Abstract
Cancer is fundamentally a genetic disease caused by the ac cumulation of somatic mutations in oncogenes and tumor suppressor genes. In the last decade, rapid advances in sequencing and bioinformatic technology led to an explosion in sequencing studies of cancer genomes, greatly expanding our knowledge of the genetic changes underlying a variety of tumor types. Several of these studies of cancer genomes have focused on pancreatic neoplasms, and cancers from the pancreas are some of the best characterized tumors at the genetic level. Pancreatic neoplasms encompass a wide array of clinical diseases, from benign cysts to deadly cancers, and the genetic alterations underlying neoplasms of the pancreas are similarly diverse. This new knowledge of pancreatic cancer genomes has deepened our understanding of tumorigenesis in the pancreas and has opened several promising new avenues for novel diagnostics and therapeutics.
Keywords: Pancreatic neoplasms, pancreatic cancer genomes
DUCTAL ADENOCARCINOMA
Pathology
Ductal adenocarcinoma is the most common malignant neoplasm of the pancreas. Grossly, these lesions are typically firm, white-yellow, poorly demarcated masses obscuring the normal pancreatic architecture. Microscopically, ductal adenocarcinoma is an invasive mucin-producing and duct-forming epithelial neoplasm with an intense stromal desmoplastic response. This desmoplastic stroma is clinically important, as it complicates diagnosis on biopsy and can be a barrier to the penetration of the tumor by chemotherapy. The neoplastic glands commonly invade nerves and vessels and in so doing spread beyond the gland. The carcinomas are graded based on the degree of differentiation of the neoplastic glands, and in general, poorly differentiated carcinomas are more aggressive than well-differentiated cases. In addition, pancreata with ductal adenocarcinoma frequently harbor intraductal noninvasive epithelial proliferations that are precursors to invasive carcinoma—these lesions, referred to as pancreatic intraepithelial neoplasia (PanIN), are graded based on architectural and cytological atypia from PanIN-1 to PanIN-3.1
Genetics
KRAS is the most frequently altered oncogene in ductal adenocarcinomas with somatic mutations clustered in specific hotspot regions in greater than 90% of these cancers.2–7KRAS encodes a small GTPase that mediates cellular signaling downstream of growth factor receptors.8,9 Rare somatic mutations have been reported in other members of this signaling pathway (such as BRAF) in pancreatic cancers (Table 1).10
TABLE 1.
Somatic Mutation Prevalence of Commonly Altered Genes in Pancreatic Neoplasms
Neoplasm | Chromosome(s) | Gene(s) | Alteration Prevalence | Gene Function |
---|---|---|---|---|
PDA | 12 | KRAS | 95% | Cell signaling (mitogen-activated protein kinase [MAPK] pathway, etc) |
9 | P16/CDKN2A | 95% | Cell cycle regulation | |
17 | TP53 | 75% | Cellular stress response | |
18 | SMAD4/DPC4 | 55% | Cell signaling (TGF-β receptor pathway) | |
IPMN | 12 | KRAS | 80% | Cell signaling (MAPK pathway etc) |
17 | RNF43 | 75% | Ubiquitin ligase | |
20 | GNAS | 60% | Cell signaling (adenylyl cyclase pathway, etc) | |
9 | P16/CDKN2A | Only in HGD/carcinoma | Cell cycle regulation | |
17 | TP53 | Only in HGD/carcinoma | Cellular stress response | |
18 | SMAD4/DPC4 | Only in HGD/carcinoma | Cell signaling (TGF-β receptor pathway) | |
3 | PIK3CA | 10% | Cell signaling (PI3K [phosphatidylinositol 3-OH kinase] pathway) | |
MCN | 12 | KRAS | 80% | Cell signaling (MAPK pathway, etc) |
17 | RNF43 | 40% | Ubiquitin ligase | |
9 | P16/CDKN2A | Only in HGD/carcinoma | Cell cycle regulation | |
17 | TP53 | Only in HGD/carcinoma | Cellular stress response | |
18 | SMAD4/DPC4 | Only in HGD/carcinoma | Cell signaling (TGF-β receptor pathway) | |
SCA | 3 | VHL | 50% | Ubiquitin ligase (HIE-1α pathway) |
SPN | 3 | CTNNB1 | 95% | Cell signaling (WNT pathway), cell adhesion |
PanNET | 11 | MEN1 | 45% | Unknown |
6/X | DAXX/ATRX | 45% | Chromatin remodeling (alternative lengthening of telomeres) | |
Multiple | mTOR pathway | 15% | Cell signaling (PI3K pathway) | |
ACC | 3 | CTNNB1 | 5% | Cell signaling (WNT pathway), cell adhesion |
5 | APC | 15% | Cell signaling (WNT pathway), cell adhesion | |
PB | 3 | CTNNB1 | 55% | Cell signaling (WNT pathway), cell adhesion |
5 | APC | 10% | Cell signaling (WNT pathway), cell adhesion | |
11 | Unknown | 85% | Unknown |
PDA indicates pancreatic ductal adenocarcinoma; ACC, acinar cell carcinoma; PB, pancreatoblastoma; HGD, high-grade dysplasia; carcinoma: invasive carcinoma; MCN, mucinous cystic neoplasm; PanNET, pancreatic neuroendocrine tumor; SCA, serous cystic neoplasm; SPN, solid-pseudopapillary neoplasm.
Several tumor suppressor genes are frequently altered in ductal adenocarcinomas. P16/CDKN2A, which encodes a protein with a crucial role in cell cycle regulation, is the most frequently altered tumor suppressor gene, with loss of protein function in greater than 90% of pancreatic cancers.5,8,11–13 This loss is mediated by several mechanisms, including intragenic mutation coupled with loss of the second allele, homozygous deletion, and promoter methylation.5,14,15 Somatic mutations in the TP53 gene occur in ~75% of ductal adenocarcinomas—these mutations occur through small intragenic mutation followed by loss of the wild-type allele.5,6,16,17 The protein encoded by TP53 plays a crucial role in the cellular stress response, and strong diffuse nuclear immunolabeling for p53 protein is associated with mutation in the TP53 gene (Fig. 1).8,16 Somatic inactivation of SMAD4/DPC4, through homozygous deletion or intragenic mutation followed by loss of the wild-type allele, occurs in approximately 55% of ductal adenocarcinomas.5,11,18–20 These mutations, which alter cellular signaling in the transforming growth factor β (TGF-β) receptor pathway, are associated with poor prognosis in ductal adenocarcinoma.8,21 Somatic alterations in other members of this signaling pathway (including TGFBR2 and ALK5) have also been reported, although they are less common than SMAD4/DPC4 mutations.22 Immunohistochemical assays for loss of Smad4 protein expression can be used as a diagnostic tool, as protein loss is correlated with gene mutation and can help distinguish adenocarcinoma from nonneoplastic pancreatic disease (Fig. 2).23
FIGURE 1.
p53 Expression in pancreatic ductal adenocarcinoma. Strong nuclear expression of the p53 protein is detectable by immunohistochemistry in the neoplastic glands of this pancreatic ductal adenocarcinoma but not in the surrounding stromal cells.
FIGURE 2.
Smad4 expression in pancreatic ductal adenocarcinoma. The neoplastic glands of this pancreatic ductal adenocarcinoma show loss of Smad4 expression by immunohistochemistry, whereas Smad4 expression is intact in the surrounding stromal cells.
Pancreatic intraepithelial neoplasias, microscopic noninvasive precursor lesions, sequentially acquire the same molecular alterations that occur in ductal adenocarcinoma. Some alterations occur early in pancreatic tumorigenesis, whereas others are limited to severely dysplastic and invasive lesions. Almost all (99%) of low-grade PanINs harbor mutations in KRAS, p16/CDKN2A, GNAS, or BRAF, whereas alterations of SMAD4/DPC4and TP53 are late events, occurring only in high-grade PanINs and invasive carcinomas.24–30 Telomere shortening is one of the most frequently occurring early events in pancreatic tumorigenesis, occurring in approximately 90% of PanIN-1A lesions.31
In addition to these frequently altered oncogenes and tumor suppressor genes, pancreatic adenocarcinomas accumulate numerous additional somatic mutations—whole-exome sequencing of pancreatic ductal adenocarcinomas revealed an average of 48 non-synonymous somatic mutations per tumor.5 These mutations occur in a variety of genes, with notable heterogeneity in the somatic alterations in each individual carcinoma. Although each individual tumor contains mutations in a unique combination of individual genes, there are 12 core cellular pathways that are altered in the majority of pancreatic carcinomas—dysregulation of these pathways (such as KRAS signaling, DNA damage control, and cell adhesion) represents a common feature of tumori-genesis in the pancreas.5 The time course of tumorigenesis can also be estimated from studies of somatic mutations in metastases and their paired primary tumors. These studies suggest a 15-year time interval between the initiating mutation and the acquisition of metastatic ability, providing a broad time window for early detection of pancreatic neoplasia.32
In addition to small intragenic mutations, large areas of genomic gain and loss also occur in pancreatic cancer, and cytogenetic analyses reveal complex karyotypes. Although some alterations (such as amplification of AKT2 or homozygous deletion of SMAD4/DPC4) target the known oncogenes and tumor suppressor genes, others involve loci whose importance in pancreatic tumorigenesis remains to be determined.33–45 In addition, ductal adenocarcinomas also contain alterations in micro-RNAs, small noncoding RNAs that negatively regulate gene expression—multiple micro-RNAs, including miR-21 and miR-221, are differentially expressed in carcinomas compared with nonneoplastic pancreas.46–49
There are several morphologic variants of ductal adeno-carcinoma, and some variants have distinct molecular features. Adenosquamous carcinoma is an uncommon variant of pancreatic carcinoma—this variant has molecular features similar to ductal adenocarcinoma, with frequent alterations in KRAS, p16/CDKN2A, TP53, and SMAD4/DPC4.50 Colloid carcinoma, which is characterized microscopically by well-differentiated neoplastic cells suspended in large extracellular pools of mucin, almost always arises in association with an intestinal-type intraductal papillary mucinous neoplasm (IPMN). Compared with ductal adenocarcinoma, colloid carcinomas have a lower prevalence of somatic mutations in KRAS (approximately 30%) and TP53 (approximately 20%).51 In addition, colloid carcinomas have a higher prevalence of somatic mutations in the oncogene GNAS, which is also frequently mutated in IPMNs.51 Hepatoid carcinoma is an extremely rare variant of pancreatic cancer that is poorly characterized at the molecular level; its genomic characteristics remain to be determined. Medullary carcinoma is another uncommon variant of pancreatic carcinoma; these carcinomas have been reported in patients with hereditary nonpolyposis colon cancer or with synchronous colorectal adenocarcinomas.52–54 Medullary carcinomas have a high prevalence of microsatellite instability and lack somatic mutations in KRAS, although oncogenic BRAF mutations have been reported in medullary carcinomas.10,53,54 The diagnosis of medullary carcinoma carries therapeutic implications; although not well studied in pancreatic medullary carcinomas, microsatellite-unstable medullary colorectal carcinomas have a better prognosis and do not respond to fluorouracil-based chemotherapy.
Undifferentiated carcinoma, another uncommon pancreatic carcinoma variant, has frequent KRAS mutations but also has frequent loss of E-cadherin protein expression.55–57 This loss is associated with E-cadherin gene (CDH1) promoter methylation in some tumors, but no somatic mutations in CDH1 have been reported.57 Molecular analyses have clarified the nature of the cell types in undifferentiated carcinoma with osteoclast-like giant cells, yet another uncommon pancreatic carcinoma variant. The atypical mononuclear cells in these neoplasms contain frequent somatic mutations in KRAS, whereas these mutations are uncommon in the osteoclast-like giant cells.58–60 Similarly, whereas the mononuclear cells variably overexpress p53, the giant cells do not.61,62 These findings support the conclusion that the atypical mononuclear cells are neoplastic, whereas the osteoclast-like giant cells are reactive—the presence of mutant KRAS DNA in the giant cells is likely due to phagocytosis of tumor DNA by these nonneoplastic cells.
Investigation of the genomic alterations in ductal adenocarcinoma has defined precursor lesions, delineated the time course for the development of pancreatic cancer, helped explain the histologic diversity of variant tumors, and has provided a genetic basis for prognostication and treatment. Moreover, as the most common neoplasm of the pancreas, the genomic features of ductal adeno-carcinoma serve as an important point of comparison for other pancreatic neoplasms.
CYSTIC NEOPLASMS OF THE PANCREAS
Intraductal Papillary Mucinous Neoplasms
Pathology
Intraductal papillary mucinous neoplasms are noninvasive papillary mucin-producing epithelial neoplasms that arise in the larger pancreatic ducts. The majority of IPMNs arise in the pancreatic head, but they can be quite large and even involve the entire pancreas. Grossly, there is dilation of the main pancreatic duct or one of its branches, and the dilated portion contains papillary projections and thick mucin. By definition, IPMNs are larger than 1 cm.63 Microscopically, IPMNs are characterized by papillary projections in the pancreatic duct system. These papillary projections are lined by mucin-secreting columnar epithelium, and there are multiple epithelial subtypes, including intestinal, gastric foveolar, pancreatobiliary, and oncocytic. Intraductal papillary mucinous neoplasms are classified by the degree of epithelial dysplasia (from low-grade to high-grade dysplasia). Approximately one third of IPMNs harbor an associated invasive adenocarcinoma; of these invasive carcinomas, half are colloid carcinomas, and half are tubular carcinomas indistinguishable for ductal adenocarcinoma arising without an IPMN. Colloid carcinomas usually are of lower stage and have a better prognosis than tubular carcinomas, and colloid carcinomas are almost universally associated with intestinal-type IPMNs.64–66 Multicentricity is common and is important for several reasons. First, patients with 1 IPMN are at risk for developing another; this means that patients should be followed up carefully for metachronous disease following the resection of an IPMN. Second, small multifocal “incipient IPMNs” are common in surgically resected pancreata with a larger IPMN. Although there is no evidence-based medicine that resecting these smaller lesions benefits the patient, it is easy for a pathologist to “over call” these at the time of intraoperative frozen section diagnosis as part of the larger IPMN, resulting in too much pancreatic parenchyma being resected.
Genetics
Intraductal papillary mucinous neoplasms contain frequent alterations in genes commonly mutated in pancreatic ductal adenocarcinoma (Table 1). Somatic mutations in the KRAS oncogene occur in 30% to 80% of IPMNs, with increasing mutation prevalence in neoplasms with high-grade dysplasia and in associated invasive carcinomas.67–73 When extremely sensitive techniques are used, 80% of IPMNs harbor KRAS mutations.51 These KRAS mutations occur in all histologic subtypes of IPMNs, and rare somatic mutations in BRAF have also been reported.68,69,71 Loss of p16 protein expression occurs in both noninvasive IPMNs and invasive carcinoma associated with IPMNs, but this loss is more prevalent in invasive carcinomas (100% of invasive carcinomas compared with 10% of noninvasive IPMNs in 1 study).74 P53 overexpression is most prevalent in areas of high-grade dysplasia and invasive carcinomas, and somatic mutations in TP53 have been reported in IPMNs with high-grade dysplasia.67,71,72,75 Loss of Smad4 expression is also a late event in IPMNs. Whereas expression is retained in the majority of noninvasive IPMNs, it is lost in approximately one third of IPMN-associated invasive carcinomas.74,76
Intraductal papillary mucinous neoplasms also contain frequent alterations in genes not involved in ductal adenocarcinoma; whole-exome sequencing revealed an average of 26 somatic mutations per IPMN, approximately half as many as in invasive ductal adenocarcinoma.77 Approximately 60% of IPMNs contain somatic mutations at a hotspot codon in the oncogene GNAS, which encodes a signaling protein that couples transmembrane receptors to their downstream signaling components.51,78 Mutations in GNAS are most prevalent in intestinal-type IPMNs.51 Inactivating somatic mutations in RNF43, which encodes an E3 ubiquitin ligase, occur in approximately 75% of IPMNs, and there is frequent loss of heterozygosity at the RNF43 locus on chromosome 17q.77 In addition, somatic mutations in PIK3CA at previously described oncogenic hotspots occur in approximately 10% of IPMNs.68,69,72 Sporadic IPMNs also rarely undergo somatic mutation in the STK11/LKB1 gene, germline alterations of which result in Peutz-Jeghers syndrome.79
Promoter hypermethylation of multiple genes has been reported in IPMNs, and this hypermethylation is more prevalent in IPMNs with associated adenocarcinoma.80 Intraductal papillary mucinous neoplasms also have altered expression of micro-RNAs, with higher expression of miR-21, miR-221, and miR-17-3p in IPMNs compared with nonmucinous pancreatic cysts.81
These many genetic alterations of IPMNs represent promising targets for the development of new screening and diagnostic assays. Mutations present in the neoplastic cells are shed into the cyst fluid and therefore can be detected in aspirated cyst fluid, although only a minority of the assayed alleles in cyst fluid samples is mutant.51,82 Still, because greater than 95% of IPMNs contain a somatic mutation in either KRAS or GNAS, molecular analyses of mutations in these genes have the potential to distinguish IPMNs from other cystic neoplasms of the pancreas based on cyst fluid samples.51
Mucinous Cystic Neoplasm
Pathology
Mucinous cystic neoplasms (MCNs) are noninvasive mucin-producing neoplasms that arise outside the large ducts of the pancreas. Mucinous cystic neoplasms occur almost exclusively in the body and tail of the pancreas and are far more common in women than in men. In contrast to IPMNs, these lesions are almost always solitary, and the cysts do not communicate with the large ducts of the pancreas. Mucinous cystic neoplasms typically are composed of multiloculated thick-walled cysts with adherent thick mucin, and they may be smooth-walled or contain papillary excrescences or mural nodules. Microscopically, there are 2 defining components: the cysts are lined by mucin-producing columnar epithelial cells with an underlying ovarian-type stroma. The cyst is often surrounded by a thick fibrous capsule. Noninvasive neoplasms are classified by the degree of epithelial dysplasia (from low-grade to high-grade dysplasia), and approximately one third of MCNs have an associated invasive adenocarcinoma. Because areas of high-grade dysplasia and even invasive carcinoma can be focal with abrupt transitions, extensive histologic sampling is required. In contrast to IPMNs, MCNs are almost always unifocal, and metachronous disease is therefore usually not a concern.
Genetics
Mucinous cystic neoplasms contain frequent alterations in genes commonly mutated in pancreatic ductal adenocarcinoma (Table 1). Somatic mutations in KRAS are common, with increasing prevalence with more severe dysplasia (30% of MCNs with low-grade dysplasia compared with 80% of MCNs with high-grade dysplasia or invasive carcinoma).70,83–85 Alterations in p16 also occur in MCNs; somatic mutation in p16/CDKN2A has been reported in an MCN with high-grade dysplasia, and promoter hypermethylation of p16/CDKN2A occurs in approximately 15% of MCNs.85,86 Aberrant p53 expression is limited to areas of high-grade dysplasia and invasive carcinoma in MCNs, and somatic mutation in TP53 has been reported in an MCN with high-grade dysplasia.84,86,87 Loss of Smad4 protein expression is associated with the transition to invasive carcinoma; Smad4 expression is usually intact in noninvasive MCNs, but a subset of invasive carcinomas shows Smad4 loss.87,88
Whole-exome sequencing of MCNs revealed an average of 16 somatic mutations per neoplasm, fewer than in both IPMNs and invasive adenocarcinomas.77 In addition to somatic mutations in KRAS and TP53, approximately 40% of MCNs contain inactivating somatic alterations in RNF43, supporting the role of this gene as a tumor suppressor in mucin-producing cystic neoplasms of the pancreas.77 In contrast to IPMNs, GNAS mutations have not been reported in MCNs.
SEROUS CYSTADENOMA
Pathology
Serous cystadenomas (SCAs) are benign cystic neoplasms lined by nonmucinous epithelium. Serous cystadenomas occur most frequently in the pancreatic body and tail. In sporadic cases, most are solitary well-demarcated masses composed of numerous small thin-walled cysts filled with clear to straw-colored watery fluid, often with a central scar. The scar may calcify, and these calcifications can be detected on computerized tomographic scans. Microscopically, SCAs are composed of numerous small cysts lined by a single layer of cuboidal cells with clear cytoplasm. This common architectural pattern is referred to as microcystic, although uncommon architectural variants exist, including macrocystic and solid variants.
Genetics
The vast majority of patients with the von Hippel-Lindau VHL syndrome, an autosomal dominant tumor predisposition syndrome caused by germline mutations in the VHL gene on chromosome 3p, develop SCAs (Table 1). These SCAs have a germline VHL mutation coupled with somatic inactivation of the second allele. Somatic inactivating mutations in VHL (point mutations coupled with loss of heterozygosity) occur in up to 50% of sporadic SCAs.77,89–91 Whole-exome sequencing of SCAs revealed 10 nonsynonymous somatic mutations per tumor, approximately half the average number of alterations in IPMNs and far fewer than in invasive adenocarcinoma.77 Serous cystadenomas lack alterations in genes frequently mutated in IPMNs and MCNs (KRAS, GNAS, RNF43, etc), further supporting the hypothesis that assays of cyst fluid mutational profile will be useful in defining cyst type.77
SOLID-PSEUDOPAPILLARY NEOPLASM
Pathology
Solid-pseudopapillary neoplasms (SPNs) are low-grade malignant neoplasms that can appear solid or cystic on imaging studies. Solid-pseudopapillary neoplasms are almost always solitary, tend to occur in young women, and are not localized to a specific region of the pancreas. Most are grossly well demarcated and have solid areas as well as areas of cystic degeneration. Microscopically, SPNs are composed of uniform poorly cohesive cells supported by delicate capillaries—these neoplastic cells often microscopically infiltrate the adjacent nonneoplastic pancreas in a distinct “insidious” pattern.92 The microscopic architecture is a mix of solid areas and degenerative areas with characteristic pseudopapillae in which poorly cohesive cells drop away, leaving a thin layer of neoplastic cells surrounding a small blood vessel.
Genetics
Somatic activating mutations in the β-catenin gene (CTNNB1) occur in 95% of SPNs, leading to abnormal nuclear localization of the β-catenin protein in almost 100% of cases (Table 1).93–96 The aberrant nuclear localization of β-catenin can be used as a diagnostic aid, as immunohistochemical labeling for β-catenin can be used to distinguish SPNs from other cellular pancreatic neoplasms (Fig. 3). Moreover, alterations in CTNNB1 are associated with changes in E-cadherin protein expression.96,97 These neoplasms do not label with antibodies to the extracellular domain of E-cadherin, whereas immunolabeling with antibodies to the intracellular domain produces an abnormal nuclear pattern of labeling.98 This aberrant E-cadherin immunolabeling can be used clinically to support a diagnosis of SPN and may explain the poorly cohesive nature of the neoplastic cells. Solid-pseudopapillary neoplasms lack alterations in genes frequently altered in pancreatic ductal adenocarcinoma (KRAS, TP53, etc) and in the other cystic neoplasms of the pancreas (GNAS, RNF43, etc). By contrast, most of the other pancreatic neoplasms lack mutations in CTNNB1.93,99,100 Recently, whole-exome sequencing of SPNs revealed an average of only 3 nonsynonymous mutations per SPN, the lowest number of any pancreatic neoplasm sequenced to date; only CTNNB1 was altered in more than 1 SPN.77
FIGURE 3.
β-Catenin expression in SPN. A, The neoplastic cells of this SPN show aberrant nuclear localization of β-catenin by immunohistochemistry. B, Adjacent nonneoplastic cells show membranous β-catenin labeling by immunohistochemistry.
PANCREATIC NEUROENDOCRINE TUMORS
Pathology
Pancreatic neuroendocrine tumors (PanNETs) are solid cellular neoplasms that are composed of neoplastic cells with neuroendocrine differentiation. Pancreatic neuroendocrine tumors can occur anywhere in the pancreas, although some functional types have specific locations (e.g., gastrinomas and somatostatinomas in the duodenum). Although patients with tumor predisposition syndromes such as multiple endocrine neoplasia 1 (MEN1) frequently have multiple PanNETs, sporadic PanNETs are usually solitary well-demarcated solid masses. Degenerative areas can occur in large neoplasms. Microscopically, PanNETs consist of organoid proliferations of cells with the cytological features of neuroendocrine cells, including granular cytoplasm and “salt and pepper” chromatin. Numerous architectural patterns have been described, including trabecular, nested, and gyriform. Grade is a major prognosticator, and PanNETs are graded based on proliferative rate, as assessed by mitotic count or Ki-67 labeling index, with high grade (grade 3) equivalent to neuroendocrine carcinoma. There are 2 histologic subtypes of neuroendocrine carcinoma: small cell carcinoma and large cell carcinoma.
Genetics
Whole-exome sequencing of PanNETs revealed an average of 16 nonsynonymous somatic mutations per tumor, far fewer than in pancreatic ductal adenocarcinomas (Table 1).101 The somatic mutations in PanNETs are distinct from those in the other neoplasms of the pancreas. Approximately 45% of sporadic PanNETs have somatic mutations in MEN1, and loss of heterozygosity at the MEN1 locus occurs in 30% to 70% of sporadic PanNETs.101–106 Mutations in MEN1 are associated with improved prognosis.101 In addition, genes involved in a chromatin remodeling complex (DAXX and ATRX) are somatically altered in approximately 45% of sporadic PanNETs.101 Mutations in these genes are inactivating and result in loss of protein expression as assayed by immunohistochemistry (Fig. 4).101 The proteins encoded by DAXX and ATRX act in a complex that plays a crucial role in telomere maintenance, and neoplasms with DAXX and ATRX mutations exhibit the ALT (alternative lengthening of telomeres) phenotype, a telomerase-independent mechanism of telomere maintenance.107 These somatic mutations in DAXX and ATRX occur late in the development of PanNETs, as DAXX and ATRX mutations occur only in large tumors and are absent from microadenomas.108 Somatic mutations in genes coding for proteins in the mammalian target of rapamycin (mTOR) cell signaling pathway occur in approximately 15% of PanNETs, including somatic mutations in PIK3CA, PTEN, and TSC2.101 The alterations in the mTOR pathway may carry clinical significance, as drugs targeting this signaling pathway have been developed for clinical use.109 Promoter hypermethylation and deletion of VHL occur in up to 25% of sporadic PanNETs, and numerous large chromosomal gains and losses have been described.110–114 Pancreatic neuroendocrine tumors lack frequent somatic alterations in genes commonly mutated in pancreatic ductal adenocarcinoma (KRAS,TP53, etc).99,101
FIGURE 4.
ATRX expression in PanNET. The neoplastic cells of this PanNET show loss of ATRX expression by immunohistochemistry, whereas ATRX expression is retained in the adjacent endothelial cells.
ACINAR CELL CARCINOMA
Pathology
Acinar cell carcinomas are rare solid cellular neoplasms composed of cells with acinar differentiation. Acinar cell carcinoma can occur anywhere in the pancreas and frequently forms a large solitary well-circumscribed solid mass. Microscopically, these neoplasms are composed of cells with architectural, histologic, or immunohistochemical evidence of acinar differentiation; acinar architecture, cytoplasmic granules, and nuclei with single prominent nucleoli are common features.
Genetics
Somatic alterations in the APC/β-catenin pathway occur in 20% to 25% of acinar cell carcinomas, including activating mutations in CTNNB1 and inactivating mutations in APC (Table 1).115 These neoplasms lack frequent alterations in genes commonly mutated in pancreatic ductal adenocarcinoma, although rare alterations in KRAS and TP53 have been reported.99,115–119 Promoter methylation of several tumor suppressor genes has been reported in acinar cell carcinomas, but the functional consequences of these alterations remain to be explored.117 Most acinar cell carcinomas also harbor numerous large chromosomal gains and losses, although no specific target genes have been identified.44,119,120
PANCREATOBLASTOMA
Pathology
Pancreatoblastomas are rare solid cellular neoplasms that occur most often in children, although cases in adults can also occur. Pancreatoblastomas are not associated with a specific location in the pancreas and usually form a solitary well-demarcated solid fleshy mass. Microscopically, pancreatoblastomas are composed of multiple cellular components; the acinar component is usually predominant, and squamoid nests are a characteristic feature, but neuroendocrine, ductal, and primitive components may also be present.
Genetics
Allelic loss of chromosome 11p is frequent in pancreatoblastomas (86% of cases in 1 study) (Table 1).121,122 Loss of this chromosome arm has also been reported in other embryonal neoplasms, suggesting the possibility of a common genetic pathway in embryonal neoplasms.123 The majority of pancreatoblastomas have somatic alterations in the APC/β-catenin pathway, including activating mutations in CTNNB1 as well as inactivating mutations in APC.121 Pancreatoblastomas lack alterations in genes commonly mutated in ductal adenocarcinoma (KRAS, TP53, etc).121 Complex karyotypes have been reported in multiple cases.124–126
CONCLUSIONS
Neoplasms of the pancreas are currently classified based on morphologic and immunohistochemical features. Molecular alterations often parallel these features, suggesting that molecular alterations can aid in diagnoses. For example, each of the 4 major cyst types of the pancreas has its own mutational profile, and this profile can be defined by analysis of cyst fluid. Knowledge of the molecular alterations underlying neoplasms of the pancreas is also likely to lead to significant improvements in the treatment of these neoplasms. For example, PanNETs with alterations of the mTOR pathway may be particularly responsive to mTOR inhibitors. As we enter the genomic era, effective clinical care of patients with pancreatic tumors will require integration of molecular analyses into the current standard of care. Moreover, future scientific breakthroughs are likely to continually transform our knowledge of the genomic features of pancreatic neoplasia, leading to clinical advances that improve the lives of patients with pancreatic neoplasms.
Footnotes
Conflicts of Interest and Sources of Funding: R.H.H. receives royalty payments from Myriad Genetics for the PalB2 invention. L.D.W. declares no conflict of interest.
REFERENCES
- 1.Hruban RH, Adsay NV, Albores-Saavedra J, et al. Pancreatic intraepithelial neoplasia: a new nomenclature and classification system for pancreatic duct lesions. Am J Surg Pathol. 2001;25:579–586. doi: 10.1097/00000478-200105000-00003. [DOI] [PubMed] [Google Scholar]
- 2.Smit VT, Boot AJ, Smits AM, et al. KRAS codon 12 mutations occur very frequently in pancreatic adenocarcinomas. Nucleic Acids Res. 1988;16:7773–7782. doi: 10.1093/nar/16.16.7773. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Almoguera C, Shibata D, Forrester K, et al. Most human carcinomas of the exocrine pancreas contain mutant c-K-ras genes. Cell. 1988;53:549–554. doi: 10.1016/0092-8674(88)90571-5. [DOI] [PubMed] [Google Scholar]
- 4.Caldas C, Kern SE. K-ras mutation and pancreatic adenocarcinoma. Int J Pancreatol. 1995;18:1–6. doi: 10.1007/BF02825415. [DOI] [PubMed] [Google Scholar]
- 5.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. doi: 10.1126/science.1164368. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Pellegata NS, Sessa F, Renault B, et al. K-ras and p53 gene mutations in pancreatic cancer: ductal and nonductal tumors progress through different genetic lesions. Cancer Res. 1994;54:1556–1560. [PubMed] [Google Scholar]
- 7.Hruban RH, van Mansfeld AD, Offerhaus GJ, et al. K-ras oncogene activation in adenocarcinoma of the human pancreas. A study of 82 carcinomas using a combination of mutant-enriched polymerase chain reaction analysis and allele-specific oligonucleotide hybridization. Am J Pathol. 1993;143:545–554. [PMC free article] [PubMed] [Google Scholar]
- 8.Vogelstein B, Kinzler KW. Cancer genes and the pathways they control. Nat Med. 2004;10:789–799. doi: 10.1038/nm1087. [DOI] [PubMed] [Google Scholar]
- 9.Vakiani E, Solit DB. KRAS and BRAF: drug targets and predictive biomarkers. J Pathol. 2011;223:219–229. doi: 10.1002/path.2796. [DOI] [PubMed] [Google Scholar]
- 10.Calhoun ES, Jones JB, Ashfaq R, et al. BRAF and FBXW7 (CDC4, FBW7, AGO, SEL10) mutations in distinct subsets of pancreatic cancer: potential therapeutic targets. Am J Pathol. 2003;163:1255–1260. doi: 10.1016/S0002-9440(10)63485-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Maitra A, Hruban RH. Pancreatic cancer. Annu Rev Pathol. 2008;3:157–188. doi: 10.1146/annurev.pathmechdis.3.121806.154305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Attri J, Srinivasan R, Majumdar S, et al. Alterations of tumor suppressor gene p16INK4a in pancreatic ductal carcinoma. BMC Gastroenterol. 2005;5:22. doi: 10.1186/1471-230X-5-22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Li J, Poi MJ, Tsai MD. Regulatory mechanisms of tumor suppressor P16(INK4A) and their relevance to cancer. Biochemistry. 2011;50:5566–5582. doi: 10.1021/bi200642e. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Caldas C, Hahn SA, da Costa LT, et al. Frequent somatic mutations and homozygous deletions of the p16 (MTS1) gene in pancreatic adenocarcinoma. Nat Genet. 1994;8:27–32. doi: 10.1038/ng0994-27. [DOI] [PubMed] [Google Scholar]
- 15.Schutte M, Hruban RH, Geradts J, et al. Abrogation of the Rb/p16 tumor-suppressive pathway in virtually all pancreatic carcinomas. Cancer Res. 1997;57:3126–3130. [PubMed] [Google Scholar]
- 16.Scarpa A, Capelli P, Mukai K, et al. Pancreatic adenocarcinomas frequently show p53 gene mutations. Am J Pathol. 1993;142:1534–1543. [PMC free article] [PubMed] [Google Scholar]
- 17.Redston MS, Caldas C, Seymour AB, et al. p53 Mutations in pancreatic carcinoma and evidence of common involvement of homocopolymer tracts in DNA microdeletions. Cancer Res. 1994;54:3025–3033. [PubMed] [Google Scholar]
- 18.Hahn SA, Schutte M, Hoque AT, et al. DPC4, a candidate tumor suppressor gene at human chromosome 18q21.1. Science. 1996;271:350–353. doi: 10.1126/science.271.5247.350. [DOI] [PubMed] [Google Scholar]
- 19.Hahn SA, Hoque AT, Moskaluk CA, et al. Homozygous deletion map at 18q21.1 in pancreatic cancer. Cancer Res. 1996;56:490–494. [PubMed] [Google Scholar]
- 20.Iacobuzio-Donahue CA, Song J, Parmiagiani G, et al. Missense mutations of MADH4: characterization of the mutational hot spot and functional consequences in human tumors. Clin Cancer Res. 2004;10:1597–1604. doi: 10.1158/1078-0432.ccr-1121-3. [DOI] [PubMed] [Google Scholar]
- 21.Blackford A, Serrano OK, Wolfgang CL, et al. SMAD4 gene mutations are associated with poor prognosis in pancreatic cancer. Clin Cancer Res. 2009;15:4674–4679. doi: 10.1158/1078-0432.CCR-09-0227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Goggins M, Shekher M, Turnacioglu K, et al. Genetic alterations of the transforming growth factor beta receptor genes in pancreatic and biliary adenocarcinomas. Cancer Res. 1998;58:5329–5332. [PubMed] [Google Scholar]
- 23.Tascilar M, Offerhaus GJ, Altink R, et al. Immunohistochemical labeling for the Dpc4 gene product is a specific marker for adenocarcinoma in biopsy specimens of the pancreas and bile duct. Am J Clin Pathol. 2001;116:831–837. doi: 10.1309/WF03-NFCE-7BRH-7C26. [DOI] [PubMed] [Google Scholar]
- 24.Moskaluk CA, Hruban RH, Kern SE. p16 and K-ras gene mutations in the intraductal precursors of human pancreatic adenocarcinoma. Cancer Res. 1997;57:2140–2143. [PubMed] [Google Scholar]
- 25.Lohr M, Kloppel G, Maisonneuve P, et al. Frequency of K-ras mutations in pancreatic intraductal neoplasias associated with pancreatic ductal adenocarcinoma and chronic pancreatitis: a meta-analysis. Neoplasia. 2005;7:17–23. doi: 10.1593/neo.04445. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Shi C, Hong SM, Lim P, et al. KRAS2 mutations in human pancreatic acinar-ductal metaplastic lesions are limited to those with PanIN: implications for the human pancreatic cancer cell of origin. Mol Cancer Res. 2009;7:230–236. doi: 10.1158/1541-7786.MCR-08-0206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Kanda M, Matthaei H, Wu J, et al. Presence of somatic mutations in most early-stage pancreatic intraepithelial neoplasia. Gastroenterology. 2012;142:730–733. e9. doi: 10.1053/j.gastro.2011.12.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.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 Res. 2000;60:2002–2006. [PubMed] [Google Scholar]
- 29.Maitra A, Adsay NV, Argani P, et al. Multicomponent analysis of the pancreatic adenocarcinoma progression model using a pancreatic intraepithelial neoplasia tissue microarray. Mod Pathol. 2003;16:902–912. doi: 10.1097/01.MP.0000086072.56290.FB. [DOI] [PubMed] [Google Scholar]
- 30.Luttges J, Galehdari H, Brocker V, et al. Allelic loss is often the first hit in the biallelic inactivation of the p53 and DPC4 genes during pancreatic carcinogenesis. Am J Pathol. 2001;158:1677–1683. doi: 10.1016/S0002-9440(10)64123-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.van Heek NT, Meeker AK, Kern SE, et al. Telomere shortening is nearly universal in pancreatic intraepithelial neoplasia. Am J Pathol. 2002;161:1541–1547. doi: 10.1016/S0002-9440(10)64432-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Yachida S, Jones S, Bozic I, et al. Distant metastasis occurs late during the genetic evolution of pancreatic cancer. Nature. 2010;467:1114–1117. doi: 10.1038/nature09515. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Ruggeri BA, Huang L, Wood M, et al. Amplification and overexpression of the AKT2 oncogene in a subset of human pancreatic ductal adenocarcinomas. Mol Carcinog. 1998;21:81–86. [PubMed] [Google Scholar]
- 34.Griffin CA, Hruban RH, Morsberger LA, et al. Consistent chromo-some abnormalities in adenocarcinoma of the pancreas. Cancer Res. 1995;55:2394–2399. [PubMed] [Google Scholar]
- 35.Aguirre AJ, Brennan C, Bailey G, et al. High-resolution characterization of the pancreatic adenocarcinoma genome. Proc Natl Acad Sci U S A. 2004;101:9067–9072. doi: 10.1073/pnas.0402932101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Heidenblad M, Schoenmakers EF, Jonson T, et al. Genome-wide array-based comparative genomic hybridization reveals multiple amplification targets and novel homozygous deletions in pancreatic carcinoma cell lines. Cancer Res. 2004;64:3052–3059. doi: 10.1158/0008-5472.can-03-3159. [DOI] [PubMed] [Google Scholar]
- 37.Iacobuzio-Donahue CA, van der Heijden MS, Baumgartner MR, et al. Large-scale allelotype of pancreaticobiliary carcinoma provides quantitative estimates of genome-wide allelic loss. Cancer Res. 2004;64:871–875. doi: 10.1158/0008-5472.can-03-2756. [DOI] [PubMed] [Google Scholar]
- 38.Mahlamaki EH, Kauraniemi P, Monni O, et al. High-resolution genomic and expression profiling reveals 105 putative amplification target genes in pancreatic cancer. Neoplasia. 2004;6:432–439. doi: 10.1593/neo.04130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Bashyam MD, Bair R, Kim YH, et al. Array-based comparative genomic hybridization identifies localized DNA amplifications and homozygous deletions in pancreatic cancer. Neoplasia. 2005;7:556–562. doi: 10.1593/neo.04586. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Chen S, Auletta T, Dovirak O, et al. Copy number alterations in pancreatic cancer identify recurrent PAK4 amplification. Cancer Biol Ther. 2008;7:1793–1802. doi: 10.4161/cbt.7.11.6840. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Doldan A, Chandramouli A, Shanas R, et al. Loss of the eukaryotic initiation factor 3f in pancreatic cancer. Mol Carcinog. 2008;47:235–244. doi: 10.1002/mc.20379. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Kwei KA, Bashyam MD, Kao J, et al. Genomic profiling identifies GATA6 as a candidate oncogene amplified in pancreatobiliary cancer. PLoS Genet. 2008;4:e1000081. doi: 10.1371/journal.pgen.1000081. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Nomoto S, Kinoshita T, Mori T, et al. Adverse prognosis of epigenetic inactivation in RUNX3 gene at 1p36 in human pancreatic cancer. Br J Cancer. 2008;98:1690–1695. doi: 10.1038/sj.bjc.6604333. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Dewald GW, Smyrk TC, Thorland EC, 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;84:801–810. doi: 10.4065/84.9.801. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Birnbaum DJ, Adelaide J, Mamessier E, et al. Genome profiling of pancreatic adenocarcinoma. Genes Chromosomes Cancer. 2011;50:456–465. doi: 10.1002/gcc.20870. [DOI] [PubMed] [Google Scholar]
- 46.Dillhoff M, Liu J, Frankel W, et al. MicroRNA-21 is overexpressed in pancreatic cancer and a potential predictor of survival. J Gastrointest Surg. 2008;12:2171–2176. doi: 10.1007/s11605-008-0584-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.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;26:4442–4452. doi: 10.1038/sj.onc.1210228. [DOI] [PubMed] [Google Scholar]
- 48.Bloomston M, Frankel WL, Petrocca F, et al. MicroRNA expression patterns to differentiate pancreatic adenocarcinoma from normal pancreas and chronic pancreatitis. JAMA. 2007;297:1901–1908. doi: 10.1001/jama.297.17.1901. [DOI] [PubMed] [Google Scholar]
- 49.Lee EJ, Gusev Y, Jiang J, et al. Expression profiling identifies microRNA signature in pancreatic cancer. Int J Cancer. 2007;120:1046–1054. doi: 10.1002/ijc.22394. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Brody JR, Costantino CL, Potoczek M, et al. Adenosquamous carcinoma of the pancreas harbors KRAS2, DPC4 and TP53 molecular alterations similar to pancreatic ductal adenocarcinoma. Mod Pathol. 2009;22:651–659. doi: 10.1038/modpathol.2009.15. [DOI] [PubMed] [Google Scholar]
- 51.Wu J, Matthaei H, Maitra A, et al. Recurrent GNAS mutations define an unexpected pathway for pancreatic cyst development. Sci Transl Med. 2011;3:92ra66. doi: 10.1126/scitranslmed.3002543. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Banville N, Geraghty R, Fox E, et al. Medullary carcinoma of the pancreas in a man with hereditary nonpolyposis colorectal cancer due to a mutation of the MSH2 mismatch repair gene. Hum Pathol. 2006;37:1498–1502. doi: 10.1016/j.humpath.2006.06.024. [DOI] [PubMed] [Google Scholar]
- 53.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+ Am J Pathol. 1998;152:1501–1507. [PMC free article] [PubMed] [Google Scholar]
- 54.Wilentz RE, Goggins M, Redston M, et al. Genetic, immunohistochemical, and clinical features of medullary carcinoma of the pancreas: a newly described and characterized entity. Am J Pathol. 2000;156:1641–1651. doi: 10.1016/S0002-9440(10)65035-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Hoorens A, Prenzel K, Lemoine NR, et al. Undifferentiated carcinoma of the pancreas: analysis of intermediate filament profile and Ki-ras mutations provides evidence of a ductal origin. J Pathol. 1998;185:53–60. doi: 10.1002/(SICI)1096-9896(199805)185:1<53::AID-PATH45>3.0.CO;2-F. [DOI] [PubMed] [Google Scholar]
- 56.Paal E, Thompson LD, Frommelt RA, et al. A clinicopathologic and immunohistochemical study of 35 anaplastic carcinomas of the pancreas with a review of the literature. Ann Diagn Pathol. 2001;5:129–140. doi: 10.1053/adpa.2001.25404. [DOI] [PubMed] [Google Scholar]
- 57.Winter JM, Ting AH, Vilardell F, et al. Absence of E-cadherin expression distinguishes noncohesive from cohesive pancreatic cancer. Clin Cancer Res. 2008;14:412–418. doi: 10.1158/1078-0432.CCR-07-0487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Sakai Y, Kupelioglu AA, Yanagisawa A, et al. Origin of giant cells in osteoclast-like giant cell tumors of the pancreas. Hum Pathol. 2000;31:1223–1229. doi: 10.1053/hupa.2000.18491. [DOI] [PubMed] [Google Scholar]
- 59.Gocke CD, Dabbs DJ, Benko FA, et al. KRAS oncogene mutations suggest a common histogenetic origin for pleomorphic giant cell tumor of the pancreas, osteoclastoma of the pancreas, and pancreatic duct adenocarcinoma. Hum Pathol. 1997;28:80–83. doi: 10.1016/s0046-8177(97)90283-5. [DOI] [PubMed] [Google Scholar]
- 60.Westra WH, Sturm P, Drillenburg P, et al. K-ras oncogene mutations in osteoclast-like giant cell tumors of the pancreas and liver: genetic evidence to support origin from the duct epithelium. Am J Surg Pathol. 1998;22:1247–1254. doi: 10.1097/00000478-199810000-00010. [DOI] [PubMed] [Google Scholar]
- 61.Deckard-Janatpour K, Kragel S, Teplitz RL, et al. Tumors of the pancreas with osteoclast-like and pleomorphic giant cells: an immunohistochemical and ploidy study. Arch Pathol Lab Med. 1998;122:266–272. [PubMed] [Google Scholar]
- 62.Molberg KH, Heffess C, Delgado R, et al. Undifferentiated carcinoma with osteoclast-like giant cells of the pancreas and periampullary region. Cancer. 1998;82:1279–1287. doi: 10.1002/(sici)1097-0142(19980401)82:7<1279::aid-cncr10>3.0.co;2-3. [DOI] [PubMed] [Google Scholar]
- 63.Hruban RH, Takaori K, Klimstra DS, et al. An illustrated consensus on the classification of pancreatic intraepithelial neoplasia and intraductal papillary mucinous neoplasms. Am J Surg Pathol. 2004;28:977–987. doi: 10.1097/01.pas.0000126675.59108.80. [DOI] [PubMed] [Google Scholar]
- 64.Sohn TA, Yeo CJ, Cameron JL, et al. Intraductal papillary mucinous neoplasms of the pancreas: an updated experience. Ann Surg. 2004;239:788–797. doi: 10.1097/01.sla.0000128306.90650.aa. discussion 97–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Seidel G, Zahurak M, Iacobuzio-Donahue C, et al. Almost all infiltrating colloid carcinomas of the pancreas and periampullary region arise from in situ papillary neoplasms: a study of 39 cases. Am J Surg Pathol. 2002;26:56–63. doi: 10.1097/00000478-200201000-00006. [DOI] [PubMed] [Google Scholar]
- 66.Adsay NV, Pierson C, Sarkar F, et al. Colloid (mucinous noncystic) carcinoma of the pancreas. Am J Surg Pathol. 2001;25:26–42. doi: 10.1097/00000478-200101000-00003. [DOI] [PubMed] [Google Scholar]
- 67.Satoh K, Shimosegawa T, Moriizumi S, et al. K-ras mutation and p53 protein accumulation in intraductal mucin-hypersecreting neoplasms of the pancreas. Pancreas. 1996;12:362–368. doi: 10.1097/00006676-199605000-00007. [DOI] [PubMed] [Google Scholar]
- 68.Schonleben F, Allendorf JD, Qiu W, et al. Mutational analyses of multiple oncogenic pathways in intraductal papillary mucinous neoplasms of the pancreas. Pancreas. 2008;36:168–172. doi: 10.1097/MPA.0b013e318158a4d2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Schonleben F, Qiu W, Remotti HE, et al. PIK3CA, KRAS, and BRAF mutations in intraductal papillary mucinous neoplasm/carcinoma (IPMN/C) of the pancreas. Langenbecks Arch Surg. 2008;393:289–296. doi: 10.1007/s00423-008-0285-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Yoshizawa K, Nagai H, Sakurai S, et al. Clonality and K-ras mutation analyses of epithelia in intraductal papillary mucinous tumor and mucinous cystic tumor of the pancreas. Virchows Arch. 2002;441:437–443. doi: 10.1007/s00428-002-0645-6. [DOI] [PubMed] [Google Scholar]
- 71.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. doi: 10.1097/PAI.0b013e31817c02c6. [DOI] [PubMed] [Google Scholar]
- 72.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. doi: 10.1007/s11605-010-1411-8. [DOI] [PubMed] [Google Scholar]
- 73.Z'Graggen K, Rivera JA, Compton CC, et al. Prevalence of activating K-ras mutations in the evolutionary stages of neoplasia in intraductal papillary mucinous tumors of the pancreas. Ann Surg. 1997;226:491–498. doi: 10.1097/00000658-199710000-00010. discussion 8–500. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.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. doi: 10.1136/gut.50.6.861. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Kawahira H, Kobayashi S, Kaneko K, et al. p53 Protein expression in intraductal papillary mucinous tumors (IPMT) of the pancreas as an indicator of tumor malignancy. Hepatogastroenterology. 2000;47:973–977. [PubMed] [Google Scholar]
- 76.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. doi: 10.1016/S0002-9440(10)64589-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.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 U S A. 2011;108:21188–21193. doi: 10.1073/pnas.1118046108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Weinstein LS, Liu J, Sakamoto A, et al. Minireview: GNAS: normal and abnormal functions. Endocrinology. 2004;145:5459–5464. doi: 10.1210/en.2004-0865. [DOI] [PubMed] [Google Scholar]
- 79.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. doi: 10.1016/S0002-9440(10)63053-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.House MG, Guo M, Iacobuzio-Donahue C, et al. Molecular progression of promoter methylation in intraductal papillary mucinous neoplasms (IPMN) of the pancreas. Carcinogenesis. 2003;24:193–198. doi: 10.1093/carcin/24.2.193. [DOI] [PubMed] [Google Scholar]
- 81.Ryu JK, Matthaei H, Dal Molin M, et al. Elevated microRNA miR-21 levels in pancreatic cyst fluid are predictive of mucinous precursor lesions of ductal adenocarcinoma. Pancreatology. 2011;11:343–350. doi: 10.1159/000329183. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Schoedel KE, Finkelstein SD, Ohori NP. K-Ras and microsatellite marker analysis of fine-needle aspirates from intraductal papillary mucinous neoplasms of the pancreas. Diagn Cytopathol. 2006;34:605–608. doi: 10.1002/dc.20511. [DOI] [PubMed] [Google Scholar]
- 83.Yanagisawa A, Kato Y, Ohtake K, et al. c-Ki-ras point mutations in ductectatic-type mucinous cystic neoplasms of the pancreas. Jpn J Cancer Res. 1991;82:1057–1060. doi: 10.1111/j.1349-7006.1991.tb01756.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Jimenez RE, Warshaw AL, Z'Graggen K, et al. Sequential accumulation of K-ras mutations and p53 overexpression in the progression of pancreatic mucinous cystic neoplasms to malignancy. Ann Surg. 1999;230:501–509. doi: 10.1097/00000658-199910000-00006. discussion 9–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Kim SG, Wu TT, Lee JH, et al. Comparison of epigenetic and genetic alterations in mucinous cystic neoplasm and serous microcystic adenoma of pancreas. Mod Pathol. 2003;16:1086–1094. doi: 10.1097/01.MP.0000094088.37888.A6. [DOI] [PubMed] [Google Scholar]
- 86.Sorio C, Capelli P, Lissandrini D, et al. Mucinous cystic carcinoma of the pancreas: a unique cell line and xenograft model of a preinvasive lesion. Virchows Arch. 2005;446:239–245. doi: 10.1007/s00428-004-1167-1. [DOI] [PubMed] [Google Scholar]
- 87.Luttges J, Feyerabend B, Buchelt T, et al. The mucin profile of noninvasive and invasive mucinous cystic neoplasms of the pancreas. Am J Surg Pathol. 2002;26:466–471. doi: 10.1097/00000478-200204000-00008. [DOI] [PubMed] [Google Scholar]
- 88.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. Am J Surg Pathol. 2000;24:1544–1548. doi: 10.1097/00000478-200011000-00011. [DOI] [PubMed] [Google Scholar]
- 89.Vortmeyer AO, Lubensky IA, Fogt F, et al. Allelic deletion and mutation of the von Hippel-Lindau (VHL) tumor suppressor gene in pancreatic microcystic adenomas. Am J Pathol. 1997;151:951–956. [PMC free article] [PubMed] [Google Scholar]
- 90.Moore PS, Zamboni G, Brighenti A, et al. Molecular characterization of pancreatic serous microcystic adenomas: evidence for a tumor suppressor gene on chromosome 10q. Am J Pathol. 2001;158:317–321. doi: 10.1016/S0002-9440(10)63971-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Mohr VH, Vortmeyer AO, Zhuang Z, et al. Histopathology and molecular genetics of multiple cysts and microcystic (serous) adenomas of the pancreas in von Hippel-Lindau patients. Am J Pathol. 2000;157:1615–1621. doi: 10.1016/S0002-9440(10)64799-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Meriden Z, Shi C, Edil BH, et al. Hyaline globules in neuroendocrine and solid-pseudopapillary neoplasms of the pancreas: a clue to the diagnosis. Am J Surg Pathol. 2011;35:981–988. doi: 10.1097/PAS.0b013e31821a9a14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Abraham SC, Klimstra DS, Wilentz RE, 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;160:1361–1369. doi: 10.1016/s0002-9440(10)62563-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Min Kim S, Sun CD, Park KC, et al. Accumulation of beta-catenin protein, mutations in exon-3 of the beta-catenin gene and a loss of heterozygosity of 5q22 in solid pseudopapillary tumor of the pancreas. J Surg Oncol. 2006;94:418–425. doi: 10.1002/jso.20509. [DOI] [PubMed] [Google Scholar]
- 95.Tanaka Y, Kato K, Notohara K, et al. Frequent beta-catenin mutation and cytoplasmic/nuclear accumulation in pancreatic solid-pseudopapillary neoplasm. Cancer Res. 2001;61:8401–8404. [PubMed] [Google Scholar]
- 96.Audard V, Cavard C, Richa H, et al. Impaired E-cadherin expression and glutamine synthetase overexpression in solid pseudopapillary neoplasm of the pancreas. Pancreas. 2008;36:80–83. doi: 10.1097/mpa.0b013e318137a9da. [DOI] [PubMed] [Google Scholar]
- 97.El-Bahrawy MA, Rowan A, Horncastle D, et al. E-cadherin/catenin complex status in solid pseudopapillary tumor of the pancreas. Am J Surg Pathol. 2008;32:1–7. doi: 10.1097/PAS.0b013e31813e0676. [DOI] [PubMed] [Google Scholar]
- 98.Chetty R, Serra S. Membrane loss and aberrant nuclear localization of E-cadherin are consistent features of solid pseudopapillary tumour of the pancreas. An immunohistochemical study using two antibodies recognizing different domains of the E-cadherin molecule. Histopathology. 2008;52:325–330. doi: 10.1111/j.1365-2559.2007.02949.x. [DOI] [PubMed] [Google Scholar]
- 99.Moore PS, Orlandini S, Zamboni G, et al. Pancreatic tumours: molecular pathways implicated in ductal cancer are involved in ampullary but not in exocrine nonductal or endocrine tumorigenesis. Br J Cancer. 2001;84:253–262. doi: 10.1054/bjoc.2000.1567. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Gerdes B, Ramaswamy A, Simon B, et al. Analysis of beta-catenin gene mutations in pancreatic tumors. Digestion. 1999;60:544–548. doi: 10.1159/000007704. [DOI] [PubMed] [Google Scholar]
- 101.Jiao Y, Shi C, Edil BH, et al. DAXX/ATRX, MEN1, and mTOR pathway genes are frequently altered in pancreatic neuroendocrine tumors. Science. 2011;331:1199–1203. doi: 10.1126/science.1200609. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Hessman O, Lindberg D, Skogseid B, et al. Mutation of the multiple endocrine neoplasia type 1 gene in nonfamilial, malignant tumors of the endocrine pancreas. Cancer Res. 1998;58:377–379. [PubMed] [Google Scholar]
- 103.Gortz B, Roth J, Krahenmann A, 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 fore-gut neoplasms. Am J Pathol. 1999;154:429–436. doi: 10.1016/S0002-9440(10)65289-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Zhuang Z, Vortmeyer AO, Pack S, et al. Somatic mutations of the MEN1 tumor suppressor gene in sporadic gastrinomas and insulinomas. Cancer Res. 1997;57:4682–4686. [PubMed] [Google Scholar]
- 105.Toliat MR, Berger W, Ropers HH, et al. Mutations in the MEN I gene in sporadic neuroendocrine tumours of gastroenteropancreatic system. Lancet. 1997;350:1223. doi: 10.1016/S0140-6736(05)63453-8. [DOI] [PubMed] [Google Scholar]
- 106.Wang EH, Ebrahimi SA, Wu AY, et al. Mutation of the MENIN gene in sporadic pancreatic endocrine tumors. Cancer Res. 1998;58:4417–4420. [PubMed] [Google Scholar]
- 107.Heaphy CM, de Wilde RF, Jiao Y, et al. Altered telomeres in tumors with ATRX and DAXX mutations. ScienceM. 2011;333:425. doi: 10.1126/science.1207313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.de Wilde RF, Heaphy CM, Maitra A, 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 doi: 10.1038/modpathol.2012.53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Yao JC, Shah MH, Ito T, et al. Everolimus for advanced pancreatic neuroendocrine tumors. N Engl J Med. 2011;364:514–523. doi: 10.1056/NEJMoa1009290. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Schmitt AM, Schmid S, Rudolph T, et al. VHL inactivation is an important pathway for the development of malignant sporadic pancreatic endocrine tumors. Endocr Relat Cancer. 2009;16:1219–1227. doi: 10.1677/ERC-08-0297. [DOI] [PubMed] [Google Scholar]
- 111.Floridia G, Grilli G, Salvatore M, et al. Chromosomal alterations detected by comparative genomic hybridization in nonfunctioning endocrine pancreatic tumors. Cancer Genet Cytogenet. 2005;156:23–30. doi: 10.1016/j.cancergencyto.2004.04.015. [DOI] [PubMed] [Google Scholar]
- 112.Jonkers YM, Claessen SM, Perren A, et al. Chromosomal instability predicts metastatic disease in patients with insulinomas. Endocr Relat Cancer. 2005;12:435–447. doi: 10.1677/erc.1.00960. [DOI] [PubMed] [Google Scholar]
- 113.Chung DC, Brown SB, Graeme-Cook F, et al. Localization of putative tumor suppressor loci by genome-wide allelotyping in human pancreatic endocrine tumors. Cancer Res. 1998;58:3706–3711. [PubMed] [Google Scholar]
- 114.Rigaud G, Missiaglia E, Moore PS, et al. High resolution allelotype of nonfunctional pancreatic endocrine tumors: identification of two molecular subgroups with clinical implications. Cancer Res. 2001;61:285–292. [PubMed] [Google Scholar]
- 115.Abraham SC, Wu TT, Hruban RH, 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;160:953–962. doi: 10.1016/s0002-9440(10)64917-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Hoorens A, Lemoine NR, McLellan E, et al. Pancreatic acinar cell carcinoma. An analysis of cell lineage markers, p53 expression, and Ki-ras mutation. Am J Pathol. 1993;143:685–698. [PMC free article] [PubMed] [Google Scholar]
- 117.de Wilde RF, Ottenhof NA, Jansen M, et al. Analysis of LKB1 mutations and other molecular alterations in pancreatic acinar cell carcinoma. Mod Pathol. 2011;24:1229–1236. doi: 10.1038/modpathol.2011.83. [DOI] [PubMed] [Google Scholar]
- 118.Terhune PG, Memoli VA, Longnecker DS. Evaluation of p53 mutation in pancreatic acinar cell carcinomas of humans and transgenic mice. Pancreas. 1998;16:6–12. doi: 10.1097/00006676-199801000-00002. [DOI] [PubMed] [Google Scholar]
- 119.Rigaud G, Moore PS, Zamboni G, et al. Allelotype of pancreatic acinar cell carcinoma. Int J Cancer. 2000;88:772–777. doi: 10.1002/1097-0215(20001201)88:5<772::aid-ijc14>3.0.co;2-w. [DOI] [PubMed] [Google Scholar]
- 120.Taruscio D, Paradisi S, Zamboni G, et al. Pancreatic acinar carcinoma shows a distinct pattern of chromosomal imbalances by comparative genomic hybridization. Genes Chromosomes Cancer. 2000;28:294–299. doi: 10.1002/1098-2264(200007)28:3<294::aid-gcc7>3.0.co;2-n. [DOI] [PubMed] [Google Scholar]
- 121.Abraham SC, Wu TT, Klimstra DS, 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;159:1619–1627. doi: 10.1016/s0002-9440(10)63008-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Kerr NJ, Fukuzawa R, Reeve AE, et al. Beckwith-Wiedemann syndrome, pancreatoblastoma, and the wnt signaling pathway. Am J Pathol. 2002;160:1541–1542. doi: 10.1016/s0002-9440(10)62580-1. author reply 2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Ruteshouser EC, Hendrickson BW, Colella S, et al. Genome-wide loss of heterozygosity analysis of WT1-wild-type and WT1-mutant Wilms tumors. Genes Chromosomes Cancer. 2005;43:172–180. doi: 10.1002/gcc.20169. [DOI] [PubMed] [Google Scholar]
- 124.Barenboim-Stapleton L, Yang X, Tsokos M, et al. Pediatric pancreatoblastoma: histopathologic and cytogenetic characterization of tumor and derived cell line. Cancer Genet Cytogenet. 2005;157:109–117. doi: 10.1016/j.cancergencyto.2004.05.017. [DOI] [PubMed] [Google Scholar]
- 125.Nagashima Y, Misugi K, Tanaka Y, et al. Pancreatoblastoma: a second report on cytogenetic findings. Cancer Genet Cytogenet. 1999;109:178–179. doi: 10.1016/s0165-4608(98)00169-1. [DOI] [PubMed] [Google Scholar]
- 126.Wiley J, Posekany K, Riley R, et al. Cytogenetic and flow cytometric analysis of a pancreatoblastoma. Cancer Genet Cytogenet. 1995;79:115–118. doi: 10.1016/0165-4608(94)00117-t. [DOI] [PubMed] [Google Scholar]