Skip to main content
Top

07-06-2016 | Pancreatic cancer | Article

Surgical and molecular pathology of pancreatic neoplasms

Journal: Diagnostic Pathology

Authors: Wenzel M. Hackeng, Ralph H. Hruban, G. Johan A. Offerhaus, Lodewijk A. A. Brosens

Publisher: BioMed Central

Abstract

Background

Histologic characteristics have proven to be very useful for classifying different types of tumors of the pancreas. As a result, the major tumor types in the pancreas have long been classified based on their microscopic appearance.

Main body

Recent advances in whole exome sequencing, gene expression profiling, and knowledge of tumorigenic pathways have deepened our understanding of the underlying biology of pancreatic neoplasia. These advances have not only confirmed the traditional histologic classification system, but also opened new doors to early diagnosis and targeted treatment.

Conclusion

This review discusses the histopathology, genetic and epigenetic alterations and potential treatment targets of the five major malignant pancreatic tumors - pancreatic ductal adenocarcinoma, pancreatic neuroendocrine tumor, solid-pseudopapillary neoplasm, acinar cell carcinoma and pancreatoblastoma.
Literature
1.
Yu J, Blackford AL, Dal Molin M, Wolfgang CL, Goggins M. Time to progression of pancreatic ductal adenocarcinoma from low-to-high tumour stages. Gut. 2015;64:1783–9.PubMedPubMedCentralCrossRef
2.
Siegel RL, Miller KD, Jemal A. Cancer statistics, 2015. CA Cancer J Clin. 2015;65:5–29.PubMedCrossRef
3.
Vincent A, Herman J, Schulick R, Hruban RH, Goggins M. Pancreatic cancer. Lancet. 2011;378:607–20.PubMedPubMedCentralCrossRef
4.
Rahib L, Smith BD, Aizenberg R, Rosenzweig AB, Fleshman JM, Matrisian LM. Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res. 2014;74:2913–21.PubMedCrossRef
5.
Bosman FT, Carneiro F, Hruban RH. WHO classification of tumours of the digestive system. World Health Organization; 2010. http://​link.​springer.​com/​article/​10.​1007%2Fs00534-006-1169-2.
6.
Hruban RH, Pitman MB, Klimstra DS. Tumors of the pancreas. American Registry of Pathology; 2007. http://​link.​springer.​com/​article/​10.​1007%2Fs00534-006-1169-2.
7.
Terada T, Ohta T, Sasaki M, Nakanuma Y, Kim YS. Expression of MUC apomucins in normal pancreas and pancreatic tumours. J Pathol. 1996;180:160–5.PubMedCrossRef
8.
Nagata K, Horinouchi M, Saitou M, Higashi M, Nomoto M, Goto M, et al. Mucin expression profile in pancreatic cancer and the precursor lesions. J Hepatobiliary Pancreat Surg. 2007;14:243–54.PubMedCrossRef
9.
Takeda S, Nakao A, Ichihara T, Suzuki Y, Nonami T, Harada A, et al. Serum concentration and immunohistochemical localization of SPan-1 antigen in pancreatic cancer. A comparison with CA19-9 antigen. Hepatogastroenterology. 1991;38:143–8.PubMed
10.
Brosens LAA, Hackeng WM, Offerhaus GJ, Hruban RH, Wood LD. Pancreatic adenocarcinoma pathology: changing “landscape”. J Gastrointest Oncol. 2015;6:358–74.PubMedPubMedCentral
11.
Basturk O, Hong S-M, Wood LD, Adsay NV, Albores-Saavedra J, Biankin AV, et al. A revised classification system and recommendations from the Baltimore Consensus Meeting for neoplastic precursor lesions in the pancreas. Am J Surg Pathol. 2015;39:1730–41.PubMedCrossRef
12.
Roberts NJ, Norris AL, Petersen GM, Bondy ML, Brand R, Gallinger S, et al. Whole genome sequencing defines the genetic heterogeneity of familial pancreatic cancer. Cancer Discov 2015:CD–15–0402.
13.
Jones S, Hruban RH, Kamiyama M, Borges M, Zhang X, Parsons DW, et al. Exomic sequencing identifies PALB2 as a pancreatic cancer susceptibility gene. Science. 2009;324:217.PubMedPubMedCentralCrossRef
14.
Zhen DB, Rabe KG, Gallinger S, Syngal S, Schwartz AG, Goggins MG, et al. BRCA1, BRCA2, PALB2, and CDKN2A mutations in familial pancreatic cancer: a PACGENE study. Genet Med. 2015;17:569–77.PubMedCrossRef
15.
Roberts NJ, Jiao Y, Yu J, Kopelovich L, Petersen GM, Bondy ML, et al. ATM mutations in patients with hereditary pancreatic cancer. Cancer Discov. 2012;2:41–6.PubMedCrossRef
16.
Hearle N, Schumacher V, Menko FH, Olschwang S, Boardman LA, Gille JJP, et al. Frequency and spectrum of cancers in the Peutz-Jeghers syndrome. Clin Cancer Res. 2006;12:3209–15.PubMedCrossRef
17.
Kastrinos F, Mukherjee B, Tayob N, Wang F, Sparr J, Raymond VM, et al. Risk of pancreatic cancer in families with Lynch syndrome. JAMA. 2009;302:1790–5.PubMedPubMedCentralCrossRef
18.
Canto MI, Harinck F, Hruban RH, Offerhaus GJ, Poley J-W, Kamel I, et al. International Cancer of the Pancreas Screening (CAPS) Consortium summit on the management of patients with increased risk for familial pancreatic cancer. 2013. p. 339–47.
19.
Le DT, Uram JN, Wang H, Bartlett BR, Kemberling H, Eyring AD, et al. PD-1 blockade in tumors with mismatch-repair deficiency. N Engl J Med. 2015;372:2509–20.PubMedCrossRef
20.
Bhalla A, Saif MW. PARP-inhibitors in BRCA-associated pancreatic cancer. JOP. 2014;15:340–3.PubMed
21.
Waddell N, Pajic M, Patch A-M, Chang DK, Kassahn KS, Bailey P, et al. Whole genomes redefine the mutational landscape of pancreatic cancer. Nature. 2015;518:495–501.PubMedPubMedCentralCrossRef
22.
Childs EJ, Mocci E, Campa D, Bracci PM, Gallinger S, Goggins M, et al. Common variation at 2p13.3, 3q29, 7p13 and 17q25.1 associated with susceptibility to pancreatic cancer. Nat Genet. 2015;47:911–6.PubMedPubMedCentralCrossRef
23.
Wolpin BM, Rizzato C, Kraft P, Kooperberg C, Petersen GM, Wang Z, et al. Genome-wide association study identifies multiple susceptibility loci for pancreatic cancer. Nat Genet. 2014;46:994–1000.PubMedPubMedCentralCrossRef
24.
Wolpin BM, Kraft P, Xu M, Steplowski E, Olsson ML, Arslan AA, et al. Variant ABO blood group alleles, secretor status, and risk of pancreatic cancer: results from the pancreatic cancer cohort consortium. Cancer Epidemiol Biomarkers Prev. 2010;19:3140–9.PubMedPubMedCentralCrossRef
25.
Biankin AV, Waddell N, Kassahn KS, Gingras M-C, Muthuswamy LB, Johns AL, et al. Pancreatic cancer genomes reveal aberrations in axon guidance pathway genes. Nature. 2012;491:399–405.PubMedPubMedCentralCrossRef
26.
Jones S, Zhang X, Parsons DW, Lin JC-H, Leary RJ, Angenendt P, et al. Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science. 2008;321:1801–6.PubMedPubMedCentralCrossRef
27.
Witkiewicz AK, McMillan EA, Balaji U, Baek G, Lin W-C, Mansour J, et al. Whole-exome sequencing of pancreatic cancer defines genetic diversity and therapeutic targets. Nat Commun. 2015;6:6744.PubMedPubMedCentralCrossRef
28.
Griffin CA, Hruban RH, Morsberger LA, Ellingham T, Long PP, Jaffee EM, et al. Consistent chromosome abnormalities in adenocarcinoma of the pancreas. Cancer Res. 1995;55:2394–9.PubMed
29.
Sausen M, Phallen J, Adleff V, Jones S, Leary RJ, Barrett MT, et al. Clinical implications of genomic alterations in the tumour and circulation of pancreatic cancer patients. Nat Commun. 2015;6:7686.PubMedPubMedCentralCrossRef
30.
Blackford A, Serrano OK, Wolfgang CL, Parmigiani G, Jones S, Zhang X, et al. SMAD4 gene mutations are associated with poor prognosis in pancreatic cancer. Clin Cancer Res. 2009;15:4674–9.PubMedPubMedCentralCrossRef
31.
Castro-Giner F, Ratcliffe P, Tomlinson I. The mini-driver model of polygenic cancer evolution. Nat Rev Cancer. 2015;15:680–5.PubMedCrossRef
32.
Yachida S, Jones S, Bozic I, Antal T, Leary R, Fu B, et al. Distant metastasis occurs late during the genetic evolution of pancreatic cancer. Nature. 2010;467:1114–7.PubMedPubMedCentralCrossRef
33.
Bettegowda C, Sausen M, Leary RJ, Kinde I, Wang Y, Agrawal N, et al. Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci Transl Med. 2014;6:224ra24-4.CrossRef
34.
Springer S, Wang Y, Dal Molin M, Masica DL, Jiao Y, Kinde I, et al. A combination of molecular markers and clinical features improve the classification of pancreatic cysts. Gastroenterology. 2015;149:1501–10.PubMedCrossRef
35.
Shimizu H, Horii A, Sunamura M, Motoi F, Egawa S, Unno M, et al. Identification of epigenetically silenced genes in human pancreatic cancer by a novel method “microarray coupled with methyl-CpG targeted transcriptional activation” (MeTA-array). Biochem Biophys Res Commun. 2011;411:162–7.PubMedCrossRef
36.
Tan AC, Jimeno A, Lin SH, Wheelhouse J, Chan F, Solomon A, et al. Characterizing DNA methylation patterns in pancreatic cancer genome. Mol Oncol. 2009;3:425–38.PubMedCrossRef
37.
Vincent A, Omura N, Hong S-M, Jaffe A, Eshleman J, Goggins M. Genome-wide analysis of promoter methylation associated with gene expression profile in pancreatic adenocarcinoma. Clin Cancer Res. 2011;17:4341–54.PubMedPubMedCentralCrossRef
38.
Nones K, Waddell N, Song S, Patch A-M, Miller D, Johns A, et al. Genome-wide DNA methylation patterns in pancreatic ductal adenocarcinoma reveal epigenetic deregulation of SLIT-ROBO, ITGA2 and MET signaling. Int J Cancer. 2014;135:1110–8.PubMedCrossRef
39.
Omura N, Li C-P, Li A, Hong S-M, Walter K, Jimeno A, et al. Genome-wide profiling of methylated promoters in pancreatic adenocarcinoma. Cancer Biol Ther. 2008;7:1146–56.PubMedPubMedCentralCrossRef
40.
Zhao Y, Sun J, Zhang H, Guo S, Gu J, Wang W, et al. High-frequency aberrantly methylated targets in pancreatic adenocarcinoma identified via global DNA methylation analysis using methylCap-seq. Clin Epigenetics. 2014;6:18.PubMedPubMedCentralCrossRef
41.
Sato N, Fukushima N, Maitra A, Matsubayashi H, Yeo CJ, Cameron JL, et al. Discovery of novel targets for aberrant methylation in pancreatic carcinoma using high-throughput microarrays. Cancer Res. 2003;63:3735–42.PubMed
42.
Goonesekere NCW, Wang X, Ludwig L, Guda C. A meta analysis of pancreatic microarray datasets yields new targets as cancer genes and biomarkers. PLoS One. 2014;9, e93046.PubMedPubMedCentralCrossRef
43.
Hong S-M, Park JY, Hruban RH, Goggins M. Molecular signatures of pancreatic cancer. Arch Pathol Lab Med. 2011;135:716–27.PubMedPubMedCentral
44.
Fukushima N, Sato N, Ueki T, Rosty C, Walter KM, Wilentz RE, et al. Aberrant methylation of preproenkephalin and p16 genes in pancreatic intraepithelial neoplasia and pancreatic ductal adenocarcinoma. Am J Pathol. 2002;160:1573–81.PubMedPubMedCentralCrossRef
45.
Moore PS, Sipos B, Orlandini S, Sorio C, Real FX, Lemoine NR, et al. Genetic profile of 22 pancreatic carcinoma cell lines. Analysis of K-ras, p53, p16 and DPC4/Smad4. Virchows Arch. 2001;439:798–802.PubMedCrossRef
46.
Ueki T, Toyota M, Sohn T, Yeo CJ, Issa JP, Hruban RH, et al. Hypermethylation of multiple genes in pancreatic adenocarcinoma. Cancer Res. 2000;60:1835–9.PubMed
47.
Matsubayashi H, Canto M, Sato N, Klein A, Abe T, Yamashita K, et al. DNA methylation alterations in the pancreatic juice of patients with suspected pancreatic disease. Cancer Res. 2006;66:1208–17.PubMedCrossRef
48.
Habbe N, Koorstra J-BM, Mendell JT, Offerhaus GJ, Ryu JK, Feldmann G, et al. MicroRNA miR-155 is a biomarker of early pancreatic neoplasia. Cancer Biol Ther. 2009;8:340–6.PubMedPubMedCentralCrossRef
49.
Frampton AE, Krell J, Jamieson NB, Gall TMH, Giovannetti E, Funel N, et al. microRNAs with prognostic significance in pancreatic ductal adenocarcinoma: a meta-analysis. Eur J Cancer. 2015;51:1389–404.PubMedCrossRef
50.
Ma M-Z, Kong X, Weng M-Z, Cheng K, Gong W, Quan Z-W, et al. Candidate microRNA biomarkers of pancreatic ductal adenocarcinoma: meta-analysis, experimental validation and clinical significance. J Exp Clin Cancer Res. 2013;32:71.PubMedPubMedCentralCrossRef
51.
Yang J-Y, Sun Y-W, Liu D-J, Zhang J-F, Li J, Hua R. MicroRNAs in stool samples as potential screening biomarkers for pancreatic ductal adenocarcinoma cancer. Am J Cancer Res. 2014;4:663–73.PubMedPubMedCentral
52.
Abue M, Yokoyama M, Shibuya R, Tamai K, Yamaguchi K, Sato I, et al. Circulating miR-483-3p and miR-21 is highly expressed in plasma of pancreatic cancer. Int J Oncol. 2015;46(2):539-47.
53.
Volinia S, Calin GA, Liu C-G, Ambs S, Cimmino A, Petrocca F, et al. A microRNA expression signature of human solid tumors defines cancer gene targets. Proc Natl Acad Sci U S A. 2006;103:2257–61.PubMedPubMedCentralCrossRef
54.
Hatley ME, Patrick DM, Garcia MR, Richardson JA, Bassel-Duby R, van Rooij E, et al. Modulation of K-Ras-dependent lung tumorigenesis by MicroRNA-21. Cancer Cell. 2010;18:282–93.PubMedPubMedCentralCrossRef
55.
Selcuklu SD, Donoghue MTA, Spillane C. miR-21 as a key regulator of oncogenic processes. Biochem Soc Trans. 2009;37:918–25.PubMedCrossRef
56.
Toste PA, Li L, Kadera BE, Nguyen AH, Tran LM, Wu N, et al. p85α is a microRNA target and affects chemosensitivity in pancreatic cancer. J Surg Res. 2015;196:285–93.PubMedCrossRef
57.
Wang C, Sun Y, Wu H, Yu S, Zhang L, Meng Y, et al. Elevated miR-483-3p expression is an early event and indicates poor prognosis in pancreatic ductal adenocarcinoma. Tumour Biol. 2015;36(12):9447-56.
58.
Bai Z, Sun J, Wang X, Wang H, Pei H, Zhang Z. MicroRNA-153 is a prognostic marker and inhibits cell migration and invasion by targeting SNAI1 in human pancreatic ductal adenocarcinoma. Oncol Rep. 2015;34:595–602.PubMedPubMedCentral
59.
Xia X, Zhang K, Cen G, Jiang T, Cao J, Huang K, et al. MicroRNA-301a-3p promotes pancreatic cancer progression via negative regulation of SMAD4. Oncotarget. 2015;6:21046–63.PubMedPubMedCentralCrossRef
60.
Zhu Z, Xu Y, Zhao J, Liu Q, Feng W, Fan J, et al. miR-367 promotes epithelial-to-mesenchymal transition and invasion of pancreatic ductal adenocarcinoma cells by targeting the Smad7-TGF-β signalling pathway. Br J Cancer. 2015;112:1367–75.PubMedPubMedCentralCrossRef
61.
Müller S, Raulefs S, Bruns P, Afonso-Grunz F, Plötner A, Thermann R, et al. Next-generation sequencing reveals novel differentially regulated mRNAs, lncRNAs, miRNAs, sdRNAs and a piRNA in pancreatic cancer. Mol Cancer. 2015;14:94.PubMedPubMedCentralCrossRef
62.
Collisson EA, Sadanandam A, Olson P, Gibb WJ, Truitt M, Gu S, et al. Subtypes of pancreatic ductal adenocarcinoma and their differing responses to therapy. Nat Med. 2011;17:500–3.PubMedPubMedCentralCrossRef
63.
Moffitt RA, Marayati R, Flate EL, Volmar KE, Loeza SGH, Hoadley KA, et al. Virtual microdissection identifies distinct tumor- and stroma-specific subtypes of pancreatic ductal adenocarcinoma. Nat Genet. 2015;47:1168–78.PubMedCrossRef
64.
Harsha HC, Kandasamy K, Ranganathan P, Rani S, Ramabadran S, Gollapudi S, et al. A compendium of potential biomarkers of pancreatic cancer. PLoS Med. 2009;6, e1000046.PubMedPubMedCentralCrossRef
65.
Wang Q, Chaerkady R, Wu J, Hwang HJ, Papadopoulos N, Kopelovich L, et al. Mutant proteins as cancer-specific biomarkers. Proc Natl Acad Sci U S A. 2011;108:2444–9.PubMedPubMedCentralCrossRef
66.
Zhang Y, Choi M. Immune therapy in pancreatic cancer: now and the future? Rev Recent Clin Trials. 2015;10:317–25.PubMedCrossRef
67.
Rosenberg SA, Restifo NP. Adoptive cell transfer as personalized immunotherapy for human cancer. Science. 2015;348:62–8.PubMedCrossRef
68.
Xu Z, Pothula SP, Wilson JS, Apte MV. Pancreatic cancer and its stroma: a conspiracy theory. World J Gastroenterol. 2014;20:11216–29.PubMedPubMedCentralCrossRef
69.
Rhim AD, Oberstein PE, Thomas DH, Mirek ET, Palermo CF, Sastra SA, et al. Stromal elements act to restrain, rather than support, pancreatic ductal adenocarcinoma. Cancer Cell. 2014;25:735–47.PubMedPubMedCentralCrossRef
70.
Stromnes IM, DelGiorno KE, Greenberg PD, Hingorani SR. Stromal reengineering to treat pancreas cancer. Carcinogenesis. 2014;35:1451–60.PubMedPubMedCentralCrossRef
71.
Halfdanarson TR, Rabe KG, Rubin J, Petersen GM. Pancreatic neuroendocrine tumors (PNETs): incidence, prognosis and recent trend toward improved survival. Ann Oncol. 2008;19:1727–33.PubMedPubMedCentralCrossRef
72.
Ries LAG, Young Jr JL, Keel GE, Eisner MP, Lin YD, Horner MJD. Cancer survival among adults. Bethesda, MD: US Department of Health and Human Services, National Institutes of Health, National Cancer Institute; 2007.
73.
Falconi M, Eriksson B, Kaltsas G, Bartsch DK, Capdevila J, Caplin M, et al. Consensus guidelines update for the management of Functional p-NETs (F-p-NETs) and Non-Functional p-NETs (NF-p-NETs). Int J Oncol. 2015;46(2):539-47.
74.
Rindi G, Falconi M, Klersy C, Albarello L, Boninsegna L, Buchler MW, et al. TNM staging of neoplasms of the endocrine pancreas: results from a large international cohort study. J Natl Cancer Inst. 2012;104:764–77.PubMedCrossRef
75.
Basturk O, Yang Z, Tang LH, Hruban RH, Adsay V, McCall CM, et al. The high-grade (WHO G3) pancreatic neuroendocrine tumor category is morphologically and biologically heterogenous and includes both well differentiated and poorly differentiated neoplasms. Am J Surg Pathol. 2015;39:683–90.PubMedPubMedCentralCrossRef
76.
Solcia E, Klöppel G, Sobin LH. Histological typing of endocrine tumours. Berlin, Heidelberg: Springer Science & Business Media; 2000.CrossRef
77.
Hammond EH, Yowell RL, Flinner RL. Neuroendocrine carcinomas: role of immunocytochemistry and electron microscopy. Hum Pathol. 1998;29:1367–71.PubMedCrossRef
78.
Yachida S, Vakiani E, White CM, Zhong Y, Saunders T, Morgan R, et al. Small cell and large cell neuroendocrine carcinomas of the pancreas are genetically similar and distinct from well-differentiated pancreatic neuroendocrine tumors. Am J Surg Pathol. 2012;36:173–84.PubMedPubMedCentralCrossRef
79.
Sipos B, Sperveslage J, Anlauf M, Hoffmeister M, Henopp T, Buch S, et al. Glucagon cell hyperplasia and neoplasia with and without glucagon receptor mutations. J Clin Endocrinol Metab. 2015;100:E783–8.PubMedCrossRef
80.
Klöppel G, Anlauf M, Perren A, Sipos B. Hyperplasia to neoplasia sequence of duodenal and pancreatic neuroendocrine diseases and pseudohyperplasia of the PP-cells in the pancreas. Endocr Pathol. 2014;25:181–5.PubMedCrossRef
81.
Esposito I, Segler A, Steiger K, Klöppel G. Pathology, genetics and precursors of human and experimental pancreatic neoplasms: an update. Int J Oncol. 2015;46(2):539-47.
82.
Jiao Y, Shi C, Edil BH, de Wilde RF, Klimstra DS, Maitra A, et al. DAXX/ATRX, MEN1, and mTOR pathway genes are frequently altered in pancreatic neuroendocrine tumors. Science. 2011;331:1199–203.PubMedPubMedCentralCrossRef
83.
Lubensky IA, Debelenko LV, Zhuang Z, Emmert-Buck MR, Dong Q, Chandrasekharappa S, et al. Allelic deletions on chromosome 11q13 in multiple tumors from individual MEN1 patients. Cancer Res. 1996;56:5272–8.PubMed
84.
Görtz B, Roth J, Krähenmann A, de Krijger RR, Muletta-Feurer S, Rütimann K, et al. Mutations and allelic deletions of the MEN1 gene are associated with a subset of sporadic endocrine pancreatic and neuroendocrine tumors and not restricted to foregut neoplasms. Am J Pathol. 1999;154:429–36.PubMedPubMedCentralCrossRef
85.
Heaphy CM, de Wilde RF, Jiao Y, Klein AP, Edil BH, Shi C, et al. Altered telomeres in tumors with ATRX and DAXX mutations. Science. 2011;333:425.PubMedPubMedCentralCrossRef
86.
Yuan F, Shi M, Ji J, Shi H, Zhou C, Yu Y, et al. KRAS and DAXX/ATRX gene mutations are correlated with the clinicopathological features, advanced diseases, and poor prognosis in Chinese patients with pancreatic neuroendocrine tumors. Int J Biol Sci. 2014;10:957–65.PubMedPubMedCentralCrossRef
87.
Marinoni I, Kurrer AS, Vassella E, Dettmer M, Rudolph T, Banz V, et al. Loss of DAXX and ATRX are associated with chromosome instability and reduced survival of patients with pancreatic neuroendocrine tumors. Gastroenterology. 2014;146:453–5.PubMedCrossRef
88.
Schmitt AM, Schmid S, Rudolph T, Anlauf M, Prinz C, Klöppel G, et al. VHL inactivation is an important pathway for the development of malignant sporadic pancreatic endocrine tumors. Endocr Relat Cancer. 2009;16:1219–27.PubMedCrossRef
89.
Nikiforova MN, Nikiforov YE, Biddinger P, Gnepp DR, Grosembacher LA, Wajchenberg BL, et al. Frequent loss of heterozygosity at chromosome 3p14.2-3p21 in human pancreatic islet cell tumours. Clin Endocrinol (Oxf). 1999;51:27–33.CrossRef
90.
Ohki R, Saito K, Chen Y, Kawase T, Hiraoka N, Saigawa R, et al. PHLDA3 is a novel tumor suppressor of pancreatic neuroendocrine tumors. Proc Natl Acad Sci U S A. 2014;111:E2404–13.PubMedPubMedCentralCrossRef
91.
Floridia G, Grilli G, Salvatore M, Pescucci C, Moore PS, Scarpa A, et al. Chromosomal alterations detected by comparative genomic hybridization in nonfunctioning endocrine pancreatic tumors. Cancer Genet Cytogenet. 2005;156:23–30.PubMedCrossRef
92.
Hessman O, Skogseid B, Westin G, Akerström G. Multiple allelic deletions and intratumoral genetic heterogeneity in men1 pancreatic tumors. J Clin Endocrinol Metab. 2001;86:1355–61.PubMed
93.
Hessman O, Lindberg D, Einarsson A, Lillhager P, Carling T, Grimelius L, et al. Genetic alterations on 3p, 11q13, and 18q in nonfamilial and MEN 1-associated pancreatic endocrine tumors. Genes Chromosomes Cancer. 1999;26:258–64.PubMedCrossRef
94.
Perren A, Anlauf M, Henopp T, Rudolph T, Schmitt A, Raffel A, et al. Multiple endocrine neoplasia type 1 (MEN1): loss of one MEN1 allele in tumors and monohormonal endocrine cell clusters but not in islet hyperplasia of the pancreas. J Clin Endocrinol Metab. 2007;92:1118–28.PubMedCrossRef
95.
de Wilde RF, Heaphy CM, Maitra A, Meeker AK, Edil BH, Wolfgang CL, et al. Loss of ATRX or DAXX expression and concomitant acquisition of the alternative lengthening of telomeres phenotype are late events in a small subset of MEN-1 syndrome pancreatic neuroendocrine tumors. Mod Pathol. 2012;25:1033–9.PubMedPubMedCentralCrossRef
96.
Stefanoli M, La Rosa S, Sahnane N, Romualdi C, Pastorino R, Marando A, et al. Prognostic relevance of aberrant DNA methylation in g1 and g2 pancreatic neuroendocrine tumors. Neuroendocrinology. 2014;100:26–34.PubMedCrossRef
97.
House MG, Herman JG, Guo MZ, Hooker CM, Schulick RD, Lillemoe KD, et al. Aberrant hypermethylation of tumor suppressor genes in pancreatic endocrine neoplasms. Ann Surg. 2003;238:423–31. discussion 431–2.PubMedPubMedCentral
98.
Malpeli G, Amato E, Dandrea M, Fumagalli C, Debattisti V, Boninsegna L, et al. Methylation-associated down-regulation of RASSF1A and up-regulation of RASSF1C in pancreatic endocrine tumors. BMC Cancer. 2011;11:351.PubMedPubMedCentralCrossRef
99.
Estrabaud E, Lassot I, Blot G, Le Rouzic E, Tanchou V, Quemeneur E, et al. RASSF1C, an isoform of the tumor suppressor RASSF1A, promotes the accumulation of beta-catenin by interacting with betaTrCP. Cancer Res. 2007;67:1054–61.PubMedCrossRef
100.
Ram RR, Mendiratta S, Bodemann BO, Torres MJ, Eskiocak U, White MA. RASSF1A inactivation unleashes a tumor suppressor/oncogene cascade with context-dependent consequences on cell cycle progression. Mol Cell Biol. 2014;34:2350–8.PubMedPubMedCentralCrossRef
101.
Roldo C, Missiaglia E, Hagan JP, Falconi M, Capelli P, Bersani S, et al. MicroRNA expression abnormalities in pancreatic endocrine and acinar tumors are associated with distinctive pathologic features and clinical behavior. J Clin Oncol. 2006;24:4677–84.PubMedCrossRef
102.
Thorns C, Schurmann C, Gebauer N, Wallaschofski H, Kümpers C, Bernard V, et al. Global microRNA profiling of pancreatic neuroendocrine neoplasias. Anticancer Res. 2014;34:2249–54.PubMed
103.
Maitra A, Hansel DE, Argani P, Ashfaq R, Rahman A, Naji A, et al. Global expression analysis of well-differentiated pancreatic endocrine neoplasms using oligonucleotide microarrays. Clin Cancer Res. 2003;9:5988–95.PubMed
104.
Speisky D, Duces A, Bièche I, Rebours V, Hammel P, Sauvanet A, et al. Molecular profiling of pancreatic neuroendocrine tumors in sporadic and Von Hippel-Lindau patients. Clin Cancer Res. 2012;18:2838–49.PubMedCrossRef
105.
Raymond E, Dahan L, Raoul J-L, Bang Y-J, Borbath I, Lombard-Bohas C, et al. Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. N Engl J Med. 2011;364:501–13.PubMedCrossRef
106.
Vinik AI, Raymond E. Pancreatic neuroendocrine tumors: approach to treatment with focus on sunitinib. Therap Adv Gastroenterol. 2013;6:396–411.PubMedPubMedCentralCrossRef
107.
Pea A, Hruban RH, Wood LD. Genetics of pancreatic neuroendocrine tumors: implications for the clinic. Expert Rev Gastroenterol Hepatol. 2015;9:1407–19.PubMedCrossRef
108.
Yao JC, Shah MH, Ito T, Bohas CL, Wolin EM, Van Cutsem E, et al. Everolimus for advanced pancreatic neuroendocrine tumors. N Engl J Med. 2011;364:514–23.PubMedPubMedCentralCrossRef
109.
Law JK, Ahmed A, Singh VK, Akshintala VS, Olson MT, Raman SP, et al. A systematic review of solid-pseudopapillary neoplasms: are these rare lesions? Pancreas. 2014;43:331–7.PubMedPubMedCentralCrossRef
110.
Notohara K, Hamazaki S, Tsukayama C, Nakamoto S, Kawabata K, Mizobuchi K, et al. Solid-pseudopapillary tumor of the pancreas: immunohistochemical localization of neuroendocrine markers and CD10. Am J Surg Pathol. 2000;24:1361–71.PubMedCrossRef
111.
Miettinen M, Partanen S, Fräki O, Kivilaakso E. Papillary cystic tumor of the pancreas. An analysis of cellular differentiation by electron microscopy and immunohistochemistry. Am J Surg Pathol. 1987;11:855–65.PubMedCrossRef
112.
El-Bahrawy MA, Rowan A, Horncastle D, Tomlinson I, Theis BA, Russell RCG, et al. E-cadherin/catenin complex status in solid pseudopapillary tumor of the pancreas. Am J Surg Pathol. 2008;32:1–7.PubMedCrossRef
113.
Guo Y, Yuan F, Deng H, Wang H-F, Jin X-L, Xiao J-C. Paranuclear dot-like immunostaining for CD99: a unique staining pattern for diagnosing solid-pseudopapillary neoplasm of the pancreas. Am J Surg Pathol. 2011;35:799–806.PubMedCrossRef
114.
Singhi AD, Lilo M, Hruban RH, Cressman KL, Fuhrer K, Seethala RR. Overexpression of lymphoid enhancer-binding factor 1 (LEF1) in solid-pseudopapillary neoplasms of the pancreas. Mod Pathol. 2014;27:1355–63.PubMedPubMedCentralCrossRef
115.
Tanaka Y, Kato K, Notohara K, Hojo H, Ijiri R, Miyake T, et al. Frequent beta-catenin mutation and cytoplasmic/nuclear accumulation in pancreatic solid-pseudopapillary neoplasm. Cancer Res. 2001;61:8401–4.PubMed
116.
Abraham SC, Klimstra DS, Wilentz RE, Yeo CJ, Conlon K, Brennan M, et al. Solid-pseudopapillary tumors of the pancreas are genetically distinct from pancreatic ductal adenocarcinomas and almost always harbor beta-catenin mutations. Am J Pathol. 2002;160:1361–9.PubMedPubMedCentralCrossRef
117.
Ruo L, Coit DG, Brennan MF, Guillem JG. Long-term follow-up of patients with familial adenomatous polyposis undergoing pancreaticoduodenal surgery. J Gastrointest Surg. 2002;6:671–5.PubMedCrossRef
118.
Inoue T, Nishi Y, Okumura F, Mizushima T, Nishie H, Iwasaki H, et al. Solid pseudopapillary neoplasm of the pancreas associated with familial adenomatous polyposis. Intern Med. 2015;54:1349–55.PubMedCrossRef
119.
Tognarini I, Tonelli F, Nesi G, Martineti V, Galli G, Gozzini A, et al. In vitro effects of oestrogens, antioestrogens and SERMs on pancreatic solid pseudopapillary neoplasm-derived primary cell culture. Cell Oncol. 2010;32:331–43.PubMedPubMedCentral
120.
Müller-Höcker J, Zietz CH, Sendelhofert A. Deregulated expression of cell cycle-associated proteins in solid pseudopapillary tumor of the pancreas. Mod Pathol. 2001;14:47–53.PubMedCrossRef
121.
Tiemann K, Heitling U, Kosmahl M, Klöppel G. Solid pseudopapillary neoplasms of the pancreas show an interruption of the Wnt-signaling pathway and express gene products of 11q. Mod Pathol. 2007;20:955–60.PubMedCrossRef
122.
Park M, Lim J-S, Lee H-J, Na K, Lee MJ, Kang CM, et al. Distinct protein expression profiles of solid-pseudopapillary neoplasms of the pancreas. J Proteome Res. 2015;14:3007–14.PubMedCrossRef
123.
Park M, Kim M, Hwang D, Park M, Kim WK, Kim SK, et al. Characterization of gene expression and activated signaling pathways in solid-pseudopapillary neoplasm of pancreas. Mod Pathol. 2014;27:580–93.PubMedCrossRef
124.
Cavard C, Audebourg A, Letourneur F, Audard V, Beuvon F, Cagnard N, et al. Gene expression profiling provides insights into the pathways involved in solid pseudopapillary neoplasm of the pancreas. J Pathol. 2009;218:201–9.PubMedCrossRef
125.
Seket B, Saurin J-C, Scoazec J-Y, Partensky C. [Pancreatic acinar cell carcinoma in a patient with familial adenomatous polyposis]. Gastroenterol Clin Biol. 2003;27:818–20.PubMed
126.
Lowery MA, Klimstra DS, Shia J, Yu KH, Allen PJ, Brennan MF, et al. Acinar cell carcinoma of the pancreas: new genetic and treatment insights into a rare malignancy. Oncologist. 2011;16:1714–20.PubMedPubMedCentralCrossRef
127.
de Wilde RF, Ottenhof NA, Jansen M, Morsink FHM, de Leng WWJ, Offerhaus GJA, et al. Analysis of LKB1 mutations and other molecular alterations in pancreatic acinar cell carcinoma. Mod Pathol. 2011;24:1229–36.PubMedCrossRef
128.
Liu W, Shia J, Gönen M, Lowery MA, O’Reilly EM, Klimstra DS. DNA mismatch repair abnormalities in acinar cell carcinoma of the pancreas: frequency and clinical significance. Pancreas. 2014;43:1264–70.PubMedCrossRef
129.
Wisnoski NC, Townsend CM, Nealon WH, Freeman JL, Riall TS. 672 patients with acinar cell carcinoma of the pancreas: a population-based comparison to pancreatic adenocarcinoma. Surgery. 2008;144:141–8.PubMedCrossRef
130.
Cingolani N, Shaco-Levy R, Farruggio A, Klimstra DS, Rosai J. Alpha-fetoprotein production by pancreatic tumors exhibiting acinar cell differentiation: study of five cases, one arising in a mediastinal teratoma. Hum Pathol. 2000;31:938–44.PubMedCrossRef
131.
Ono J, Sakamoto H, Sakoda K, Yagi Y, Hagio S, Sato E, et al. Acinar cell carcinoma of the pancreas with elevated serum alpha-fetoprotein. Int Surg. 1984;69:361–4.PubMed
132.
Klimstra DS, Heffess CS, Oertel JE, Rosai J. Acinar cell carcinoma of the pancreas. A clinicopathologic study of 28 cases. Am J Surg Pathol. 1992;16:815–37.PubMedCrossRef
133.
La Rosa S, Franzi F, Marchet S, Finzi G, Clerici M, Vigetti D, et al. The monoclonal anti-BCL10 antibody (clone 331.1) is a sensitive and specific marker of pancreatic acinar cell carcinoma and pancreatic metaplasia. Virchows Arch. 2009;454:133–42.PubMedCrossRef
134.
Hosoda W, Sasaki E, Murakami Y, Yamao K, Shimizu Y, Yatabe Y. BCL10 as a useful marker for pancreatic acinar cell carcinoma, especially using endoscopic ultrasound cytology specimens. Pathol Int. 2013;63:176–82.PubMedCrossRef
135.
Yasumoto M, Hamabashiri M, Akiba J, Ogasawara S, Naito Y, Taira T, et al. The utility of a novel antibody in the pathological diagnosis of pancreatic acinar cell carcinoma. J Clin Pathol. 2012;65:327–32.PubMedCrossRef
136.
Abraham SC, Wu T-T, Hruban RH, Lee J-H, Yeo CJ, Conlon K, et al. Genetic and immunohistochemical analysis of pancreatic acinar cell carcinoma: frequent allelic loss on chromosome 11p and alterations in the APC/beta-catenin pathway. Am J Pathol. 2002;160:953–62.PubMedPubMedCentralCrossRef
137.
Jiao Y, Yonescu R, Offerhaus GJA, Klimstra DS, Maitra A, Eshleman JR, et al. Whole-exome sequencing of pancreatic neoplasms with acinar differentiation. J Pathol. 2014;232:428–35.PubMedPubMedCentralCrossRef
138.
Rigaud G, Moore PS, Zamboni G, Orlandini S, Taruscio D, Paradisi S, et al. Allelotype of pancreatic acinar cell carcinoma. Int J Cancer. 2000;88:772–7.PubMedCrossRef
139.
La Rosa S, Sessa F, Capella C. Acinar cell carcinoma of the pancreas: overview of clinicopathologic features and insights into the molecular pathology. Front Med (Lausanne). 2015;2:41.
140.
Dewald GW, Smyrk TC, Thorland EC, McWilliams RR, Van Dyke DL, Keefe JG, et al. Fluorescence in situ hybridization to visualize genetic abnormalities in interphase cells of acinar cell carcinoma, ductal adenocarcinoma, and islet cell carcinoma of the pancreas. Mayo Clin Proc. 2009;84:801–10.PubMedPubMedCentralCrossRef
141.
Chmielecki J, Hutchinson KE, Frampton GM, Chalmers ZR, Johnson A, Shi C, et al. Comprehensive genomic profiling of pancreatic acinar cell carcinomas identifies recurrent RAF fusions and frequent inactivation of DNA repair genes. Cancer Discov. 2014;4:1398–405.PubMedCrossRef
142.
Furlan D, Sahnane N, Bernasconi B, Frattini M, Tibiletti MG, Molinari F, et al. APC alterations are frequently involved in the pathogenesis of acinar cell carcinoma of the pancreas, mainly through gene loss and promoter hypermethylation. Virchows Arch. 2014;464:553–64.PubMedCrossRef
143.
Guo M, Jia Y, Yu Z, House MG, Esteller M, Brock MV, et al. Epigenetic changes associated with neoplasms of the exocrine and endocrine pancreas. Discov Med. 2014;17:67–73.PubMedPubMedCentral
144.
Dhebri AR, Connor S, Campbell F, Ghaneh P, Sutton R, Neoptolemos JP. Diagnosis, treatment and outcome of pancreatoblastoma. Pancreatology. 2004;4:441–51. discussion 452–3.PubMedCrossRef
145.
Salman B, Brat G, Yoon Y-S, Hruban RH, Singhi AD, Fishman EK, et al. The diagnosis and surgical treatment of pancreatoblastoma in adults: a case series and review of the literature. J Gastrointest Surg. 2013;17:2153–61.PubMedCrossRef
146.
Weksberg R, Shuman C, Beckwith JB. Beckwith-Wiedemann syndrome. Eur J Hum Genet. 2010;18:8–14.PubMedCrossRef
147.
Abraham SC, Wu TT, Klimstra DS, Finn LS, Lee JH, Yeo CJ, et al. Distinctive molecular genetic alterations in sporadic and familial adenomatous polyposis-associated pancreatoblastomas : frequent alterations in the APC/beta-catenin pathway and chromosome 11p. Am J Pathol. 2001;159:1619–27.PubMedPubMedCentralCrossRef
148.
Maher ER, Reik W. Beckwith-Wiedemann syndrome: imprinting in clusters revisited. J Clin Invest. 2000;105:247–52.PubMedPubMedCentralCrossRef
149.
Wong IHN, Chan J, Wong J, Tam PKH. Ubiquitous aberrant RASSF1A promoter methylation in childhood neoplasia. Clin Cancer Res. 2004;10:994–1002.PubMedCrossRef
150.
Honda S, Okada T, Miyagi H, Minato M, Suzuki H, Taketomi A. Spontaneous rupture of an advanced pancreatoblastoma: aberrant RASSF1A methylation and CTNNB1 mutation as molecular genetic markers. J Pediatr Surg. 2013;48:e29–32.PubMedCrossRef
151.
Ghiorzo P. Genetic predisposition to pancreatic cancer. World J Gastroenterol. 2014;20:10778–89.PubMedPubMedCentralCrossRef