Skip to main content Accessibility help
×
Hostname: page-component-848d4c4894-4hhp2 Total loading time: 0 Render date: 2024-05-28T18:51:06.120Z Has data issue: false hasContentIssue false

48 - Pancreatic cancer

from Part 3.1 - Molecular pathology: carcinomas

Published online by Cambridge University Press:  05 February 2015

Siong-Seng Liau
Affiliation:
Hepatopancreatobiliary Surgery Unit, Department of Surgery, Addenbrooke’s Hospital, and Medical Research Council Cancer Cell Unit, Hutchison-MRC Research Center, University of Cambridge, Cambridge, UK
David A. Tuveson
Affiliation:
Department of Oncology, Addenbrooke’s Hospital, University of Cambridge, Cambridge, UK
Edward P. Gelmann
Affiliation:
Columbia University, New York
Charles L. Sawyers
Affiliation:
Memorial Sloan-Kettering Cancer Center, New York
Frank J. Rauscher, III
Affiliation:
The Wistar Institute Cancer Centre, Philadelphia
Get access

Summary

Introduction

Pancreatic cancer is usually fatal. Various factors have been ascribed to this grim reality, including the lack of early disease detection methods and the poor response of pancreatic patients to therapies (1,2). Surgery is the only curative therapy (Figure 48.1), but only a small minority of patients qualify for surgery (10–20%) and most of these patients still succumb to local or metastatic recurrence. In advanced disease, chemotherapy remains the only option, however few agents are approved and they show only a modest survival benefit. To make progress against this cancer, a better understanding at the molecular and tissue level is urgently required, which should serve as a “roadmap” to identify better therapeutic agents.

Histopathology of pancreatic adenocarcinoma

Pancreatic ductal adenocarcinoma (PDA) is the most common exocrine pancreatic tumor, accounting for 85% of all malignant pancreatic tumors. Histologically, PDA is a malignant epithelial neoplasm which demonstrates gland formation (3). A characteristic feature of PDA is the intense peritumoural desmoplastic reaction (Figure 48.1). PDA evolves through a multi-step process with cells gaining cumulative genetic and phenotypic alterations resulting in pathological progression from normal pancreas to pre-invasive pre-neoplasms and invasive cancer (4; Figure 48.2). These pre-neoplasms include pancreatic intra-epithelial neoplasms (PanINs), intra-ductal papillary mucinous neoplasms (IPMNs), and mucinous cystic neoplasms (MCNs). Compelling evidence supports the notion that PDA develops from pancreatic pre-neoplasms, similar to the proposed adenoma–carcinoma sequence in colorectal adenocarcinoma (5).

Type
Chapter
Information
Molecular Oncology
Causes of Cancer and Targets for Treatment
, pp. 557 - 568
Publisher: Cambridge University Press
Print publication year: 2013

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Bramhall, SR, Allum, WH, Jones, AG, et al. Treatment and survival in 13,560 patients with pancreatic cancer, and incidence of the disease, in the West Midlands: an epidemiological study. British Journal of Surgery 1995;82:111–15.CrossRefGoogle ScholarPubMed
Hedberg, M, Borgstrom, A, Genell, S, Janzon, L. Survival following pancreatic carcinoma: a follow-up study of all cases recorded in Malmo, Sweden, 1977–1991. British Journal of Surgery 1998;85:1641–4.CrossRefGoogle Scholar
Hruban, R, Klimstra, D, Pitman, M. Tumors of the Pancreas, 4th edn. Washington: Armed Forces Institute of Pathology; 2006.
Hruban, RH, Goggins, M, Parsons, J, Kern, SE. Progression model for pancreatic cancer. Clinical Cancer Research 2000;6:2969–72.
Vogelstein, B, Fearon, ER, Hamilton, SR, et al. Genetic alterations during colorectal-tumor development. New England Journal of Medicine 1988;319:525–32.CrossRefGoogle ScholarPubMed
Jaffee, EM, Hruban, RH, Canto, M, Kern, SE. Focus on pancreas cancer. Cancer Cell 2002;2:25–8.CrossRef
Wilentz, RE, Iacobuzio-Donahue, CA, Argani, P, et al. Loss of expression of Dpc4 in pancreatic intraepithelial neoplasia: evidence that DPC4 inactivation occurs late in neoplastic progression. Cancer Research 2000;60:2002–6.
Satoh, K, Shimosegawa, T, Moriizumi, S, Koizumi, M, Toyota, T. K-ras mutation and p53 protein accumulation in intraductal mucin-hypersecreting neoplasms of the pancreas. Pancreas 1996;12:362–8.CrossRef
Furukawa, T, Fujisaki, R, Yoshida, Y, et al. Distinct progression pathways involving the dysfunction of DUSP6/MKP-3 in pancreatic intraepithelial neoplasia and intraductal papillary-mucinous neoplasms of the pancreas. Modern Pathology 2005;18:1034–42.CrossRef
Iacobuzio-Donahue, CA, Klimstra, DS, Adsay, NV, et al. Dpc-4 protein is expressed in virtually all human intraductal papillary mucinous neoplasms of the pancreas: comparison with conventional ductal adenocarcinomas. American Journal of Pathology 2000;157:755–61.CrossRefGoogle ScholarPubMed
Sahin, F, Maitra, A, Argani, P, et al. Loss of Stk11/Lkb1 expression in pancreatic and biliary neoplasms. Modern Pathology 2003;16:686–91.CrossRef
Jimenez, RE, Warshaw, AL, Z’Graggen, K, et al. Sequential accumulation of K-ras mutations and p53 over-expression in the progression of pancreatic mucinous cystic neoplasms to malignancy. Annals of Surgery 1999;230:501–9; discussion 509–11.
Iacobuzio-Donahue, CA, Wilentz, RE, Argani, P, et al. Dpc4 protein in mucinous cystic neoplasms of the pancreas: frequent loss of expression in invasive carcinomas suggests a role in genetic progression. American Journal of Surgical Pathology 2000;24:1544–8.CrossRefGoogle ScholarPubMed
Malumbres, M, Barbacid, M. RAS oncogenes: the first 30 years. Nature Reviews Cancer 2003;3:459–65.CrossRef
Goldstein, AM, Fraser, MC, Struewing, JP, et al. Increased risk of pancreatic cancer in melanoma-prone kindreds with p16INK4 mutations. New England Journal of Medicine 1995;333:970–4.CrossRefGoogle ScholarPubMed
Yan, L, McFaul, C, Howes, N, et al. Molecular analysis to detect pancreatic ductal adenocarcinoma in high-risk groups. Gastroenterology 2005;128:2124–30.CrossRef
Feldmann, G, Beaty, R, Hruban, RH, Maitra, A. Molecular genetics of pancreatic intraepithelial neoplasia. Journal of Hepatobiliary and Pancreatic Surgery 2007;14:224–32.CrossRefGoogle ScholarPubMed
Campbell, PJ, Yachida, S, Mudie, LJ, et al. The patterns and dynamics of genomic instability in metastatic pancreatic cancer. Nature 2010;467:1109–13.CrossRef
Yachida, S, Jones, S, Bozic, I, et al. Distant metastasis occurs late during the genetic evolution of pancreatic cancer. Nature 2010;467:1114–17.CrossRef
Goggins, M, Hruban, RH, Kern, SE. BRCA2 is inactivated late in the development of pancreatic intraepithelial neoplasia: evidence and implications. American Journal of Pathology 2000;156:1767–71.CrossRefGoogle ScholarPubMed
Jones, S, Hruban, RH, Kamiyama, M, et al. Exomic sequencing identifies PALB2 as a pancreatic cancer susceptibility gene. Science 2009;324:217.CrossRef
Jones, S, Zhang, X, Parsons, DW, et al. Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science 2008;321:1801–6.CrossRef
Tan, AC, Jimeno, A, Lin, SH, et al. Characterizing DNA methylation patterns in pancreatic cancer genome. Molecular Oncology 2009;3:425–38.CrossRef
Bloomston, M, Frankel, WL, Petrocca, F, et al. MicroRNA expression patterns to differentiate pancreatic adenocarcinoma from normal pancreas and chronic pancreatitis. Journal of the American Medical Association 2007;297:1901–8.CrossRefGoogle ScholarPubMed
Szafranska, AE, Davison, TS, John, J, et al. MicroRNA expression alterations are linked to tumorigenesis and non-neoplastic processes in pancreatic ductal adenocarcinoma. Oncogene 2007;26:4442–52.CrossRef
Lee, EJ, Gusev, Y, Jiang, J, et al. Expression profiling identifies microRNA signature in pancreatic cancer. International Journal of Cancer 2007;120:1046–54.CrossRefGoogle ScholarPubMed
van Heek, NT, Meeker, AK, Kern, SE, et al. Telomere shortening is nearly universal in pancreatic intraepithelial neoplasia. American Journal of Pathology 2002;161:1541–7.CrossRefGoogle ScholarPubMed
Artandi, SE, Chang, S, Lee, SL, et al. Telomere dysfunction promotes non-reciprocal translocations and epithelial cancers in mice. Nature 2000;406:641–5.CrossRef
Berman, DM, Karhadkar, SS, Maitra, A, et al. Widespread requirement for Hedgehog ligand stimulation in growth of digestive tract tumours. Nature 2003;425:846–51.CrossRef
Thayer, SP, di Magliano, MP, Heiser, PW, et al. Hedgehog is an early and late mediator of pancreatic cancer tumorigenesis. Nature 2003;425:851–6.CrossRef
Radtke, F, Raj, K. The role of Notch in tumorigenesis: oncogene or tumour suppressor? Nature Reviews Cancer 2003;3:756–67.
Friedmann, DR, Wilson, JJ, Kovall, RA. RAM-induced allostery facilitates assembly of a notch pathway active transcription complex. Journal of Biological Chemistry 2008;283:14 781–91.CrossRefGoogle ScholarPubMed
Hruban, RH, Adsay, NV, Albores-Saavedra, J, et al. Pathology of genetically engineered mouse models of pancreatic exocrine cancer: consensus report and recommendations. Cancer Research 2006;66:95–106.CrossRef
Hruban, RH, Rustgi, AK, Brentnall, TA, et al. Pancreatic cancer in mice and man: the Penn Workshop 2004. Cancer Research 2006;66:14–17.CrossRef
Hingorani, SR, Petricoin, EF, Maitra, A, et al. Preinvasive and invasive ductal pancreatic cancer and its early detection in the mouse. Cancer Cell 2003;4:437–50.CrossRef
Tuveson, DA, Shaw, AT, Willis, NA, et al. Endogenous oncogenic K-ras(G12D) stimulates proliferation and widespread neoplastic and developmental defects. Cancer Cell 2004;5:375–87.CrossRef
Olive, KP, Tuveson, DA, Ruhe, ZC, et al. Mutant p53 gain of function in two mouse models of Li-Fraumeni syndrome. Cell 2004;119:847–60.CrossRef
Hingorani, SR, Wang, L, Multani, AS, et al. Trp53R172H and KrasG12D cooperate to promote chromosomal instability and widely metastatic pancreatic ductal adenocarcinoma in mice. Cancer Cell 2005;7:469–83.CrossRef
Bardeesy, N, Cheng, KH, Berger, JH, et al. Smad4 is dispensable for normal pancreas development yet critical in progression and tumor biology of pancreas cancer. Genes and Development 2006;20:3130–46.CrossRef
Ijichi, H, Chytil, A, Gorska, AE, et al. Aggressive pancreatic ductal adenocarcinoma in mice caused by pancreas-specific blockade of transforming growth factor-beta signaling in cooperation with active Kras expression. Genes and Development 2006;20:3147–60.CrossRef
Hill, R, Calvopina, JH, Kim, C, et al. PTEN loss accelerates KrasG12D-induced pancreatic cancer development. Cancer Research 2010;70:7114–24.CrossRef
Kojima, K, Vickers, SM, Adsay, NV, et al. Inactivation of Smad4 accelerates Kras(G12D)-mediated pancreatic neoplasia. Cancer Research 2007;67:8121–30.CrossRef
Olive, KP, Jacobetz, MA, Davidson, CJ, et al. Inhibition of Hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer. Science 2009;324:1457–61.CrossRef
Burris, HA 3rd, Moore, MJ, Andersen, J, et al. Improvements in survival and clinical benefit with gemcitabine as first-line therapy for patients with advanced pancreas cancer: a randomized trial. Journal of Clinical Oncology 1997;15:2403–13.CrossRefGoogle ScholarPubMed
Neoptolemos, JP, Stocken, DD, Bassi, C, et al. Adjuvant chemotherapy with fluorouracil plus folinic acid vs gemcitabine following pancreatic cancer resection: a randomized controlled trial. Journal of the American Medical Association 2010;304:1073–81.CrossRefGoogle ScholarPubMed
Korc, M, Chandrasekar, B, Yamanaka, Y, et al. Overexpression of the epidermal growth factor receptor in human pancreatic cancer is associated with concomitant increases in the levels of epidermal growth factor and transforming growth factor alpha. Journal of Clinical Investigation 1992;90:1352–60.CrossRefGoogle ScholarPubMed
Moore, MJ, Goldstein, D, Hamm, J, et al. Erlotinib plus gemcitabine compared to gemcitabine alone in patients with advanced pancreatic cancer: a Phase III trial of the National Cancer Institute of Canada Clinical Trials Group [NCIC-CTG]. Journal of Clinical Oncology (Meeting Abstracts) 2005;23:1.Google Scholar
Whyte, DB, Kirschmeier, P, Hockenberry, TN, et al. K- and N-Ras are geranylgeranylated in cells treated with farnesyl protein transferase inhibitors. Journal of Biological Chemistry 1997;272:14 459–64.CrossRefGoogle ScholarPubMed
Yang, JC, Haworth, L, Sherry, RM, et al. A randomized trial of bevacizumab, an anti-vascular endothelial growth factor antibody, for metastatic renal cancer. New England Journal of Medicine 2003;349:427–34.CrossRefGoogle ScholarPubMed
Baker, CH, Solorzano, CC, Fidler, IJ. Blockade of vascular endothelial growth factor receptor and epidermal growth factor receptor signaling for therapy of metastatic human pancreatic cancer. Cancer Research 2002;62:1996–2003.
Bockhorn, M, Tsuzuki, Y, Xu, L, et al. Differential vascular and transcriptional responses to anti-vascular endothelial growth factor antibody in orthotopic human pancreatic cancer xenografts. Clinical Cancer Research 2003;9:4221–6.
Tsuzuki, Y, Mouta Carreira, C, Bockhorn, M, et al. Pancreas microenvironment promotes VEGF expression and tumor growth: novel window models for pancreatic tumor angiogenesis and microcirculation. Laboratory Investigation 2001;81:1439–51.CrossRef
Kindler, HL, Niedzwiecki, D, Hollis, D, et al. Gemcitabine plus bevacizumab compared with gemcitabine plus placebo in patients with advanced pancreatic cancer: Phase III trial of the Cancer and Leukemia Group B (CALGB 80303). Journal of Clinical Oncology; 28:3617–22.CrossRef
Kindler, HL, Wroblewski, K, Wallace, JA, et al. Gemcitabine plus sorafenib in patients with advanced pancreatic cancer: a Phase II trial of the University of Chicago Phase II Consortium. Investigational New Drugs2012;30:382–6.
Sakurada, A, Suzuki, A, Sato, M, et al. Infrequent genetic alterations of the PTEN/MMAC1 gene in Japanese patients with primary cancers of the breast, lung, pancreas, kidney, and ovary. Japanese Journal of Cancer Research 1997;88:1025–8.CrossRefGoogle ScholarPubMed
Schlieman, MG, Fahy, BN, Ramsamooj, R, Beckett, L, Bold, RJ. Incidence, mechanism and prognostic value of activated AKT in pancreas cancer. British Journal of Cancer 2003;89:2110–15.CrossRefGoogle ScholarPubMed
Yamamoto, S, Tomita, Y, Hoshida, Y, et al. Prognostic significance of activated Akt expression in pancreatic ductal adenocarcinoma. Clinical Cancer Research 2004;10:2846–50.CrossRef
Bramhall, SR, Schulz, J, Nemunaitis, J, et al. A double-blind placebo-controlled, randomised study comparing gemcitabine and marimastat with gemcitabine and placebo as first line therapy in patients with advanced pancreatic cancer. British Journal of Cancer 2002;87:161–7.CrossRefGoogle ScholarPubMed
Coussens, LM, Fingleton, B, Matrisian, LM. Matrix metalloproteinase inhibitors and cancer: trials and tribulations. Science 2002;295:2387–92.CrossRef
Lynch, HT, Deters, CA, Lynch, JF, Brand, RE. Familial pancreatic carcinoma in Jews. Familial Cancer 2004;3:233–40.CrossRef
Ozcelik, H, Schmocker, B, Di Nicola, N, et al. Germline BRCA2 6174delT mutations in Ashkenazi Jewish pancreatic cancer patients. Nature Genetics 1997;16:17–18.CrossRef
Goggins, M, Schutte, M, Lu, J, et al. Germline BRCA2 gene mutations in patients with apparently sporadic pancreatic carcinomas. Cancer Research 1996;56:5360–4.
Lynch, HT, Deters, CA, Snyder, CL, et al. BRCA1 and pancreatic cancer: pedigree findings and their causal relationships. Cancer Genetics and Cytogenetics 2005;158:119–25.CrossRef
Hofstatter, EW, Domchek, SM, Miron, A, et al. PALB2 mutations in familial breast and pancreatic cancer. Familial Cancer 2011;10:225–31.CrossRef
Bartsch, DK, Sina-Frey, M, Lang, S, et al. CDKN2A germline mutations in familial pancreatic cancer. Annals of Surgery 2002;236:730–7.CrossRef
Parker, JF, Florell, SR, Alexander, A, et al. Pancreatic carcinoma surveillance in patients with familial melanoma. Archives of Dermatology 2003;139:1019–25.CrossRef
Lowenfels, AB, Maisonneuve, P, DiMagno, EP, et al. Hereditary pancreatitis and the risk of pancreatic cancer. International Hereditary Pancreatitis Study Group. Journal of the National Cancer Institute 1997;89:442–6.CrossRefGoogle ScholarPubMed
Giardiello, FM, Offerhaus, GJ, Lee, DH, et al. Increased risk of thyroid and pancreatic carcinoma in familial adenomatous polyposis. Gut 1993;34:1394–6.CrossRef
Giardiello, FM, Brensinger, JD, Tersmette, AC, et al. Very high risk of cancer in familial Peutz-Jeghers syndrome. Gastroenterology 2000;119:1447–53.CrossRef
Su, GH, Hruban, RH, Bansal, RK, et al. Germline and somatic mutations of the STK11/LKB1 Peutz-Jeghers gene in pancreatic and biliary cancers. American Journal of Pathology 1999;154:1835–40.CrossRefGoogle ScholarPubMed
Varley, JM. Germline TP53 mutations and Li-Fraumeni syndrome. Human Mutation 2003;21:313–20.CrossRef
Kleihues, P, Schauble, B, zur Hausen, A, Esteve, J, Ohgaki, H. Tumors associated with p53 germline mutations: a synopsis of 91 families. American Journal of Pathology 1997;150:1–13.Google ScholarPubMed
Goggins, M, Offerhaus, GJ, Hilgers, W, et al. Pancreatic adenocarcinomas with DNA replication errors (RER+) are associated with wild-type K-ras and characteristic histopathology: poor differentiation, a syncytial growth pattern, and pushing borders suggest RER+. American Journal of Pathology 1998;152:1501–7.Google ScholarPubMed
Yamamoto, H, Itoh, F, Nakamura, H, et al. Genetic and clinical features of human pancreatic ductal adenocarcinomas with widespread microsatellite instability. Cancer Research 2001;61:3139–44.
Couch, FJ, Johnson, MR, Rabe, K, et al. Germ line Fanconi anemia complementation group C mutations and pancreatic cancer. Cancer Research 2005;65:383–6.
van der Heijden, MS, Yeo, CJ, Hruban, RH, Kern, SE. Fanconi anemia gene mutations in young-onset pancreatic cancer. Cancer Research 2003;63:2585–8.
Almoguera, C, Shibata, D, Forrester, K, et al. Most human carcinomas of the exocrine pancreas contain mutant c-K-ras genes. Cell 1988;53:549–54.CrossRef
Liau, SS, Rocha, F, Matros, E, Redston, M, Whang, E. High mobility group AT-hook 1 (HMGA1) is an independent prognostic factor and novel therapeutic target in pancreatic adenocarcinoma. Cancer 2008;113:302–14.CrossRef
Hristov, AC, Cope, L, Di Cello, F, et al. M. HMGA1 correlates with advanced tumor grade and decreased survival in pancreatic ductal adenocarcinoma. Modern Pathology 2010;23:98–104.CrossRef
Wei, D, Le, X, Zheng, L, et al. Stat3 activation regulates the expression of vascular endothelial growth factor and human pancreatic cancer angiogenesis and metastasis. Oncogene 2003;22:319–29.CrossRef
Scholz, A, Heinze, S, Detjen, KM, et al. Activated signal transducer and activator of transcription 3 (STAT3) supports the malignant phenotype of human pancreatic cancer. Gastroenterology 2003;125:891–905.CrossRef
Thomas, RM, Toney, K, Fenoglio-Preiser, C, et al. The RON receptor tyrosine kinase mediates oncogenic phenotypes in pancreatic cancer cells and is increasingly expressed during pancreatic cancer progression. Cancer Research 2007;67:6075–82.CrossRef
Seo, Y, Baba, H, Fukuda, T, Takashima, M, Sugimachi, K. High expression of vascular endothelial growth factor is associated with liver metastasis and a poor prognosis for patients with ductal pancreatic adenocarcinoma. Cancer 2000;88:2239–45.3.0.CO;2-V>CrossRef
Furukawa, T, Duguid, WP, Kobari, M, Matsuno, S, Tsao, MS. Hepatocyte growth factor and Met receptor expression in human pancreatic carcinogenesis. American Journal of Pathology 1995;147:889–95.Google ScholarPubMed
Doucas, H, Mann, CD, Sutton, CD, et al. Expression of nuclear Notch3 in pancreatic adenocarcinomas is associated with adverse clinical features, and correlates with the expression of STAT3 and phosphorylated Akt. Journal of Surgical Oncology 2008;97:63–8.CrossRefGoogle ScholarPubMed
Hakam, A, Fang, Q, Karl, R, Coppola, D. Coexpression of IGF-1R and c-Src proteins in human pancreatic ductal adenocarcinoma. Digestive Diseases and Sciences 2003;48:1972–8.CrossRef
Bloomston, M, Bhardwaj, A, Ellison, EC, Frankel, WL. Epidermal growth factor receptor expression in pancreatic carcinoma using tissue microarray technique. Digestive Surgery 2006;23:74–9.CrossRef
Wang, W, Abbruzzese, JL, Evans, DB, et al. The nuclear factor-kappa B RelA transcription factor is constitutively activated in human pancreatic adenocarcinoma cells. Clinical Cancer Research 1999;5:119–27.
Sclabas, GM, Fujioka, S, Schmidt, C, Evans, DB, Chiao, PJ. NF-kappaB in pancreatic cancer. International Journal of Gastrointestinal Cancer 2003;33:15–26.CrossRefGoogle ScholarPubMed
Zeng, G, Germinaro, M, Micsenyi, A, et al. Aberrant Wnt/beta-catenin signaling in pancreatic adenocarcinoma. Neoplasia 2006;8:279–89.CrossRef
Furuyama, K, Doi, R, Mori, T, et al. Clinical significance of focal adhesion kinase in resectable pancreatic cancer. World Journal Surgery 2006;30:219–26.CrossRef
Safran, H, Steinhoff, M, Mangray, S, et al. Overexpression of the HER-2/neu oncogene in pancreatic adenocarcinoma. American Journal of Clinical Oncology 2001;24:496–9.CrossRefGoogle ScholarPubMed
Dugan, MC, Dergham, ST, Kucway, R, et al. HER-2/neu expression in pancreatic adenocarcinoma: relation to tumor differentiation and survival. Pancreas 1997;14:229–36.CrossRef
Ruggeri, BA, Huang, L, Wood, M, Cheng, JQ, Testa, JR. Amplification and overexpression of the AKT2 oncogene in a subset of human pancreatic ductal adenocarcinomas. Molecular Carcinogenesis 1998;21:81–6.3.0.CO;2-R>CrossRef
Hu, YX, Watanabe, H, Ohtsubo, K, et al. Frequent loss of p16 expression and its correlation with clinicopathological parameters in pancreatic carcinoma. Clinical Cancer Research 1997;3:1473–7.
Wilentz, RE, Geradts, J, Maynard, R, et al. Inactivation of the p16 (INK4A) tumor-suppressor gene in pancreatic duct lesions: loss of intranuclear expression. Cancer Research 1998;58:4740–4.
Rozenblum, E, Schutte, M, Goggins, M, et al. Tumor-suppressive pathways in pancreatic carcinoma. Cancer Research 1997;57:1731–4.
Zhang, SY, Ruggeri, B, Agarwal, P, et al. Immunohistochemical analysis of p53 expression in human pancreatic carcinomas. Archives of Pathology and Laboratory Medicine 1994;118:150–4.
Goggins, M, Shekher, M, Turnacioglu, K, et al. Genetic alterations of the transforming growth factor beta receptor genes in pancreatic and biliary adenocarcinomas. Cancer Research 1998;58:5329–32.
Hiyama, E, Kodama, T, Shinbara, K, et al. Telomerase activity is detected in pancreatic cancer but not in benign tumors. Cancer Research 1997;57:326–31.
Tucker, ON, Dannenberg, AJ, Yang, EK, et al. Cyclooxygenase-2 expression is up-regulated in human pancreatic cancer. Cancer Research 1999;59:987–90.
Moore, MJ, Hamm, J, Dancey, J, et al. Comparison of gemcitabine versus the matrix metalloproteinase inhibitor BAY 12–9566 in patients with advanced or metastatic adenocarcinoma of the pancreas: a Phase III trial of the National Cancer Institute of Canada Clinical Trials Group. Journal of Clinical Oncology 2003;21:3296–302.CrossRefGoogle ScholarPubMed
Van Cutsem, E, van de Velde, H, Karasek, P, et al. Phase III trial of gemcitabine plus tipifarnib compared with gemcitabine plus placebo in advanced pancreatic cancer. Journal of Clinial Oncology 2004;22:1430–8.CrossRefGoogle ScholarPubMed
Philip, PA, Benedetti, J, Corless, CL, et al. Phase III study comparing gemcitabine plus cetuximab versus gemcitabine in patients with advanced pancreatic adenocarcinoma: Southwest Oncology Group-directed intergroup trial S0205. Journal of Clinical Oncology 2010;28:3605–10.CrossRefGoogle ScholarPubMed
Van Cutsem, E, Vervenne, WL, Bennouna, J, et al. Phase III trial of bevacizumab in combination with gemcitabine and erlotinib in patients with metastatic pancreatic cancer. Journal of Clinical Oncology 2009;27:2231–7.CrossRefGoogle ScholarPubMed
Maitra, A, Adsay, NV, Argani, P, et al. Multicomponent analysis of the pancreatic adenocarcinoma progression model using a pancreatic intraepithelial neoplasia tissue microarray. Modern Pathology 2003;16:902–12.CrossRef
Omura, N, Li, CP, Li, A, et al. Genome-wide profiling of methylated promoters in pancreatic adenocarcinoma. Cancer Biology and Therapeutics 2008;7:1146–56.CrossRef

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

  • Pancreatic cancer
    • By Siong-Seng Liau, Hepatopancreatobiliary Surgery Unit, Department of Surgery, Addenbrooke’s Hospital, and Medical Research Council Cancer Cell Unit, Hutchison-MRC Research Center, University of Cambridge, Cambridge, UK, David A. Tuveson, Department of Oncology, Addenbrooke’s Hospital, University of Cambridge, Cambridge, UK
  • Edited by Edward P. Gelmann, Columbia University, New York, Charles L. Sawyers, Memorial Sloan-Kettering Cancer Center, New York, Frank J. Rauscher, III
  • Book: Molecular Oncology
  • Online publication: 05 February 2015
  • Chapter DOI: https://doi.org/10.1017/CBO9781139046947.049
Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

  • Pancreatic cancer
    • By Siong-Seng Liau, Hepatopancreatobiliary Surgery Unit, Department of Surgery, Addenbrooke’s Hospital, and Medical Research Council Cancer Cell Unit, Hutchison-MRC Research Center, University of Cambridge, Cambridge, UK, David A. Tuveson, Department of Oncology, Addenbrooke’s Hospital, University of Cambridge, Cambridge, UK
  • Edited by Edward P. Gelmann, Columbia University, New York, Charles L. Sawyers, Memorial Sloan-Kettering Cancer Center, New York, Frank J. Rauscher, III
  • Book: Molecular Oncology
  • Online publication: 05 February 2015
  • Chapter DOI: https://doi.org/10.1017/CBO9781139046947.049
Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

  • Pancreatic cancer
    • By Siong-Seng Liau, Hepatopancreatobiliary Surgery Unit, Department of Surgery, Addenbrooke’s Hospital, and Medical Research Council Cancer Cell Unit, Hutchison-MRC Research Center, University of Cambridge, Cambridge, UK, David A. Tuveson, Department of Oncology, Addenbrooke’s Hospital, University of Cambridge, Cambridge, UK
  • Edited by Edward P. Gelmann, Columbia University, New York, Charles L. Sawyers, Memorial Sloan-Kettering Cancer Center, New York, Frank J. Rauscher, III
  • Book: Molecular Oncology
  • Online publication: 05 February 2015
  • Chapter DOI: https://doi.org/10.1017/CBO9781139046947.049
Available formats
×