Elsevier

Pancreatology

Volume 15, Issue 6, November–December 2015, Pages 598-610
Pancreatology

Review article
Pathology, genetics and precursors of human and experimental pancreatic neoplasms: An update

https://doi.org/10.1016/j.pan.2015.08.007Get rights and content

Abstract

Over the past decade, there have been substantial improvements in our knowledge of pancreatic neoplasms and their precursor lesions. Extensive genetic analyses, recently using high-throughput molecular techniques and next-generation sequencing methodologies, and the development of sophisticated genetically engineered mouse models closely recapitulating human disease, have improved our understanding of the genetic basis of pancreatic neoplasms. These advances are paving the way for refined, molecular-based classifications of pancreatic neoplasms with the potential to better predict prognosis and, possibly, response to therapy. Another major development resides in the identification of subsets of pancreatic exocrine and endocrine neoplasms which occur in the context of hereditary syndromes and whose genetic basis and tumor development have been at least partially defined. However, despite all molecular progress, correct and careful morphological characterization of tissue specimens both in the context of experimental and routine diagnostic pathology represents the basis for any further genetic investigation or clinical decision. This review focuses on the current and new concepts of classification and on the current models of tumor development, both in the field of exocrine and endocrine neoplasms, and underscores the importance of applying standardized terminology to allow adequate data interpretation and promote scientific exchange in the field of pancreas research.

Introduction

The pancreas is an organ with dual function exerted by an exocrine and endocrine cell compartment. Thus, the neoplasms arising therefrom display different phenotypes in terms of morphology and biology, and are driven by distinct genetic alterations. In order to improve the prognosis and treatment options, especially for those tumors with unfavorable outcome, it is essential to identify precursor lesions and their driving molecular alterations. With this knowledge we will improve our understanding of the tumors' natural history, we may then better select the patients who profit from existing therapeutic approaches and might finally define new therapeutic targets.

The development of sophisticated genetically engineered mouse models that closely recapitulate human disease, together with the extensive use of high-throughput molecular techniques and next-generation sequencing methodologies have improved our understanding of the genetic basis of pancreatic neoplasms and may prepare our way for clinic-translational approaches. The bottom line for these approaches is the consistent use of established, internationally accepted classification systems and a unanimous nomenclature both in research and routine pathology.

The 2010 World Health Organization classification [1] of pancreatic tumors is widely accepted. It separates the tumors in epithelial (including exocrine and neuroendocrine tumors) and non-epithelial neoplasms and subtypes them according to their biological behavior in benign, premalignant and malignant (Table 1).

Human neoplasms can be further classified in sporadic, familial and hereditary. A neoplasm occurring in individuals who do not have a germline mutation that confers increased susceptibility to cancer or without a family history of cancer is defined as sporadic. Cancer that occurs in families more often than would be expected by chance and usually at an earlier age compared to the general population is defined as familial. Shared environmental or lifestyle factors play a role in familial cancer. If a germline mutation predisposes to cancer development, the term hereditary neoplasm is used (for definitions see also www.cancer.gov/dictionary).

Among the pancreatic neoplasms the sporadic epithelial non-neuroendocrine tumors are most frequent and account for about 90%. They display either an acinar or ductal cell phenotype. In addition, there are some rare tumors whose phenotypic appearance is not attributable to one of the known cell types. Genetically, the neoplasms with a ductal phenotype usually harbor KRAS mutations (“KRAS-positive”), while those with other phenotypes such as the acinar cell carcinomas, neuroendocrine neoplasms, serous cystic neoplasms and solid pseudopapillary neoplasms have a different genetic profile. In this review, the pancreatic neoplasms and their precursors are classified according to their KRAS-status and their cellular phenotype (see Table 2). In addition, their counterparts in genetically engineered mice are briefly described for comparison. Finally, we present a conceptual approach to the development of pancreatic neoplasms.

Section snippets

KRAS-positive neoplasms with ductal phenotype

Pancreatic ductal adenocarcinoma (PDAC) is a highly malignant tumor that histologically imitates small and medium-sized pancreatic ducts. PDAC affects usually old and only rarely patients younger than 40 years, occurs slightly more in men than women and is the most common pancreatic neoplasm (85–90%) [2], [3], [4]. For the last reason, the term “pancreatic cancer” is often used as synonymous to PDAC. However, since the pancreas gives also rise to other types of “cancers” distinct from PDAC, the

KRAS-negative neoplasms with acinar cell phenotype

Acinar cell carcinoma (ACC) is a rare neoplasm (<2% of pancreatic malignancies) that can occur anywhere in the pancreas and typically forms a large well-demarcated solid and nodular, and only rarely cystic, mass (Fig. 2A). Men are more frequently affected than women (mean age 55–65 years). The tumor cells produce pancreatic enzymes, notably trypsin and lipase that can be detected by immunohistochemistry (Fig. 2B) [2], [39]. Interestingly, some ACCs may contain substantial proportions (>30%) of

KRAS-negative neoplasms with uncertain phenotype and various genetic changes

Solid-pseudopapillary neoplasm (SPN) is a low-grade malignant tumor that typically affects young women. It presents as a solitary, well-circumscribed mass, which often undergoes pseudocystic hemorrhagic degeneration. For this reason, SPNs are usually included among the cystic pancreatic neoplasms. Microscopically, SCNs are composed of monomorphic epithelial cells forming solid and pseudopapillary structures.

Genetically, 95% of all SPNs harbor somatic activating mutations in the β-catenin gene (

KRAS-negative neoplasms with neuroendocrine phenotype

Neuroendocrine neoplasms (NENs) account for 1–2% of all pancreatic tumors. They affect both genders evenly and may occur at a wide range of age (30–60 years). The WHO classification distinguishes two main groups: the well-differentiated neuroendocrine tumors (PanNETs) and the poorly-differentiated neuroendocrine carcinomas (PanNECs). These subtypes are characterized by their histological differentiation and proliferative activity, assessed by mitotic count or preferably by the Ki-67 index.

Familial/hereditary pancreatic neoplasms

PDAC are reported in approximately 5–10% of patients with a family history of pancreatic cancer [82]. The terms hereditary and familial pancreatic cancer have been frequently interchangeably used in the literature, sometimes causing confusion among their meaning and definition. While the term “hereditary pancreatic cancer” refers to pancreatic cancers arising in patients with recognized genetic syndromes (Table 4), “familial pancreatic cancer” (FPC) defines patients with PDAC having at least

Comparative pathology in animal models of pancreatic neoplasms

PanIN PDAC. First approaches to mimic human PDAC in animal models focused on chemical induction of PDAC. These include tumors induced by N-nitrosobis(2-oxopropyl)amine (BOP) in syrian hamsters [100] and by 9,10-dimethyl-1,2-benzanthracene (DMBA) in rat and mice and exhibit close histological and genetic similarities to their human counterparts (recently reviewed by Murtaugh [101]). The first GEMM that successfully recapitulated the human disease was introduced by Hingorani et al., in 2003 [102]

Conclusions

Recent progress in defining the detailed pathological features of pancreatic tumors and their precursor lesions in correlation with underlying molecular changes and experimental models has enormously improved our knowledge about tumor development in the pancreas. These improvements may help in defining biomarkers of early disease, predicting prognosis, detecting targets for medical treatment and acquire new insights into tumorigenesis. Fig. 5 summarizes our conceptual approach on the origin of

Acknowledgments

This work was supported by the COST action BM1204 “EU Pancreas: An integrated European platform for pancreas cancer research: from basic science to clinical and public health interventions for a rare disease” and was written on behalf of the members of working group 1 (eupancreas.com).

References (134)

  • R.E. Wilentz et al.

    Genetic, immunohistochemical, and clinical features of medullary carcinoma of the pancreas: a newly described and characterized entity

    Am J Pathol

    (2000)
  • S. La Rosa et al.

    Improved histologic and clinicopathologic criteria for prognostic evaluation of pancreatic endocrine tumors

    Hum Pathol

    (2009)
  • A. Agaimy et al.

    ISL1 expression is not restricted to pancreatic well-differentiated neuroendocrine neoplasms, but is also commonly found in well and poorly differentiated neuroendocrine neoplasms of extrapancreatic origin

    Mod Pathol Off J U S Can Acad Pathol Inc

    (2013)
  • R.F. de Wilde 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 Off J U S Can Acad Pathol Inc

    (2012)
  • R.H. Hruban et al.

    Update on familial pancreatic cancer

    Adv Surg

    (2010)
  • L.C. Murtaugh et al.

    A case of mistaken identity? Nonductal origins of pancreatic “ductal” cancers

    Cancer Cell

    (2007)
  • D.E. Bockman et al.

    Origin and development of the precursor lesions in experimental pancreatic cancer in rats

    Laboratory Investig J Tech Methods Pathol

    (2003)
  • I.A. Lubensky et al.

    Multiple neuroendocrine tumors of the pancreas in von Hippel-Lindau disease patients: histopathological and molecular genetic analysis

    Am J Pathol

    (1998)
  • O. Mete et al.

    Precursor lesions of endocrine system neoplasms

    Pathology

    (2013)
  • F.T. Bosman et al.

    Theise ND WHO classification of tumours of the digestive system

  • D.S. Klimstra et al.

    Tumors of the exocrine pancreas

  • G. Klöppel et al.

    Pathology of exocrine pancreatic tumors

    (2008)
  • F. Bergmann et al.

    Molecular characterisation of pancreatic ductal adenocarcinoma in patients under 40

    J Clin Pathol

    (2006)
  • I. Esposito et al.

    Most pancreatic cancer resections are R1 resections

    Ann Surg Oncol

    (2008)
  • W. Hartwig et al.

    Pancreatic cancer surgery in the new millennium: better prediction of outcome

    Ann Surg

    (2011)
  • A.M. Schlitter et al.

    Definition of microscopic tumor clearance (r0) in pancreatic cancer resections

    Cancers

    (2010)
  • N. Waddell et al.

    Whole genomes redefine the mutational landscape of pancreatic cancer

    Nature

    (2015)
  • E.A. Collisson et al.

    Subtypes of pancreatic ductal adenocarcinoma and their differing responses to therapy

    Nat Med

    (2011)
  • S. Jones et al.

    Core signaling pathways in human pancreatic cancers revealed by global genomic analyses

    Science

    (2008)
  • R.H. Hruban et al.

    An illustrated consensus on the classification of pancreatic intraepithelial neoplasia and intraductal papillary mucinous neoplasms

    Am J Surg Pathol

    (2004)
  • C. Shi et al.

    Increased prevalence of precursor lesions in familial pancreatic cancer patients

    Clin Cancer Res Off J Am Assoc Cancer Res

    (2009)
  • J.B. Koorstra et al.

    Morphogenesis of pancreatic cancer: role of pancreatic intraepithelial neoplasia (PanINs)

    Langenbeck's Arch Surgery/Deutsche Gesellschaft fur Chir

    (2008)
  • M. Kanda et al.

    Presence of somatic mutations in most early-stage pancreatic intraepithelial neoplasia

    Gastroenterology

    (2012)
  • T. Furukawa et al.

    Prognostic relevance of morphological types of intraductal papillary mucinous neoplasms of the pancreas

    Gut

    (2011)
  • L.D. Wood et al.

    Pathology and molecular genetics of pancreatic neoplasms

    Cancer J

    (2012)
  • M.C. Tan et al.

    GNAS and KRAS mutations define separate progression pathways in intraductal papillary mucinous neoplasm-associated carcinoma

    J Am Coll Surg

    (2015)
  • A.V. Biankin 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)
  • B. Chadwick et al.

    Histologic, immunohistochemical, and molecular classification of 52 IPMNs of the pancreas

    Appl Immunohistochem Mol Morphol AIMM/Off Publ Soc Appl Immunohistochem

    (2009)
  • J. Wu et al.

    Whole-exome sequencing of neoplastic cysts of the pancreas reveals recurrent mutations in components of ubiquitin-dependent pathways

    Proc Natl Acad Sci U S A

    (2011)
  • M. Dal Molin et al.

    Clinicopathological correlates of activating GNAS mutations in intraductal papillary mucinous neoplasm (IPMN) of the pancreas

    Ann Surg Oncol

    (2013)
  • W. Hosoda et al.

    GNAS mutation is a frequent event in pancreatic intraductal papillary mucinous neoplasms and associated adenocarcinomas

    Virchows Arch

    (2015)
  • N. Ideno et al.

    Clinical significance of GNAS mutation in intraductal papillary mucinous neoplasm of the pancreas with concomitant pancreatic ductal adenocarcinoma

    Pancreas

    (2015)
  • M.G. House et al.

    Molecular progression of promoter methylation in intraductal papillary mucinous neoplasms (IPMN) of the pancreas

    Carcinogenesis

    (2003)
  • D. Mohri et al.

    Different subtypes of intraductal papillary mucinous neoplasm in the pancreas have distinct pathways to pancreatic cancer progression

    J Gastroenterol

    (2012)
  • K.E. Schoedel et al.

    K-Ras and microsatellite marker analysis of fine-needle aspirates from intraductal papillary mucinous neoplasms of the pancreas

    Diagn Cytopathol

    (2006)
  • J. Wu et al.

    Recurrent GNAS mutations define an unexpected pathway for pancreatic cyst development

    Sci Transl Med

    (2011)
  • A. Schlitter et al.

    Klassifikation und diagnose zystischer Pankreastumoren

    Viszeralmedizin

    (2011)
  • M. Distler et al.

    Precursor lesions for sporadic pancreatic cancer: PanIN, IPMN, and MCN

    BioMed Res Int

    (2014)
  • H. Matthaei et al.

    Cystic precursors to invasive pancreatic cancer

    Nat Rev Gastroenterol Hepatol

    (2011)
  • Cited by (0)

    1

    Equally contributing first authors.

    View full text