mutations. American Journal of Pathology, 145(3), 696–701.PubMed
154.
go back to reference Efrat, S., et al. (1988). Beta-cell lines derived from transgenic mice expressing a hybrid insulin gene-oncogene. Proc Natl Acad Sci USA, 85(23), 9037–9041.PubMed Efrat, S., et al. (1988). Beta-cell lines derived from transgenic mice expressing a hybrid insulin gene-oncogene. Proc Natl Acad Sci USA, 85(23), 9037–9041.PubMed
155.
go back to reference Bottinger, E. P., et al. (1997). Expression of a dominant-negative mutant TGF-beta type II receptor in transgenic mice reveals essential roles for TGF-beta in regulation of growth and differentiation in the exocrine pancreas. EMBO Journal, 16(10), 2621–2633.PubMed Bottinger, E. P., et al. (1997). Expression of a dominant-negative mutant TGF-beta type II receptor in transgenic mice reveals essential roles for TGF-beta in regulation of growth and differentiation in the exocrine pancreas. EMBO Journal, 16(10), 2621–2633.PubMed
156.
go back to reference Yamaoka, T., et al. (2000). Diabetes and pancreatic tumours in transgenic mice expressing Pa x 6. Diabetologia, 43(3), 332–339.PubMed Yamaoka, T., et al. (2000). Diabetes and pancreatic tumours in transgenic mice expressing Pa x 6. Diabetologia, 43(3), 332–339.PubMed
157.
go back to reference Wagner, M., et al. (2001). A murine tumor progression model for pancreatic cancer recapitulating the genetic alterations of the human disease. Genes & Development, 15(3), 286–293. Wagner, M., et al. (2001). A murine tumor progression model for pancreatic cancer recapitulating the genetic alterations of the human disease. Genes & Development, 15(3), 286–293.
158.
go back to reference Schreiner, B., et al. (2003). Pattern of secondary genomic changes in pancreatic tumors of Tgf alpha/Trp53+/− transgenic mice. Genes, Chromosomes & Cancer, 38(3), 240–248. Schreiner, B., et al. (2003). Pattern of secondary genomic changes in pancreatic tumors of Tgf alpha/Trp53+/− transgenic mice. Genes, Chromosomes & Cancer, 38(3), 240–248.
159.
go back to reference Bardeesy, N., et al. (2002). Obligate roles for p16(Ink4a) and p19(Arf)-p53 in the suppression of murine pancreatic neoplasia. Molecular and Cellular Biology, 22(2), 635–643.PubMed Bardeesy, N., et al. (2002). Obligate roles for p16(Ink4a) and p19(Arf)-p53 in the suppression of murine pancreatic neoplasia. Molecular and Cellular Biology, 22(2), 635–643.PubMed
160.
go back to reference Adrian, K., et al. (2009). Tgfbr1 haploinsufficiency inhibits the development of murine mutant Kras-induced pancreatic precancer. Cancer Research, 69(24), 9169–9174.PubMed Adrian, K., et al. (2009). Tgfbr1 haploinsufficiency inhibits the development of murine mutant Kras-induced pancreatic precancer. Cancer Research, 69(24), 9169–9174.PubMed
161.
go back to reference Grippo, P.J., et al., Concurrent PEDF deficiency and Kras mutation induce invasive pancreatic cancer and adipose-rich stroma in mice. Gut, 2012;61:1454-1464. Grippo, P.J., et al., Concurrent PEDF deficiency and Kras mutation induce invasive pancreatic cancer and adipose-rich stroma in mice. Gut, 2012;61:1454-1464.
162.
go back to reference Krantz, S. B., et al. (2011). MT1-MMP cooperates with Kras(G12D) to promote pancreatic fibrosis through increased TGF-beta signaling. Molecular Cancer Research, 9(10), 1294–1304.PubMed Krantz, S. B., et al. (2011). MT1-MMP cooperates with Kras(G12D) to promote pancreatic fibrosis through increased TGF-beta signaling. Molecular Cancer Research, 9(10), 1294–1304.PubMed
163.
go back to reference Sandgren, E. P., et al. (1993). Transforming growth factor alpha dramatically enhances oncogene-induced carcinogenesis in transgenic mouse pancreas and liver. Molecular and Cellular Biology, 13(1), 320–330.PubMed Sandgren, E. P., et al. (1993). Transforming growth factor alpha dramatically enhances oncogene-induced carcinogenesis in transgenic mouse pancreas and liver. Molecular and Cellular Biology, 13(1), 320–330.PubMed
164.
go back to reference Fisher, G. H., et al. (1999). Development of a flexible and specific gene delivery system for production of murine tumor models. Oncogene, 18(38), 5253–5260.PubMed Fisher, G. H., et al. (1999). Development of a flexible and specific gene delivery system for production of murine tumor models. Oncogene, 18(38), 5253–5260.PubMed
165.
go back to reference Morton, J. P., et al. (2008). Trp53 deletion stimulates the formation of metastatic pancreatic tumors. American Journal of Pathology, 172(4), 1081–1087.PubMed Morton, J. P., et al. (2008). Trp53 deletion stimulates the formation of metastatic pancreatic tumors. American Journal of Pathology, 172(4), 1081–1087.PubMed
166.
go back to reference Song, S. Y., et al. (1999). Expansion of Pdx1-expressing pancreatic epithelium and islet neogenesis in transgenic mice overexpressing transforming growth factor alpha. Gastroenterology, 117(6), 1416–1426.PubMed Song, S. Y., et al. (1999). Expansion of Pdx1-expressing pancreatic epithelium and islet neogenesis in transgenic mice overexpressing transforming growth factor alpha. Gastroenterology, 117(6), 1416–1426.PubMed
167.
go back to reference Greten, F. R., et al. (2001). TGF alpha transgenic mice. A model of pancreatic cancer development. Pancreatology, 1(4), 363–368.PubMed Greten, F. R., et al. (2001). TGF alpha transgenic mice. A model of pancreatic cancer development. Pancreatology, 1(4), 363–368.PubMed
168.
go back to reference Siveke, J. T., et al. (2007). Concomitant pancreatic activation of Kras(G12D) and Tgfa results in cystic papillary neoplasms reminiscent of human IPMN. Cancer Cell, 12(3), 266–279.PubMed Siveke, J. T., et al. (2007). Concomitant pancreatic activation of Kras(G12D) and Tgfa results in cystic papillary neoplasms reminiscent of human IPMN. Cancer Cell, 12(3), 266–279.PubMed
169.
go back to reference Schutte, M., et al. (1996). DPC4 gene in various tumor types. Cancer Research, 56(11), 2527–2530.PubMed Schutte, M., et al. (1996). DPC4 gene in various tumor types. Cancer Research, 56(11), 2527–2530.PubMed
170.
go back to reference Bardeesy, N., et al. (2006). Smad4 is dispensable for normal pancreas development yet critical in progression and tumor biology of pancreas cancer. Genes & Development, 20(22), 3130–3146. Bardeesy, N., et al. (2006). Smad4 is dispensable for normal pancreas development yet critical in progression and tumor biology of pancreas cancer. Genes & Development, 20(22), 3130–3146.
171.
go back to reference Ijichi, H., et al. (2006). Aggressive pancreatic ductal adenocarcinoma in mice caused by pancreas-specific blockade of transforming growth factor-beta signaling in cooperation with active Kras expression. Genes & Development, 20(22), 3147–3160. Ijichi, H., et al. (2006). Aggressive pancreatic ductal adenocarcinoma in mice caused by pancreas-specific blockade of transforming growth factor-beta signaling in cooperation with active Kras expression. Genes & Development, 20(22), 3147–3160.
172.
go back to reference Kojima, K., et al. (2007). Inactivation of Smad4 accelerates Kras(G12D)-mediated pancreatic neoplasia. Cancer Research, 67(17), 8121–8130.PubMed Kojima, K., et al. (2007). Inactivation of Smad4 accelerates Kras(G12D)-mediated pancreatic neoplasia. Cancer Research, 67(17), 8121–8130.PubMed
173.
go back to reference Akhurst, R. J. (2002). TGF-beta antagonists: Why suppress a tumor suppressor? The Journal of Clinical Investigation, 109(12), 1533–1536.PubMed Akhurst, R. J. (2002). TGF-beta antagonists: Why suppress a tumor suppressor? The Journal of Clinical Investigation, 109(12), 1533–1536.PubMed
174.
go back to reference Mathur, A., et al. (2009). Pancreatic steatosis promotes dissemination and lethality of pancreatic cancer. Journal of the American College of Surgery, 208(5), 989–994. discussion 994–6. Mathur, A., et al. (2009). Pancreatic steatosis promotes dissemination and lethality of pancreatic cancer. Journal of the American College of Surgery, 208(5), 989–994. discussion 994–6.
175.
go back to reference Yamagishi, S., et al. (2010). Positive association of circulating levels of advanced glycation end products (AGEs) with pigment epithelium-derived factor (PEDF) in a general population. Pharmacological Research, 61(2), 103–107.PubMed Yamagishi, S., et al. (2010). Positive association of circulating levels of advanced glycation end products (AGEs) with pigment epithelium-derived factor (PEDF) in a general population. Pharmacological Research, 61(2), 103–107.PubMed
176.
go back to reference Olive, K. P., et al. (2009). Inhibition of hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer. Science, 324(5933), 1457–1461.PubMed Olive, K. P., et al. (2009). Inhibition of hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer. Science, 324(5933), 1457–1461.PubMed
177.
go back to reference Guerra, C., et al. (2007). Chronic pancreatitis is essential for induction of pancreatic ductal adenocarcinoma by K-Ras oncogenes in adult mice. Cancer Cell, 11(3), 291–302.PubMed Guerra, C., et al. (2007). Chronic pancreatitis is essential for induction of pancreatic ductal adenocarcinoma by K-Ras oncogenes in adult mice. Cancer Cell, 11(3), 291–302.PubMed
178.
go back to reference Shields, J. M., et al. (2000). Understanding Ras: ‘It ain’t over ‘til it’s over’. Trends in Cell Biology, 10(4), 147–154.PubMed Shields, J. M., et al. (2000). Understanding Ras: ‘It ain’t over ‘til it’s over’. Trends in Cell Biology, 10(4), 147–154.PubMed
179.
go back to reference Guerra, C., et al. (2003). Tumor induction by an endogenous K-ras oncogene is highly dependent on cellular context. Cancer Cell, 4(2), 111–120.PubMed Guerra, C., et al. (2003). Tumor induction by an endogenous K-ras oncogene is highly dependent on cellular context. Cancer Cell, 4(2), 111–120.PubMed
180.
go back to reference Tuveson, D. A., et al. (2004). Endogenous oncogenic K-ras(G12D) stimulates proliferation and widespread neoplastic and developmental defects. Cancer Cell, 5(4), 375–387.PubMed Tuveson, D. A., et al. (2004). Endogenous oncogenic K-ras(G12D) stimulates proliferation and widespread neoplastic and developmental defects. Cancer Cell, 5(4), 375–387.PubMed
181.
go back to reference Pin, C. L., Bonvissuto, A. C., & Konieczny, S. F. (2000). Mist1 expression is a common link among serous exocrine cells exhibiting regulated exocytosis. Anatomical Record, 259(2), 157–167.PubMed Pin, C. L., Bonvissuto, A. C., & Konieczny, S. F. (2000). Mist1 expression is a common link among serous exocrine cells exhibiting regulated exocytosis. Anatomical Record, 259(2), 157–167.PubMed
182.
go back to reference Pin, C. L., et al. (2001). The bHLH transcription factor Mist1 is required to maintain exocrine pancreas cell organization and acinar cell identity. The Journal of Cell Biology, 155(4), 519–530.PubMed Pin, C. L., et al. (2001). The bHLH transcription factor Mist1 is required to maintain exocrine pancreas cell organization and acinar cell identity. The Journal of Cell Biology, 155(4), 519–530.PubMed
183.
go back to reference Tuveson, D. A., et al. (2006). Mist1-KrasG12D knock-in mice develop mixed differentiation metastatic exocrine pancreatic carcinoma and hepatocellular carcinoma. Cancer Research, 66(1), 242–247.PubMed Tuveson, D. A., et al. (2006). Mist1-KrasG12D knock-in mice develop mixed differentiation metastatic exocrine pancreatic carcinoma and hepatocellular carcinoma. Cancer Research, 66(1), 242–247.PubMed
184.
go back to reference Shi, G., et al. (2009). Loss of the acinar-restricted transcription factor Mist1 accelerates Kras-induced pancreatic intraepithelial neoplasia. Gastroenterology, 136(4), 1368–1378.PubMed Shi, G., et al. (2009). Loss of the acinar-restricted transcription factor Mist1 accelerates Kras-induced pancreatic intraepithelial neoplasia. Gastroenterology, 136(4), 1368–1378.PubMed
185.
go back to reference Mao, X., Fujiwara, Y., & Orkin, S. H. (1999). Improved reporter strain for monitoring Cre recombinase-mediated DNA excisions in mice. Proc Natl Acad Sci USA, 96(9), 5037–5042.PubMed Mao, X., Fujiwara, Y., & Orkin, S. H. (1999). Improved reporter strain for monitoring Cre recombinase-mediated DNA excisions in mice. Proc Natl Acad Sci USA, 96(9), 5037–5042.PubMed
186.
go back to reference Lakso, M., et al. (1992). Targeted oncogene activation by site-specific recombination in transgenic mice. Proc Natl Acad Sci USA, 89(14), 6232–6236.PubMed Lakso, M., et al. (1992). Targeted oncogene activation by site-specific recombination in transgenic mice. Proc Natl Acad Sci USA, 89(14), 6232–6236.PubMed
187.
go back to reference Jackson, E. L., et al. (2001). Analysis of lung tumor initiation and progression using conditional expression of oncogenic K-ras. Genes & Development, 15(24), 3243–3248. Jackson, E. L., et al. (2001). Analysis of lung tumor initiation and progression using conditional expression of oncogenic K-ras. Genes & Development, 15(24), 3243–3248.
188.
go back to reference Carriere, C., et al. (2007). The nestin progenitor lineage is the compartment of origin for pancreatic intraepithelial neoplasia. Proc Natl Acad Sci USA, 104(11), 4437–4442.PubMed Carriere, C., et al. (2007). The nestin progenitor lineage is the compartment of origin for pancreatic intraepithelial neoplasia. Proc Natl Acad Sci USA, 104(11), 4437–4442.PubMed
189.
go back to reference Leach, S. D. (2005). Epithelial differentiation in pancreatic development and neoplasia: New niches for nestin and Notch. Journal of Clinical Gastroenterology, 39(4 Suppl 2), S78–S82.PubMed Leach, S. D. (2005). Epithelial differentiation in pancreatic development and neoplasia: New niches for nestin and Notch. Journal of Clinical Gastroenterology, 39(4 Suppl 2), S78–S82.PubMed
190.
go back to reference Fendrich, V., et al. (2008). Hedgehog signaling is required for effective regeneration of exocrine pancreas. Gastroenterology, 135(2), 621–631.PubMed Fendrich, V., et al. (2008). Hedgehog signaling is required for effective regeneration of exocrine pancreas. Gastroenterology, 135(2), 621–631.PubMed
191.
go back to reference Habbe, N., et al. (2008). Spontaneous induction of murine pancreatic intraepithelial neoplasia (mPanIN) by acinar cell targeting of oncogenic Kras in adult mice. Proc Natl Acad Sci USA, 105(48), 18913–18918.PubMed Habbe, N., et al. (2008). Spontaneous induction of murine pancreatic intraepithelial neoplasia (mPanIN) by acinar cell targeting of oncogenic Kras in adult mice. Proc Natl Acad Sci USA, 105(48), 18913–18918.PubMed
192.
go back to reference Trobridge, P., et al. (2009). TGF-beta receptor inactivation and mutant Kras induce intestinal neoplasms in mice via a beta-catenin-independent pathway. Gastroenterology, 136(5), 1680–1688. e7.PubMed Trobridge, P., et al. (2009). TGF-beta receptor inactivation and mutant Kras induce intestinal neoplasms in mice via a beta-catenin-independent pathway. Gastroenterology, 136(5), 1680–1688. e7.PubMed
193.
go back to reference Kim, W. Y., et al. (2009). HIF2alpha cooperates with RAS to promote lung tumorigenesis in mice. The Journal of Clinical Investigation, 119(8), 2160–2170.PubMed Kim, W. Y., et al. (2009). HIF2alpha cooperates with RAS to promote lung tumorigenesis in mice. The Journal of Clinical Investigation, 119(8), 2160–2170.PubMed
194.
go back to reference Fan, H. Y., et al. (2009). Cell type-specific targeted mutations of Kras and Pten document proliferation arrest in granulosa cells versus oncogenic insult to ovarian surface epithelial cells. Cancer Research, 69(16), 6463–6472.PubMed Fan, H. Y., et al. (2009). Cell type-specific targeted mutations of Kras and Pten document proliferation arrest in granulosa cells versus oncogenic insult to ovarian surface epithelial cells. Cancer Research, 69(16), 6463–6472.PubMed
195.
go back to reference Dail, M., et al. (2010). Mutant Ikzf1, KrasG12D, and Notch1 cooperate in T lineage leukemogenesis and modulate responses to targeted agents. Proc Natl Acad Sci USA, 107(11), 5106–5111.PubMed Dail, M., et al. (2010). Mutant Ikzf1, KrasG12D, and Notch1 cooperate in T lineage leukemogenesis and modulate responses to targeted agents. Proc Natl Acad Sci USA, 107(11), 5106–5111.PubMed
196.
go back to reference Sund, N. J., et al. (2001). Tissue-specific deletion of Foxa2 in pancreatic beta cells results in hyperinsulinemic hypoglycemia. Genes & Development, 15(13), 1706–1715. Sund, N. J., et al. (2001). Tissue-specific deletion of Foxa2 in pancreatic beta cells results in hyperinsulinemic hypoglycemia. Genes & Development, 15(13), 1706–1715.
197.
go back to reference Su, Y., et al., N-Cadherin haploinsufficiency increases survival in a mouse model of pancreatic cancer. Oncogene, 2012;31;4484-4489. Su, Y., et al., N-Cadherin haploinsufficiency increases survival in a mouse model of pancreatic cancer. Oncogene, 2012;31;4484-4489.
198.
go back to reference Karlsson, R., et al. (2009). Rho GTPase function in tumorigenesis. Biochimica et Biophysica Acta, 1796(2), 91–98.PubMed Karlsson, R., et al. (2009). Rho GTPase function in tumorigenesis. Biochimica et Biophysica Acta, 1796(2), 91–98.PubMed
199.
go back to reference Heid, I., et al. (2011). Early requirement of Rac1 in a mouse model of pancreatic cancer. Gastroenterology, 141(2), 719–730. 730 e1-7.PubMed Heid, I., et al. (2011). Early requirement of Rac1 in a mouse model of pancreatic cancer. Gastroenterology, 141(2), 719–730. 730 e1-7.PubMed
200.
go back to reference Nakhai, H., et al. (2008). Conditional ablation of Notch signaling in pancreatic development. Development, 135(16), 2757–2765.PubMed Nakhai, H., et al. (2008). Conditional ablation of Notch signaling in pancreatic development. Development, 135(16), 2757–2765.PubMed
201.
go back to reference Stanger, B. Z., et al. (2005). Pten constrains centroacinar cell expansion and malignant transformation in the pancreas. Cancer Cell, 8(3), 185–195.PubMed Stanger, B. Z., et al. (2005). Pten constrains centroacinar cell expansion and malignant transformation in the pancreas. Cancer Cell, 8(3), 185–195.PubMed
202.
go back to reference Rowley, M., et al. (2011). Inactivation of Brca2 promotes Trp53-associated but inhibits KrasG12D-dependent pancreatic cancer development in mice. Gastroenterology, 140(4), 1303–1313. e1-3.PubMed Rowley, M., et al. (2011). Inactivation of Brca2 promotes Trp53-associated but inhibits KrasG12D-dependent pancreatic cancer development in mice. Gastroenterology, 140(4), 1303–1313. e1-3.PubMed
203.
go back to reference Feldmann, G., et al. (2011). Inactivation of Brca2 cooperates with Trp53(R172H) to induce invasive pancreatic ductal adenocarcinomas in mice: A mouse model of familial pancreatic cancer. Cancer Biology & Therapy, 11(11), 959–968. Feldmann, G., et al. (2011). Inactivation of Brca2 cooperates with Trp53(R172H) to induce invasive pancreatic ductal adenocarcinomas in mice: A mouse model of familial pancreatic cancer. Cancer Biology & Therapy, 11(11), 959–968.
204.
go back to reference Bardeesy, N., et al. (2006). Both p16(Ink4a) and the p19(Arf)-p53 pathway constrain progression of pancreatic adenocarcinoma in the mouse. Proc Natl Acad Sci USA, 103(15), 5947–5952.PubMed Bardeesy, N., et al. (2006). Both p16(Ink4a) and the p19(Arf)-p53 pathway constrain progression of pancreatic adenocarcinoma in the mouse. Proc Natl Acad Sci USA, 103(15), 5947–5952.PubMed
205.
go back to reference Corcoran, R. B., et al. (2011). STAT3 plays a critical role in KRAS-induced pancreatic tumorigenesis. Cancer Research, 71(14), 5020–5029.PubMed Corcoran, R. B., et al. (2011). STAT3 plays a critical role in KRAS-induced pancreatic tumorigenesis. Cancer Research, 71(14), 5020–5029.PubMed
206.
go back to reference Fukuda, A., et al. (2011). Stat3 and MMP7 contribute to pancreatic ductal adenocarcinoma initiation and progression. Cancer Cell, 19(4), 441–455.PubMed Fukuda, A., et al. (2011). Stat3 and MMP7 contribute to pancreatic ductal adenocarcinoma initiation and progression. Cancer Cell, 19(4), 441–455.PubMed
207.
go back to reference Siveke, J. T., et al. (2008). Notch signaling is required for exocrine regeneration after acute pancreatitis. Gastroenterology, 134(2), 544–555.PubMed Siveke, J. T., et al. (2008). Notch signaling is required for exocrine regeneration after acute pancreatitis. Gastroenterology, 134(2), 544–555.PubMed
208.
go back to reference Mazur, P. K., et al. (2010). Notch2 is required for progression of pancreatic intraepithelial neoplasia and development of pancreatic ductal adenocarcinoma. Proc Natl Acad Sci USA, 107(30), 13438–13443.PubMed Mazur, P. K., et al. (2010). Notch2 is required for progression of pancreatic intraepithelial neoplasia and development of pancreatic ductal adenocarcinoma. Proc Natl Acad Sci USA, 107(30), 13438–13443.PubMed
209.
go back to reference Morton, J. P., et al. (2010). LKB1 haploinsufficiency cooperates with Kras to promote pancreatic cancer through suppression of p21-dependent growth arrest. Gastroenterology, 39(2), 586–597. 597 e1-6. Morton, J. P., et al. (2010). LKB1 haploinsufficiency cooperates with Kras to promote pancreatic cancer through suppression of p21-dependent growth arrest. Gastroenterology, 39(2), 586–597. 597 e1-6.
210.
go back to reference Murtaugh, L. C., et al. (2003). Notch signaling controls multiple steps of pancreatic differentiation. Proc Natl Acad Sci USA, 100(25), 14920–14925.PubMed Murtaugh, L. C., et al. (2003). Notch signaling controls multiple steps of pancreatic differentiation. Proc Natl Acad Sci USA, 100(25), 14920–14925.PubMed
211.
go back to reference Aguirre, A. J., et al. (2003). Activated Kras and Ink4a/Arf deficiency cooperate to produce metastatic pancreatic ductal adenocarcinoma. Genes & Development, 17(24), 3112–3126. Aguirre, A. J., et al. (2003). Activated Kras and Ink4a/Arf deficiency cooperate to produce metastatic pancreatic ductal adenocarcinoma. Genes & Development, 17(24), 3112–3126.
212.
go back to reference Morton, J. P., et al. (2010). Mutant p53 drives metastasis and overcomes growth arrest/senescence in pancreatic cancer. Proc Natl Acad Sci USA, 107(1), 246–251.PubMed Morton, J. P., et al. (2010). Mutant p53 drives metastasis and overcomes growth arrest/senescence in pancreatic cancer. Proc Natl Acad Sci USA, 107(1), 246–251.PubMed
213.
go back to reference Vincent, D. F., et al. (2009). Inactivation of TIF1gamma cooperates with Kras to induce cystic tumors of the pancreas. PLoS Genetics, 5(7), e1000575.PubMed Vincent, D. F., et al. (2009). Inactivation of TIF1gamma cooperates with Kras to induce cystic tumors of the pancreas. PLoS Genetics, 5(7), e1000575.PubMed
214.
go back to reference Skoulidis, F., et al. (2010). Germline Brca2 heterozygosity promotes Kras(G12D)-driven carcinogenesis in a murine model of familial pancreatic cancer. Cancer Cell, 18(5), 499–509.PubMed Skoulidis, F., et al. (2010). Germline Brca2 heterozygosity promotes Kras(G12D)-driven carcinogenesis in a murine model of familial pancreatic cancer. Cancer Cell, 18(5), 499–509.PubMed
215.
go back to reference Maniati, E., et al. (2011). Crosstalk between the canonical NF-kappaB and Notch signaling pathways inhibits Ppargamma expression and promotes pancreatic cancer progression in mice. The Journal of Clinical Investigation, 121(12), 4685–4699.PubMed Maniati, E., et al. (2011). Crosstalk between the canonical NF-kappaB and Notch signaling pathways inhibits Ppargamma expression and promotes pancreatic cancer progression in mice. The Journal of Clinical Investigation, 121(12), 4685–4699.PubMed
216.
go back to reference Hanlon, L., et al. (2010). Notch1 functions as a tumor suppressor in a model of K-ras-induced pancreatic ductal adenocarcinoma. Cancer Research, 70(11), 4280–4286.PubMed Hanlon, L., et al. (2010). Notch1 functions as a tumor suppressor in a model of K-ras-induced pancreatic ductal adenocarcinoma. Cancer Research, 70(11), 4280–4286.PubMed
217.
go back to reference De La, O. J., et al. (2008). Notch and Kras reprogram pancreatic acinar cells to ductal intraepithelial neoplasia. Proc Natl Acad Sci USA, 105(48), 18907–18912. De La, O. J., et al. (2008). Notch and Kras reprogram pancreatic acinar cells to ductal intraepithelial neoplasia. Proc Natl Acad Sci USA, 105(48), 18907–18912.
218.
go back to reference Tinder, T. L., et al. (2008). MUC1 enhances tumor progression and contributes toward immunosuppression in a mouse model of spontaneous pancreatic adenocarcinoma. Journal of Immunology, 181(5), 3116–3125. Tinder, T. L., et al. (2008). MUC1 enhances tumor progression and contributes toward immunosuppression in a mouse model of spontaneous pancreatic adenocarcinoma. Journal of Immunology, 181(5), 3116–3125.
219.
go back to reference Mukherjee, P., et al. (2009). Progression of pancreatic adenocarcinoma is significantly impeded with a combination of vaccine and COX-2 inhibition. Journal of Immunology, 182(1), 216–224. Mukherjee, P., et al. (2009). Progression of pancreatic adenocarcinoma is significantly impeded with a combination of vaccine and COX-2 inhibition. Journal of Immunology, 182(1), 216–224.
220.
go back to reference Sharpless, N. E., et al. (2001). Loss of p16Ink4a with retention of p19Arf predisposes mice to tumorigenesis. Nature, 413(6851), 86–91.PubMed Sharpless, N. E., et al. (2001). Loss of p16Ink4a with retention of p19Arf predisposes mice to tumorigenesis. Nature, 413(6851), 86–91.PubMed
221.
go back to reference Collado, M., et al. (2005). Tumour biology: Senescence in premalignant tumours. Nature, 436(7051), 642.PubMed Collado, M., et al. (2005). Tumour biology: Senescence in premalignant tumours. Nature, 436(7051), 642.PubMed
222.
go back to reference Yan, K. P., et al. (2004). Molecular cloning, genomic structure, and expression analysis of the mouse transcriptional intermediary factor 1 gamma gene. Gene, 334, 3–13.PubMed Yan, K. P., et al. (2004). Molecular cloning, genomic structure, and expression analysis of the mouse transcriptional intermediary factor 1 gamma gene. Gene, 334, 3–13.PubMed
223.
go back to reference Su, G. H., et al. (2001). ACVR1B (ALK4, activin receptor type 1B) gene mutations in pancreatic carcinoma. Proc Natl Acad Sci USA, 98(6), 3254–3257.PubMed Su, G. H., et al. (2001). ACVR1B (ALK4, activin receptor type 1B) gene mutations in pancreatic carcinoma. Proc Natl Acad Sci USA, 98(6), 3254–3257.PubMed
224.
go back to reference Gu, Z., et al. (1998). The type I activin receptor ActRIB is required for egg cylinder organization and gastrulation in the mouse. Genes & Development, 12(6), 844–857. Gu, Z., et al. (1998). The type I activin receptor ActRIB is required for egg cylinder organization and gastrulation in the mouse. Genes & Development, 12(6), 844–857.
225.
go back to reference Qiu, W., et al. (2011). Conditional activin receptor type 1B (Acvr1b) knockout mice reveal hair loss abnormality. The Journal of Investigative Dermatology, 131(5), 1067–1076.PubMed Qiu, W., et al. (2011). Conditional activin receptor type 1B (Acvr1b) knockout mice reveal hair loss abnormality. The Journal of Investigative Dermatology, 131(5), 1067–1076.PubMed
226.
go back to reference Schonleben, F., et al. (2008). PIK3CA, KRAS, and BRAF mutations in intraductal papillary mucinous neoplasm/carcinoma (IPMN/C) of the pancreas. Langenbeck's Archives of Surgery, 393(3), 289–296.PubMed Schonleben, F., et al. (2008). PIK3CA, KRAS, and BRAF mutations in intraductal papillary mucinous neoplasm/carcinoma (IPMN/C) of the pancreas. Langenbeck's Archives of Surgery, 393(3), 289–296.PubMed
227.
go back to reference Okami, K., et al. (1998). Analysis of PTEN/MMAC1 alterations in aerodigestive tract tumors. Cancer Research, 58(3), 509–511.PubMed Okami, K., et al. (1998). Analysis of PTEN/MMAC1 alterations in aerodigestive tract tumors. Cancer Research, 58(3), 509–511.PubMed
228.
go back to reference Ying, H., et al. (2011). Pten is a major tumor suppressor in pancreatic ductal adenocarcinoma and regulates an NF-kappaB-cytokine network. Cancer Discov, 1(2), 158–169.PubMed Ying, H., et al. (2011). Pten is a major tumor suppressor in pancreatic ductal adenocarcinoma and regulates an NF-kappaB-cytokine network. Cancer Discov, 1(2), 158–169.PubMed
229.
go back to reference Hill, R., et al. (2010). PTEN loss accelerates KrasG12D-induced pancreatic cancer development. Cancer Research, 70(18), 7114–7124.PubMed Hill, R., et al. (2010). PTEN loss accelerates KrasG12D-induced pancreatic cancer development. Cancer Research, 70(18), 7114–7124.PubMed
230.
go back to reference Hruban, R. H., et al. (1999). Familial pancreatic cancer. Annals of Oncology, 4, 69–73. Hruban, R. H., et al. (1999). Familial pancreatic cancer. Annals of Oncology, 4, 69–73.
231.
go back to reference Couch, F. J., et al. (2007). The prevalence of BRCA2 mutations in familial pancreatic cancer. Cancer Epidemiology, Biomarkers & Prevention, 16(2), 342–346. Couch, F. J., et al. (2007). The prevalence of BRCA2 mutations in familial pancreatic cancer. Cancer Epidemiology, Biomarkers & Prevention, 16(2), 342–346.
232.
go back to reference Su, G. H., et al. (1999). Germline and somatic mutations of the STK11/LKB1 Peutz-Jeghers gene in pancreatic and biliary cancers. American Journal of Pathology, 154, 1835–1840.PubMed Su, G. H., et al. (1999). Germline and somatic mutations of the STK11/LKB1 Peutz-Jeghers gene in pancreatic and biliary cancers. American Journal of Pathology, 154, 1835–1840.PubMed
233.
go back to reference Giardiello, F. M., et al. (2000). Very high risk of cancer in familial Peutz-Jeghers syndrome. Gastroenterology, 119(6), 1447–1453.PubMed Giardiello, F. M., et al. (2000). Very high risk of cancer in familial Peutz-Jeghers syndrome. Gastroenterology, 119(6), 1447–1453.PubMed
234.
go back to reference Ylikorkala, A., et al. (2001). Vascular abnormalities and deregulation of VEGF in Lkb1-deficient mice. Science, 293(5533), 1323–1326.PubMed Ylikorkala, A., et al. (2001). Vascular abnormalities and deregulation of VEGF in Lkb1-deficient mice. Science, 293(5533), 1323–1326.PubMed
235.
go back to reference Grote, V.A., et al., The Associations of advanced glycation end products and its soluble receptor with pancreatic cancer risk: A case–control study within the prospective EPIC cohort. Cancer Epidemiol Biomarkers Prev, 2012;21:619-628. Grote, V.A., et al., The Associations of advanced glycation end products and its soluble receptor with pancreatic cancer risk: A case–control study within the prospective EPIC cohort. Cancer Epidemiol Biomarkers Prev, 2012;21:619-628.
236.
go back to reference DiNorcia, J., et al. (2012). RAGE gene deletion inhibits the development and progression of ductal neoplasia and prolongs survival in a murine model of pancreatic cancer. Journal of Gastrointestinal Surgery, 16(1), 104–112. discussion 112.PubMed DiNorcia, J., et al. (2012). RAGE gene deletion inhibits the development and progression of ductal neoplasia and prolongs survival in a murine model of pancreatic cancer. Journal of Gastrointestinal Surgery, 16(1), 104–112. discussion 112.PubMed
237.
go back to reference Meylan, E., et al. (2009). Requirement for NF-kappaB signalling in a mouse model of lung adenocarcinoma. Nature, 462(7269), 104–107.PubMed Meylan, E., et al. (2009). Requirement for NF-kappaB signalling in a mouse model of lung adenocarcinoma. Nature, 462(7269), 104–107.PubMed
238.
go back to reference Fendrich, V., et al. (2010). The angiotensin-I-converting enzyme inhibitor enalapril and aspirin delay progression of pancreatic intraepithelial neoplasia and cancer formation in a genetically engineered mouse model of pancreatic cancer. Gut, 59(5), 630–637.PubMed Fendrich, V., et al. (2010). The angiotensin-I-converting enzyme inhibitor enalapril and aspirin delay progression of pancreatic intraepithelial neoplasia and cancer formation in a genetically engineered mouse model of pancreatic cancer. Gut, 59(5), 630–637.PubMed
239.
go back to reference Ling, J., et al. (2012). KrasG12D-induced IKK2/beta/NF-kappaB activation by IL-1alpha and p62 feedforward loops is required for development of pancreatic ductal adenocarcinoma. Cancer Cell, 21(1), 105–120.PubMed Ling, J., et al. (2012). KrasG12D-induced IKK2/beta/NF-kappaB activation by IL-1alpha and p62 feedforward loops is required for development of pancreatic ductal adenocarcinoma. Cancer Cell, 21(1), 105–120.PubMed
240.
go back to reference Gendler, S. J., & Spicer, A. P. (1995). Epithelial mucin genes. Annual Review of Physiology, 57, 607–634.PubMed Gendler, S. J., & Spicer, A. P. (1995). Epithelial mucin genes. Annual Review of Physiology, 57, 607–634.PubMed
241.
go back to reference Rowse, G. J., et al. (1998). Tolerance and immunity to MUC1 in a human MUC1 transgenic murine model. Cancer Research, 58(2), 315–321.PubMed Rowse, G. J., et al. (1998). Tolerance and immunity to MUC1 in a human MUC1 transgenic murine model. Cancer Research, 58(2), 315–321.PubMed
242.
go back to reference Whipple, C.A., A.L. Young, and M. Korc, A Kras(G12D)-driven genetic mouse model of pancreatic cancer requires glypican-1 for efficient proliferation and angiogenesis. Oncogene, 2012;31:2535-2544. Whipple, C.A., A.L. Young, and M. Korc, A Kras(G12D)-driven genetic mouse model of pancreatic cancer requires glypican-1 for efficient proliferation and angiogenesis. Oncogene, 2012;31:2535-2544.
243.
go back to reference Aikawa, T., et al. (2008). Glypican-1 modulates the angiogenic and metastatic potential of human and mouse cancer cells. The Journal of Clinical Investigation, 118(1), 89–99.PubMed Aikawa, T., et al. (2008). Glypican-1 modulates the angiogenic and metastatic potential of human and mouse cancer cells. The Journal of Clinical Investigation, 118(1), 89–99.PubMed
244.
go back to reference Agbunag, C., & Bar-Sagi, D. (2004). Oncogenic K-ras drives cell cycle progression and phenotypic conversion of primary pancreatic duct epithelial cells. Cancer Research, 64(16), 5659–5663.PubMed Agbunag, C., & Bar-Sagi, D. (2004). Oncogenic K-ras drives cell cycle progression and phenotypic conversion of primary pancreatic duct epithelial cells. Cancer Research, 64(16), 5659–5663.PubMed
245.
go back to reference Pasca di Magliano, M. (2007). Common activation of canonical Wnt signaling in pancreatic adenocarcinoma. PLoS One, 2(11), e1155.PubMed Pasca di Magliano, M. (2007). Common activation of canonical Wnt signaling in pancreatic adenocarcinoma. PLoS One, 2(11), e1155.PubMed
246.
go back to reference Morris, J. P., et al. (2010). Beta-catenin blocks Kras-dependent reprogramming of acini into pancreatic cancer precursor lesions in mice. The Journal of Clinical Investigation, 120(2), 508–520.PubMed Morris, J. P., et al. (2010). Beta-catenin blocks Kras-dependent reprogramming of acini into pancreatic cancer precursor lesions in mice. The Journal of Clinical Investigation, 120(2), 508–520.PubMed
247.
go back to reference Heiser, P. W., et al. (2008). Stabilization of beta-catenin induces pancreas tumor formation. Gastroenterology, 135(4), 1288–1300.PubMed Heiser, P. W., et al. (2008). Stabilization of beta-catenin induces pancreas tumor formation. Gastroenterology, 135(4), 1288–1300.PubMed
248.
go back to reference Feldmann, G., et al. (2008). Hedgehog inhibition prolongs survival in a genetically engineered mouse model of pancreatic cancer. Gut, 57(10), 1420–1430.PubMed Feldmann, G., et al. (2008). Hedgehog inhibition prolongs survival in a genetically engineered mouse model of pancreatic cancer. Gut, 57(10), 1420–1430.PubMed
249.
go back to reference Plentz, R., et al. (2009). Inhibition of gamma-secretase activity inhibits tumor progression in a mouse model of pancreatic ductal adenocarcinoma. Gastroenterology, 136(5), 1741–1749. e6.PubMed Plentz, R., et al. (2009). Inhibition of gamma-secretase activity inhibits tumor progression in a mouse model of pancreatic ductal adenocarcinoma. Gastroenterology, 136(5), 1741–1749. e6.PubMed
250.
go back to reference Funahashi, H., et al. (2007). Delayed progression of pancreatic intraepithelial neoplasia in a conditional Kras(G12D) mouse model by a selective cyclooxygenase-2 inhibitor. Cancer Research, 67(15), 7068–7071.PubMed Funahashi, H., et al. (2007). Delayed progression of pancreatic intraepithelial neoplasia in a conditional Kras(G12D) mouse model by a selective cyclooxygenase-2 inhibitor. Cancer Research, 67(15), 7068–7071.PubMed
251.
go back to reference Ijichi, H., et al. (2011). Inhibiting Cxcr2 disrupts tumor-stromal interactions and improves survival in a mouse model of pancreatic ductal adenocarcinoma. The Journal of Clinical Investigation, 121(10), 4106–4117.PubMed Ijichi, H., et al. (2011). Inhibiting Cxcr2 disrupts tumor-stromal interactions and improves survival in a mouse model of pancreatic ductal adenocarcinoma. The Journal of Clinical Investigation, 121(10), 4106–4117.PubMed
252.
go back to reference Diep, C. H., et al. (2011). Synergistic effect between erlotinib and MEK inhibitors in KRAS wild-type human pancreatic cancer cells. Clinical Cancer Research, 17(9), 2744–2756.PubMed Diep, C. H., et al. (2011). Synergistic effect between erlotinib and MEK inhibitors in KRAS wild-type human pancreatic cancer cells. Clinical Cancer Research, 17(9), 2744–2756.PubMed
253.
go back to reference Branda, C. S., & Dymecki, S. M. (2004). Talking about a revolution: The impact of site-specific recombinases on genetic analyses in mice. Developmental Cell, 6(1), 7–28.PubMed Branda, C. S., & Dymecki, S. M. (2004). Talking about a revolution: The impact of site-specific recombinases on genetic analyses in mice. Developmental Cell, 6(1), 7–28.PubMed
254.
go back to reference Zheng, B., et al. (2000). Engineering mouse chromosomes with Cre-loxP: Range, efficiency, and somatic applications. Molecular and Cellular Biology, 20(2), 648–655.PubMed Zheng, B., et al. (2000). Engineering mouse chromosomes with Cre-loxP: Range, efficiency, and somatic applications. Molecular and Cellular Biology, 20(2), 648–655.PubMed
255.
go back to reference Brayton, C. F., Treuting, P. M., & Ward, J. M. (2012). Pathobiology of aging mice and GEM: Background strains and experimental design. Veterinary Pathology, 49(1), 85–105.PubMed Brayton, C. F., Treuting, P. M., & Ward, J. M. (2012). Pathobiology of aging mice and GEM: Background strains and experimental design. Veterinary Pathology, 49(1), 85–105.PubMed
256.
go back to reference Jorgenson, T. C., et al. (2010). Identification of susceptibility loci in a mouse model of KRASG12D-driven pancreatic cancer. Cancer Research, 70(21), 8398–8406.PubMed Jorgenson, T. C., et al. (2010). Identification of susceptibility loci in a mouse model of KRASG12D-driven pancreatic cancer. Cancer Research, 70(21), 8398–8406.PubMed
257.
go back to reference Yu, R., et al. (2011). Pancreatic neuroendocrine tumors in glucagon receptor-deficient mice. PLoS One, 6(8), e23397.PubMed Yu, R., et al. (2011). Pancreatic neuroendocrine tumors in glucagon receptor-deficient mice. PLoS One, 6(8), e23397.PubMed
258.
go back to reference Rhim, A. D., et al. (2012). EMT and dissemination precede pancreatic tumor formation. Cell, 148(1–2), 349–361.PubMed Rhim, A. D., et al. (2012). EMT and dissemination precede pancreatic tumor formation. Cell, 148(1–2), 349–361.PubMed
259.
go back to reference Collins, M. A., et al. (2012). Oncogenic Kras is required for both the initiation and maintenance of pancreatic cancer in mice. The Journal of Clinical Investigation, 122(2), 639–653.PubMed Collins, M. A., et al. (2012). Oncogenic Kras is required for both the initiation and maintenance of pancreatic cancer in mice. The Journal of Clinical Investigation, 122(2), 639–653.PubMed
260.
go back to reference Wang, Y., et al. (2011). Restoring expression of wild-type p53 suppresses tumor growth but does not cause tumor regression in mice with a p53 missense mutation. The Journal of Clinical Investigation, 121(3), 893–904.PubMed Wang, Y., et al. (2011). Restoring expression of wild-type p53 suppresses tumor growth but does not cause tumor regression in mice with a p53 missense mutation. The Journal of Clinical Investigation, 121(3), 893–904.PubMed
261.
go back to reference Ventura, A., et al. (2007). Restoration of p53 function leads to tumour regression in vivo. Nature, 445(7128), 661–665.PubMed Ventura, A., et al. (2007). Restoration of p53 function leads to tumour regression in vivo. Nature, 445(7128), 661–665.PubMed
Metadata
Title
Challenges and advances in mouse modeling for human pancreatic tumorigenesis and metastasis
Authors
Wanglong Qiu
Gloria H. Su
Publication date
01-06-2013
Publisher
Springer US
Published in
Cancer and Metastasis Reviews / Issue 1-2/2013
Print ISSN: 0167-7659
Electronic ISSN: 1573-7233
DOI
https://doi.org/10.1007/s10555-012-9408-2

Other articles of this Issue 1-2/2013

Cancer and Metastasis Reviews 1-2/2013 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