Targeted next-generation sequencing of cancer genes dissects the molecular profiles of intraductal papillary neoplasms of the pancreas

Eliana Amato, Marco Dal Molin, Andrea Mafficini, Jun Yu, Giuseppe Malleo, Borislav Rusev, Matteo Fassan, Davide Antonello, Yoshihiko Sadakari, Paola Castelli, Giuseppe Zamboni, Anirban Maitra, Roberto Salvia, Ralph H Hruban, Claudio Bassi, Paola Capelli, Rita T Lawlor, Michael Goggins, Aldo Scarpa, Eliana Amato, Marco Dal Molin, Andrea Mafficini, Jun Yu, Giuseppe Malleo, Borislav Rusev, Matteo Fassan, Davide Antonello, Yoshihiko Sadakari, Paola Castelli, Giuseppe Zamboni, Anirban Maitra, Roberto Salvia, Ralph H Hruban, Claudio Bassi, Paola Capelli, Rita T Lawlor, Michael Goggins, Aldo Scarpa

Abstract

Intraductal neoplasms are important precursors to invasive pancreatic cancer and provide an opportunity to detect and treat pancreatic neoplasia before an invasive carcinoma develops. The diagnostic evaluation of these lesions is challenging, as diagnostic imaging and cytological sampling do not provide accurate information on lesion classification, the grade of dysplasia or the presence of invasion. Moreover, the molecular driver gene mutations of these precursor lesions have yet to be fully characterized. Fifty-two intraductal papillary neoplasms, including 48 intraductal papillary mucinous neoplasms (IPMNs) and four intraductal tubulopapillary neoplasms (ITPNs), were subjected to the mutation assessment in 51 cancer-associated genes, using ion torrent semiconductor-based next-generation sequencing. P16 and Smad4 immunohistochemistry was performed on 34 IPMNs and 17 IPMN-associated carcinomas. At least one somatic mutation was observed in 46/48 (96%) IPMNs; 29 (60%) had multiple gene alterations. GNAS and/or KRAS mutations were found in 44/48 (92%) of IPMNs. GNAS was mutated in 38/48 (79%) IPMNs, KRAS in 24/48 (50%) and these mutations coexisted in 18/48 (37.5%) of IPMNs. RNF43 was the third most commonly mutated gene and was always associated with GNAS and/or KRAS mutations, as were virtually all the low-frequency mutations found in other genes. Mutations in TP53 and BRAF genes (10% and 6%) were only observed in high-grade IPMNs. P16 was lost in 7/34 IPMNs and 9/17 IPMN-associated carcinomas; Smad4 was lost in 1/34 IPMNs and 5/17 IPMN-associated carcinomas. In contrast to IPMNs, only one of four ITPNs had detectable driver gene (GNAS and NRAS) mutations. Deep sequencing DNA from seven cyst fluid aspirates identified 10 of the 13 mutations detected in their associated IPMN. Using next-generation sequencing to detect cyst fluid mutations has the potential to improve the diagnostic and prognostic stratification of pancreatic cystic neoplasms.

Keywords: IPMN; biomarkers; next-generation sequencing (NGS); pancreatic tumours.

© 2014 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.

Figures

Figure 1
Figure 1
Representative haematoxylin and eosin (H&E) images and differential mucins immunolabelling of pancreatic intraductal neoplasms (original magnification = ×20). PB, pancreaticobiliary; ITPN, intraductal tubulopapillary neoplasm.
Figure 2
Figure 2
Mutational profiles of matched non-invasive and invasive IPMN components identified by ion torrent sequencing. (A) Three cases (128 fp, 129fp and 138fp) presented a non-invasive intermediate-grade (IG) or high-grade (HG) dysplastic component and an invasive adenocarcinoma (K) component, both of which were analysed using the 50-gene Ampliseq Hotspot Cancer Panel; representative H&E images from case 138fp are presented (original magnifications = ×2 and ×20). (B) For each pair of samples, the first column denotes mutations detected in IG or HG dysplastic samples and the second column represents mutations detected in cancer samples. Rows are the genes in which mutations were detected, and the orange bars represent the mutation. Seven mutations observed in the GNAS, KRAS, BRAF, KRAS and STK11 genes were common to both samples; three mutations were observed in only one of the matched lesions.
Figure 3
Figure 3
Concomitant IPMN and ITPN sharing a common GNAS mutation. Patient 130fp presented two different neoplastic lesions, an intestinal-type IPMN and an ITPN coexisting in pancreatic head. The two phenotypically different lesions showed a common GNAS R201H mutation; the ITPN component presented a NRAS Q61L mutation; representative H&E images of the lesions are shown (original magnifications = ×20). On the right of each sample is a representation of the reads aligned to the reference genome, as provided by the Integrative Genomics Viewer software (IGV v 2.1, Broad Institute) for the hotspot mutations in the GNAS and NRAS genes.
Figure 4
Figure 4
IPMN showing a KRAS mutation V14I. The intestinal-type IPMN 128fp presented a V14I KRAS mutation in both the high grade (HG) dysplastic and the invasive carcinomatous (K) component, analysed separately using the 50-gene Ampliseq Hotspot Cancer Panel; a representative H&E image of the lesions is shown (original magnification = ×20). On the right there is a representation of the reads aligned to the reference genome, as provided by the Integrative Genomics Viewer software (IGV v 2.1, Broad Institute).
Figure 5
Figure 5
TP53 mutational status corresponds to p53 protein accumulation. (A) Case 939 showed a heterogeneous pattern of staining, which is consistent with TP53 mutational status (21% of mutated alleles, I195N). (B) Case 108, with no evident p53 labelling, corresponds to a homozygous stop mutation (83% of mutated alleles, R306*). For each sample, a representative p53 immunohistochemical image (original magnification = ×20) and a representation of the reads aligned to the reference genome, as provided by the Integrative Genomics Viewer software (IGV v 2.1, Broad Institute), are presented.
Figure 6
Figure 6
P16 and Smad4 immunoexpression in IPMN. Representative images of p16 (A–D) and Smad4 (E–H) immunoreactions are shown. (A) In normal pancreatic parenchyma, p16 is expressed in Langerhans’ islets and in random acinar and duct cells. (B) A barcode-like p16 positivity in a gastric-type IPMN. (C) Strong nuclear and cytoplasmic p16 expression in an oncocytic-type IPMN. (D) p16 expression heterogeneity in a case of pancreatobiliary-type IPMN (positive and negative p16 components are evident). (E) Normal pancreas shows a strong Smad4 expression in all epithelial and stromal cell types, with stronger positivity in Langerhans’ islets. Strong Smad4 expression in intestinal-type (F) and oncocytic-type (G) IPMNs. (H) A Smad4 negative invasive carcinoma infiltrating the stroma surrounding a pancreatobiliary-type IPMN. Original magnifications = ×10 and ×20.

References

    1. Adsay NV, Fukushima N, Furukawa T. Intraductal neoplasms of the pancreas. In: Bosman FT, Carneiro F, Hruban RH, et al., editors. WHO Classification of Tumours of the Digestive System. 4th edn. Lyon: IARC; 2010.
    1. Luttges J, Zamboni G, Longnecker D, et al. The immunohistochemical mucin expression pattern distinguishes different types of intraductal papillary mucinous neoplasms of the pancreas and determines their relationship to mucinous noncystic carcinoma and ductal adenocarcinoma. Am J Surg Pathol. 2001;25::942–948.
    1. Chadwick B, Willmore-Payne C, Tripp S, et al. Histologic, immunohistochemical, and molecular classification of 52 IPMNs of the pancreas. Appl Immunohistochem Mol Morphol. 2009;17::31–39.
    1. Mohri D, Asaoka Y, Ijichi H, et al. Different subtypes of intraductal papillary mucinous neoplasm in the pancreas have distinct pathways to pancreatic cancer progression. J Gastroenterol. 2012;47::203–213.
    1. Schonleben F, Qiu W, Ciau NT, et al. PIK3CA mutations in intraductal papillary mucinous neoplasm/carcinoma of the pancreas. Clin Cancer Res. 2006;12::3851–3855.
    1. Abe K, Suda K, Arakawa A, et al. Different patterns of p16INK4A and p53 protein expressions in intraductal papillary–mucinous neoplasms and pancreatic intraepithelial neoplasia. Pancreas. 2007;34::85–91.
    1. Biankin AV, Biankin SA, Kench JG, et al. Aberrant p16(INK4A) and DPC4/Smad4 expression in intraductal papillary mucinous tumours of the pancreas is associated with invasive ductal adenocarcinoma. Gut. 2002;50::861–868.
    1. Sato N, Rosty C, Jansen M, et al. STK11 LKB1 Peutz–Jeghers gene inactivation in intraductal papillary–mucinous neoplasms of the pancreas. Am J Pathol. 2001;159::2017–2022.
    1. Schonleben F, Qiu W, Allendorf JD, et al. Molecular analysis of PIK3CA, BRAF, and RAS oncogenes in periampullary and ampullary adenomas and carcinomas. J Gastrointest Surg. 2009;13::1510–1516.
    1. Wu J, Jiao Y, Dal Molin M, et al. Whole-exome sequencing of neoplastic cysts of the pancreas reveals recurrent mutations in components of ubiquitin-dependent pathways. Proc Natl Acad Sci USA. 2011;108::21188–21193.
    1. Wu J, Matthaei H, Maitra A, et al. Recurrent GNAS mutations define an unexpected pathway for pancreatic cyst development. Sci Transl Med. 2011;3:
    1. Yamaguchi H, Kuboki Y, Hatori T, et al. Somatic mutations in PIK3CA and activation of AKT in intraductal tubulopapillary neoplasms of the pancreas. Am J Surg Pathol. 2011;35::1812–1817.
    1. 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. Am J Pathol. 2000;157::755–761.
    1. Kanda M, Sadakari Y, Borges M, et al. Mutant TP53 in duodenal samples of pancreatic juice from patients with pancreatic cancer or high-grade dysplasia. Clin Gastroenterol Hepatol. 2013;11::719–730.
    1. Sato N, Ueki T, Fukushima N, et al. Aberrant methylation of CpG islands in intraductal papillary mucinous neoplasms of the pancreas. Gastroenterology. 2002;123::365–372.
    1. Lubezky N, Ben-Haim M, Marmor S, et al. High-throughput mutation profiling in intraductal papillary mucinous neoplasm (IPMN) J Gastrointest Surg. 2011;15::503–511.
    1. Xiao HD, Yamaguchi H, Dias-Santagata D, et al. Molecular characteristics and biological behaviours of the oncocytic and pancreatobiliary subtypes of intraductal papillary mucinous neoplasms. J Pathol. 2011;224::508–516.
    1. Yamaguchi H, Kuboki Y, Hatori T, et al. The discrete nature and distinguishing molecular features of pancreatic intraductal tubulopapillary neoplasms and intraductal papillary mucinous neoplasms of the gastric type, pyloric gland variant. J Pathol. 2013;231::335–341.
    1. Scarpa A, Sikora K, Fassan M, et al. Molecular typing of lung adenocarcinoma on cytological samples using a multigene next generation sequencing panel. PLoS One. 2013;8::e80478.
    1. Beadling C, Neff TL, Heinrich MC, et al. Combining highly multiplexed PCR with semiconductor-based sequencing for rapid cancer genotyping. J Mol Diagn. 2013;15::171–176.
    1. Luchini C, Capelli P, Fassan M, et al. Next-generation histopathological diagnosis: a lesson from a hepatic carcinosarcoma. J Clin Oncol. 2014
    1. Simbolo M, Gottardi M, Corbo V, et al. DNA qualification workflow for next generation sequencing of histopathological samples. PLoS One. 2013;8::e62692.
    1. Zamo A, Bertolaso A, van Raaij AW, et al. Application of microfluidic technology to the BIOMED-2 protocol for detection of B-cell clonality. J Mol Diagn. 2012;14::30–37.
    1. Wilentz RE, Su GH, Dai JL, et al. Immunohistochemical labeling for dpc4 mirrors genetic status in pancreatic adenocarcinomas: a new marker of DPC4 inactivation. Am J Pathol. 2000;156::37–43.
    1. 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 Res. 58::4740–4744.
    1. Furukawa T, Kloppel G, Volkan Adsay N, et al. Classification of types of intraductal papillary–mucinous neoplasm of the pancreas: a consensus study. Virchows Arch. 2005;447::794–799.
    1. Nakamura A, Horinouchi M, Goto M, et al. New classification of pancreatic intraductal papillary-mucinous tumour by mucin expression: its relationship with potential for malignancy. J Pathol. 2002;197::201–210.
    1. Yonezawa S, Taira M, Osako M, et al. MUC-1 mucin expression in invasive areas of intraductal papillary mucinous tumors of the pancreas. Pathol Int. 1998;48::319–322.
    1. Fassan M, Simbolo M, Bria E, et al. High-throughput mutation profiling identifies novel molecular dysregulation in high-grade intraepithelial neoplasia and early gastric cancers. Gastric Cancer. 2013
    1. Schubbert S, Zenker M, Rowe SL, et al. Germline KRAS mutations cause Noonan syndrome. Nat Genet. 2006;38::331–336.
    1. Tyner JW, Erickson H, Deininger MW, et al. High-throughput sequencing screen reveals novel, transforming RAS mutations in myeloid leukemia patients. Blood. 2009;113::1749–1755.
    1. Furukawa T, Kuboki Y, Tanji E, et al. Whole-exome sequencing uncovers frequent GNAS mutations in intraductal papillary mucinous neoplasms of the pancreas. Sci Rep. 2011;1::161.
    1. Biankin AV, Waddell N, Kassahn KS, et al. Pancreatic cancer genomes reveal aberrations in axon guidance pathway genes. Nature. 2012;491::399–405.
    1. Jones S, Zhang X, Parsons DW, et al. Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science. 2008;321::1801–1806.
    1. Garcia-Carracedo D, Turk A, Fine S, et al. Loss of PTEN expression predicts poor prognosis in patients with intraductal papillary mucinous neoplasms of the pancreas. Clin Cancer Res. 2013;19::6830–6841.
    1. Dal Molin M, Matthaei H, Wu J, et al. Clinicopathological correlates of activating GNAS mutations in intraductal papillary mucinous neoplasm (IPMN) of the pancreas. Ann Surg Oncol. 2013;20::3802–3808.
    1. Tanaka M, Fernandez-del Castillo C, Adsay V, et al. International consensus guidelines 2012 for the management of IPMN and MCN of the pancreas. Pancreatology. 2012;12::183–197.
    1. Kinde I, Wu J, Papadopoulos N, et al. Detection and quantification of rare mutations with massively parallel sequencing. Proc Natl Acad Sci USA. 2011;108::9530–9535.

Source: PubMed

3
Sottoscrivi