Introduction
Pancreatic cancer is the fourth leading cause of cancer death in the United States 1,2. Approximately 32 000 individuals in the USA and over 200 000 individuals worldwide die from the disease each year 1,2,3. The incidence approximates the mortality rate, which reflects the poor prognosis of pancreatic cancer. Although there have been many advances in pancreatic cancer research, the 5-year survival rate for affected patients remains under 5% 2.
The aggressiveness that characterizes pancreatic cancer arises from multiple heterogeneous genetic changes that occur before the onset of clinical symptoms. Studies performed over the past decade have shed some light on the molecular and histological events that are associated with pancreatic carcinogenesis. This chapter will focus on the genetics and pathology of pancreatic ductal adenocarcinoma. Future progress in this area will hopefully lead to improved diagnostic tests, early detection, and new treatments for patients who suffer from this devastating disease.
Genetics
The accumulation of multiple nonrandom genetic changes over time is a hallmark of pancreatic cancer. Genetic abnormalities include alterations in chromosome or gene copy number, microsatellite instability, epigenetic silencing, intragenic point mutations, and gene overexpression secondary to increased transcription (Table I) 4.
Table I. Overview of the genetic abnormalities described in this paper.
| Abnormalities observed in pancreatic ductal adenocarcinoma | Example |
|---|---|
| Chromosome alterations | Losses: 9p, 17p, 18q, 3p, 8p, 6q Gains: 3q, 5p, 7p, 8q, 20q |
| Microsatellite instability | Medullary carcinoma with mutations in hMLH1 |
| Epigenetic silencing | Hypermethylation of promoter CpG islands in ppENK |
| Mutations | p16, p53, DPC4 |
| Gene overexpression | Mesothelin |
Chromosomal alterations
The most common techniques used to study chromosome losses and gains are karyotyping (G-banding), comparative genomic hybridization (CGH), and allelotyping 4. Metaphase spread karyotyping (G-banding) is a cytogenetic technique, developed in the 1960s, where chromosomes are stained and their banding patterns are examined during metaphase. Karyotyping can reveal large deletions, insertions, translocations, inversions, and other rearrangements, but suffers from a lack of submicroscopic resolution. Conventional CGH has an improved resolution (5–10 mB) 5, but in the post-genomic era, even this technology has become arcane. Array-based CGH, a molecular cytogenetic technique developed in the early 1990s, is an application of CGH using microarray technology. In microarray analysis, thousands of molecules may be immobilized on an insoluble solid support. This strategy enables the simultaneous detection of a large number of analytes 6. A second application of microarray technology, gene expression analysis, will be discussed in greater detail below. The general principles of microarray analysis 7, applicable to both CGH and global gene expression studies, are illustrated in Figure 1.
Figure 1. .
Microarray technology is based on high throughput competitive hybridization of fluorescently labeled sample DNA and control DNA (target sequences) with an organized array of sequences deposited on a biochip (probe sequences). Molecular cytogenetics (e.g. CGH) and gene expression analysis (e.g. cDNA microarray) are two widely used applications of microarray technology. The cartoon is based on a schematic from Coe and Antler 7.
As demonstrated in Figure 1, fragments of tumor DNA and non-tumor DNA are differentially labeled with fluorescent dye and allowed to compete for hybridization sites in either a metaphase chromosome spread (conventional CGH) or in an array of genomic probes (array CGH). The color and intensity of the fluorescence emitted by the hybridized fragments of labeled DNA reveal areas of the genome that may be deleted or amplified in the test sample relative to the control sample 5,8. In allelotyping analysis, microsatellite markers (short tandem repeats of DNA that are interspersed throughout the genome) in normal and cancer DNA from a single patient are PCR amplified and sequenced. Since individuals are often heterozygous at microsatellite loci, genomic deletions can be detected by evaluating microsatellite markers in tumor DNA for losses of heterozygosity (LOH). Chromosome positions that have frequent LOH in pancreatic cancer often harbor important tumor suppressor genes 9,10,11.
Chromosome losses are more common than chromosome gains in pancreatic cancer 4. According to allelotyping data in pancreatic cancer xenografts, DNA loss in xenografts ranges from 1.6% to 32% of the genome 10. Despite this variability, certain reproducible patterns have been observed. The most common regions of genomic loss in primary pancreatic cancers and pancreatic cancer xenografts include chromosome arms 9p, 17p, 18q, 3p, 8p, and 6q 10,12,13. Some of these loci contain known tumor suppressor genes such as CDKN2A/P16/MTS1 (9p21), p53 (17p13), and MADH4/SMAD4/DPC4 (18q). Further investigations may reveal additional tumor suppressor genes in the other LOH ‘hot spots’ 10.
Frequent gains of DNA, secondary to unbalanced chromosome rearrangements, have been observed in primary pancreatic cancers on chromosome arms 3q, 5p, 7p, 8q, and 20q 12,14. Fluorescent in situ hybridization (FISH) performed on pancreatic cancer cell lines demonstrates amplified regions of DNA at 19q, 12p, 12q, 17q, and 20q. These amplified sites correspond to the locations of the following oncogenes: AKT2, KRAS2, MDM2, ERBB2, and AIB1, respectively 13,15.
Microsatellite instability
Medullary carcinoma, a subtype of pancreatic adenocarcinoma seen in approximately 5% of operative cases, often contains a defective DNA mismatch repair (MMR) mechanism 16. Altered MMR gives rise to microsatellite instability (MSI), characterized by hypermutability at so-called minisatellite repeat sequences. Unlike most ductal adenocarcinomas of the pancreas, MSI tumors show minimal LOH and aneuploidy 17. They also frequently lack mutations in KRAS2 and P53. Histologically, medullary cancers are poorly differentiated. They grow in a syncytial pattern, exhibit pushing borders, and demonstrate extensive necrosis. Interestingly, these tumors appear to have a better prognosis than the more common ductal adenocarcinoma 16.
Epigenetic silencing
Epigenetic phenomena are heritable DNA modifications that do not involve alterations in DNA sequence 18. Examples include structural changes to chromatin, such as post-translation histone modifications and nucleosome rearrangements, and deregulation of cytosine methylation at promoter CpG islands. The expression of many tumor suppressor genes and oncogenes appears to be influenced through this mechanism. Genes that are silenced in pancreatic cancer by epigenetic changes include ppENK (90%), RARB (20%), CDKN2A/P16 (18%), CACNA1G (16%), TIMP3 (11%), CDH1 (7%), THBBS1 (7%), and hMLH1 (4%) 19,20. Approximately 60% of pancreatic cancers have multiple genes that are silenced by hypermethylation of promoter cytosines 20.
Mutations and specific gene alterations
Specific genes that play an important role in carcinogenesis can be classified as tumor suppressor genes, oncogenes, or genes involved in genomic maintenance. Many tumor suppressors regulate cell proliferation. Therefore, biallelic inactivation of a tumor suppressor gene can provide a growth advantage for tumors. Gene loss may result from allelic deletion, intragenic mutation, chromosomal recombination, epigenetic phenomena, or a combination thereof. Conversely, oncogenes promote cell growth. Genetic alterations that activate oncogenes include gain-of-function mutations, translocations, amplifications, and gene overexpression. Table II provides a list of genes that affect pancreatic carcinogenesis through intragenic mutations 21.
Table II. Genes that may be mutated in pancreatic cancer.
| Gene | Chromosome location | Percentage of pancreatic cancers with mutations |
|---|---|---|
| Oncogenes | ||
| KRAS2 | 12p | 95 |
| BRAF | 7q | 3 |
| Tumor suppressors | ||
| p16 | 9p | 95 |
| p53 | 17p | 75 |
| DPC4 | 18q | 55 |
| LKB1/STK11 | 19p | 5 |
| MKK4 | 17p | 4 |
| TGFBR2 | 3p | 4 |
| ACVR2 | 2q | 4 |
| ACVR1B | 12q | 2 |
| TGFBR1 | 9q | 1 |
| Genome maintenance | ||
| BRCA2 | 13q | 7–10 |
| FANCC | 9q | 6 |
| FANCG | 9p | 3 |
| MLH1 | 3p | 3 |
Table modified from Sohn 21.
CDKN2A/p16/MTS1, located on chromosome arm 9p, is the most frequently inactivated tumor suppressor gene in pancreatic cancer (95%). The gene can be inactivated through homozygous deletion (40%), single allele loss combined with an intragenic mutation in the second allele (40%), or promoter hypermethylation (15%) 22,23. p16 prevents Rb-1 inhibition by CDK4-phosphorylation during G1 of the cell cycle (Figure 2). When p16 is inactivated, phosphorylation of Rb-1 by G1-CDKs increases, facilitating progression through the G1/S-cell cycle checkpoint 24.
Figure 2. .
p16 and p53 regulation of the cell cycle. In normal cells, cyclin D complexes with Cdk4 during G1 of the cell cycle. This complex inhibits Rb by phosphorylating it, allowing the cell to progress into S-phase. p16 prevents cell proliferation during stress by interfering with the cyclin D-Cdk complex 24. p53 responds to DNA damage by inducing p21. Activated p21 binds and inhibits G1 and G1/S-Cdks (Cdk2), causing the cell to arrest in G1. p53 prevents entry into mitosis through the induction of 14-3-3σ, p21, and Gadd45. These proteins inactivate M-Cdks (Cdk1, Cdc2) 27. Inactivation of p16 or p53 in pancreatic cancer promotes cell tumorigenesis.
p53, located on chromsome arm 17p, is the second most commonly inactivated tumor suppressor gene in pancreatic cancer (50–75%) 25,26. Usually, one p53 allele is deleted while the other allele acquires an intragenic mutation 25. Following DNA damage to normal cells, activated p53 causes the cell to arrest in G1 or G2 of the cell cycle (Figure 2) 27. p53 also responds to genetic injury by activating apoptotic pathways. Thus, p53 inactivation allows the tumor to proliferate despite the continued accumulation of other genetic defects.
MADH4/SMAD4/DPC4, located on chromosome arm 18q, is the third most frequently inactivated tumor suppressor gene in pancreatic cancer (55%). Approximately 30% of pancreatic cancers have homozygous deletions at the SMAD4 locus, and 25% have allelic loss with an intragenic mutation in the second allele 28. SMAD4 is a downstream component of the transforming growth factor (TGF)-β and activin signaling pathways (discussed below, Figure 3). Like p16, loss of SMAD4 impairs the cell cycle check mechanism during the G1/S transition. SMAD4 loss is also believed to affect pro-apoptotic signaling 29.
Figure 3. .
Signaling pathways in pancreas development. Genes that maintain epithelial progenitor cells, and have oncogenic properties in pancreatic cancer, appear on the left of the figure. Genes that stimulate endocrine differentiation, and have tumor suppressor properties in pancreatic cancer, appear on the right of the figure. The role of three specific signaling pathways in pancreatic development are summarized. (a) Shh signaling inhibits endocrine differentiation. (b) Notch signaling inhibits endocrine differentiation. (c) TGF-β and activin signaling promote endocrine differentiation. Modified from Kim et al 85.
Mutations have been reported in additional components of the TGF-β and activin signaling pathways, such as TGFβR1 (1% homozygous deletion rate), TGFβR2 (4%), ACVR1B (2%), and ACVR2 (4%) 30,31. Rare mutations have also been identified in genes outside of the TGF-β and activin pathways. MKK4, a component of the mitogen activated protein kinase pathway involved in stress and cytokine-induced signaling, has a 4% mutation rate in pancreatic cancer 32. STK11/LKB1, a serine/threonine kinase mutated in Peutz-Jeghers syndrome, is also mutated in 4% of pancreatic cancers 33. Recent studies demonstrate that STK11 is important to the maintenance of intracellular polarity and intracellular energy metabolism 34. BRCA2, a genomic maintenance gene in the Fanconi anemia (FA) pathway, is altered in 7% of pancreatic cancers 35, but germline mutations can be seen in a larger proportion of patients (17%) that cluster in pancreatic cancer families (see below) 36. FANCC and FANCG are two other components in the FA pathway that have been shown to be mutated in pancreatic cancer (∼5%) 37. FA signaling directs homologous recombination after double-strand DNA breaks.
KRAS2, located on chromosome arm 12p, is the most commonly mutated oncogene in pancreatic cancer (90%) 38,39,40. In the majority of cases, aspartic acid is substituted for glycine at codon 12. Additional mutational ‘hot spots’ occur in codons 13 and 61 41. KRAS2 functions as a GTPase involved in intracellular signaling. Gain-of-function mutations that impair KRAS2 GTPase activity augment downstream signaling and increase cell proliferation. Pancreatic cancers with wild-type KRAS2 may have mutations in BRAF (3%) 42. BRAF is a serine/threonine kinase and a key downstream effector of KRAS2.
Gene overexpression
Altered gene expression in tumors can result from chromosomal rearrangements, epigenetic silencing, or mutations in genes that are upstream of a target gene. Gene expression may be examined one gene at a time, using techniques like Northern blot analysis, real-time PCR, and mRNA in situ hybridization 43. However, recently developed techniques such as serial analysis of gene expression (SAGE) and microarray analysis permit simultaneous evaluation of virtually the entire human transcriptome 43. In SAGE analysis, mRNA is reverse transcribed into cDNA, and then cleaved close to the poly A tail (3’ end). These steps produce 10–14 base pair fragments, or tags, that correspond to the transcripts in the original sample RNA pool. The tags are linked together, amplified, and sequenced. The prevalence of each tag in the amplified product corresponds to the expression level of a unique transcript in the tumor RNA library 43. Data from the target pool can be compared to data from a control RNA library to identify changes in gene expression. An advantage of the SAGE technique is that genomes do not have to be characterized before analysis 43.
Gene expression microarrays, like genomic microarray analysis (i.e. CGH), involve hybridization reactions between labeled targets and a library of probes (either cDNA or oligonucleotides) that are adhered to a biochip. However, in the case of gene expression analysis, the labeled targets are made from cDNA instead of genomic fragments. Therefore, the readout provides expression data rather than cytogenetic data. cDNA microarray analysis follows the general scheme for microarray experiments illustrated in Figure 1. Specifically, the RNA from tumor and non-tumor cells is differentially labeled with fluorescent dye during cDNA synthesis. The target cDNAs are allowed to hybridize to the cDNA probes on the biochip in a competitive fashion. The colors and intensities of the hybridization products are analyzed to determine the level of gene expression in the tumor compared to the reference sample 7,44. In oligonucleotide microarray analysis (trademarked as GeneChip by Affymetrix), probes that are 25 nucleotides long are synthesized on a biochip, one nucleotide at a time. Labeled tumor cRNA, synthesized through sequential reverse transcription and in vitro transcription steps, is allowed to hybridize to the chip in a non-competitive fashion. The intensity of the signals on the GeneChip corresponds to the level of transcript present in the tumor RNA sample. These data can be compared to the signal produced by the hybridization of reference cRNA with a separate, but identical, gene chip 7. Gene expression microarray analysis is perhaps better suited than SAGE analysis for high-throughput investigations, but the technique requires prior characterization of a group of genes in order to synthesize a library of probes.
Global gene expression technologies have identified hundreds of overexpressed genes in pancreatic ductal adenocarcinoma 45,46,47,48,49,50,51,52,53. Our group at Johns Hopkins identified 422 overexpressed genes using SAGE, cDNA microarrays, and U133 oligonucleotide arrays 45. Forty genes were identified by more than one method, and six genes were detected by all three methods. The six genes detected by each technique are keratin 19, transglutaminase 2, stratifin retinoic acid-induced 3, secretory leukocyte protease inhibitor, and tetraspan1. Increased expression of the first three genes listed was observed in earlier studies using antibody detection or mass spectrometry 54,55,56.
Familial pancreatic cancer
Focused investigations on high-risk families have yielded new insights into the genetics of pancreatic cancer. Registries, such as the National Familial Pancreas Tumor Registry (NFPTR) at Johns Hopkins, track patients and family members at risk for pancreatic cancer 57,58. Inherited mutations may account for 2–10% of all pancreatic cancers 58,59,60,61. Individuals with one, two, or three first degree relatives (parent, child, or sibling) with a history of pancreatic cancer demonstrate risk increases for pancreatic cancer that are approximately 6-, 18-, and 57-fold above baseline, respectively 58. Pancreatic cancer surveillance protocols in high-risk families using endoscopic ultrasound and/or endoscopic retrograde cholangiopancreatography demonstrate that routine screening, combined with directed pancreatectomy for suspicious lesions, can effectively identify and treat patients with pancreatic dysplasia prior to the onset of invasive pancreatic cancer 62,63. EUS-based surveillance at Johns Hopkins identified one high-risk individual with an asymptomatic invasive ductal adenocarcinoma. The patient had a disease-free survival of >5 years when Canto et al. published the study 63.
Five hereditary tumor predisposition syndromes associated with pancreatic cancer have been identified thus far (Table III) 64, although these known syndromes account for only a minority (<20%) of familial cases. Hereditary nonpolyposis colorectal cancer (HNPCC) is an autosomal dominant disorder responsible for roughly 3% of colorectal cancers, and has an estimated population prevalence of 1/740 65. At least six distinct DNA mismatch repair (MMR) genes have been linked to HNPCC. Mutations in MMR genes result in the MSI phenotype with enhanced tumorigenesis. Individuals with HNPCC have a 1–5% lifetime risk of developing pancreatic cancer 60,66,67. Other extra-colonic cancers in HNPCC patients include endometrial cancer (60%), stomach cancer (10%), ovarian cancer (12%), genitourinary cancer (4%), biliary cancer (2%), nervous system (4%), and small bowel cancer (1–4%) 68.
Table III. Hereditary tumor predisposition syndromes associated with pancreatic cancer.
| Syndrome | Features | Estimated lifetime risk of pancreatic cancer* | Gene | Locus |
|---|---|---|---|---|
| HNPCC | Colorectal cancers; less commonly cancers of the uterus, stomach, ovaries, genitourinary system, hepatobiliary system, brain, and small intestine | 1–5% | Common | |
| MSH2 | 2p15-16 | |||
| MLH1 | 3p21 | |||
| Rare | ||||
| PMS1 | 2q31 | |||
| PMS2 | 2p15-16 | |||
| MSH6 | 2p15-16 | |||
| Peutz-Jeghers syndrome | Mucocutaneous pigmentations, hamartomatous polyps of the GI tract, and increased risk of GI and non-GI malignancy | 30% | STK11/LKB1 kinase | 19p13.3 |
| FAMMM | Multiple nevi, melanomas, and increased risk of pancreatic cancer | 15% | CDKN2A/p16 | 19p13.3 |
| Hereditary breast and ovarian cancer (BRCA2) | Increased risk of breast, ovarian, and pancreatic cancer | 5–10% | BRCA2 | 13q12.3 |
| Hereditary pancreatitis (HP) | Recurrent pancreatitis and pancreatic cancer | 40% | PRSS1 | 7q35 |
| SPINK1 | 5q32 | |||
| Family X | Absence of extra-pancreatic cancer and pancreatitis; pancreatic insufficiency prior to the onset of cancer | 80% | Unidentified | 4q32-34 |
Peutz-Jeghers syndrome (PJS) is an autosomal dominant disorder with an incidence of 1:25 000. The disease is characterized by mucocutaneous pigmentations and hamartomatous intestinal polyps, due to mutations in the serine/threonine kinase 11 (STK11/LKB1) gene. Patients with PJS have a 132-fold increased risk of developing pancreatic cancer; which translates into a lifetime risk of >30% 60,67,69.
Familial atypical multiple mole-melanoma syndrome (FAMMM) is associated with multiple nevi, melanomas, and extracutaneous tumors. The disease is caused by a mutation in CDKN2A/p16 and accounts for approximately 12% of familial pancreatic cancers 60. Affected individuals have a 20-fold increased risk for developing pancreatic cancer, and a lifetime risk that approaches 15% in some families 70.
Familial breast and ovarian cancer syndrome is due to mutations in the BRCA1 or BRCA2 genes. Individuals with germline BRCA1 mutations have a twofold risk increase of pancreatic cancer; those with BRCA2 mutations have between a 4- and 13-fold risk increase 71,72,73. Inherited BRCA2 mutations are responsible for roughly 17% of familial pancreatic cancer cases, which makes BRCA2 the most common inherited defect contributing to pancreatic cancer identified to date 35. Ashkenazi Jews have the highest incidence of BRCA2-deficient pancreatic cancer. Affected individuals in this subpopulation usually harbor the 6174delT BRCA2 mutation, which occurs in 1% of all Ashkenazi Jews 72. Familial breast cancer syndrome due to a mutated BRCA2 gene carries a lifetime risk for pancreatic cancer which approximates 7% 74.
In addition to the above described familial cancer predisposition syndromes, familial pancreatic cancer can be associated with hereditary pancreatitis (HP). HP is an autosomal dominant disease caused in 70% of the cases by a mutation in cationic trypsinogen (protease serine 1; PRSS1) 75. Mutations in the serine protease inhibitor Kazal type 1 gene (SPINK1) have also been linked to HP 76. Mutated PRSS1 is resistant to inactivation, leading to pancreatic autodigestion. Affected individuals have a 50-fold increased risk of developing pancreatic cancer; their lifetime risk approaches 40% 77.
An unidentified pancreatic cancer gene has been mapped to chromosome arm 4q32-34 through linkage analysis in one family, referred to as ‘family X’ 61. Affected family members have an 80% lifetime risk of pancreatic cancer and do not appear to be at increased risk for extra-pancreatic tumors. The cancers develop earlier (median age 43 years) than sporadic pancreatic cancer (median age 65 years) 78 and are often associated with a prodrome of pancreatic insufficiency 61.
In the overwhelming majority (∼80%) of familial pancreatic cancer cases, the underlying genetic predisposition remains unknown. Some groups have aggressively proposed that unaffected family members undergo annual surveillance 61 to exclude development of pancreatic cancer prospectively (see EUS studies above). There is an urgent need to identify the gene or genes responsible for the vast majority of familial cases, so that a ‘gene test’ akin to other familial cancers like colon or breast cancer can be formulated for unaffected relatives of probands.
Pathology
Pancreatic cancer and development
The embryologic development of normal pancreatic tissue can be separated into three phases. First, dorsal and ventral buds arise from foregut endoderm. Next, cells differentiate into exocrine and endocrine cells. Finally, morphogenesis occurs through growth and branching. These events result in three distinct cell types derived from a common precursor: exocrine acinar cells synthesize and secrete digestive proenzymes into pancreatic ducts; ductal cells line the pancreatic ducts and secrete alkaline fluid; and endocrine cells, primarily involved in glucose homeostasis, are clustered into structures called islets of Langerhans 79.
The cell type of origin in pancreatic cancer has not been definitively characterized. Expression of acinar, ductal, and islet cell markers have each been identified in pancreatic ductal adenocarcinoma. In fact, markers for gastric, duodenal, and colonic epithelium have also been detected 80. Experiments in transgenic mice have led some investigators to speculate that normal pancreata undergoes a metaplastic change into an embryonic-like epithelium early in cancer development. This theory is based on animal studies which demonstrate that metaplastic mouse pancreata develop histological and genetic changes that are similar to changes observed in pancreatic cancer 81,82,83.
The link between pancreatic development and pancreatic cancer, supported mainly by animal studies, has increased interest in certain developmental signaling pathways. The pathways that are best characterized include sonic hedgehog (Shh), Notch, and TGF-β signaling (Figure 3).
In the Hedgehog (Hh) pathway, Shh binds and inhibits the transmembrane receptor, Patched (PTCH) (Figure 3a) 84. This action allows Smoothened (SMO) to initiate downstream signals that stabilize Gli transcription factors, leading to the induction of target Hh genes 84,85. The pathway is repressed during pancreatic development by TGF-β/activin signaling 85. This action is believed to maintain a precursor population and curb the progression of cells towards differentiated endocrine tissue 84,85. Inactivated Shh during embryogenesis can result in developmental abnormalities such as ectopic pancreatic tissue or islet cell hyperplasia 85. Conversely, increased Shh expression leads to the loss of pancreatic lineage markers and the transformation of pancreatic mesenchyme into gut mesoderm 85. Shh pathway components are activated in pancreatic cancer 86,87. Increased expression of Hh factors were observed in pancreatic cancer cell lines and xenografts. Developmental misexpression of Shh in murine pancreata under the control of a Pdx1 promoter resulted in genetic and histological abnormalities that resemble those seen in pancreatic cancer precursor lesions 86. Furthermore, cyclopamine (a SMO antagonist) and a monoclonal antibody against Shh had a growth inhibitory effect on pancreatic cancer cells in vitro, suggesting a requirement for the pathway in growth sustenance.
Notch signaling also regulates epithelial progenitors and inhibits endocrine differentiation in the developing pancreas (Figure 3b) 84,85,88. The Notch pathway is triggered when Delta, Serrate, and Jagged ligands bind Notch receptors. The intracellular component of the activated Notch receptor interacts with the DNA-binding protein RBP-Jκ, to activate the transcription of the hairy/enhancer-of-split genes (Hes). Hes activation directs basic helix-loop-helix factors (bHLH) to down-regulate differentiation-associated genes, like the endocrine activator ngn3 85. Oligonucleotide microarray studies detected overexpression of key Notch pathway components in pancreatic cancer 83. Transfection of the Notch1 and Notch2 receptors into murine pancreatic epithelium induced acinar-to-ductal metaplasia 83. This process can be disrupted in vitro by γ-secretase, an enzymatic inhibitor of Notch signaling 83.
Unlike the Notch and Hh pathways, TGF-β and activin signaling promote endocrine cell development (Figure 3c). The TGF-β and activin ligands bind their respective type II receptors. The type II receptors in turn bind TGF-β and activin type I receptors, and activate intracellular SMAD proteins 30,85. Since the TGF-β and activin pathways counter Hh and Notch pathways during pancreatic development, it is not surprising that TGF-β and activin signaling affect tumor growth in a different manner than the Hh and Notch pathways. As described earlier, multiple factors in the TGF-β and activin pathways harbor inactivating mutations, suggesting that these genes are important tumor suppressors 29.
Pancreatic intraepithelial neoplasia
Dysregulated molecular signaling and the accumulation of genetic changes over time result in incremental changes in pancreatic epithelium that can be appreciated microscopically 89. A unified nomenclature was developed in 1999 to classify intraductal precursor lesions of the pancreas, referred to as pancreatic intraepithelial neoplasia (PanINs) (Figure 4) 90,91. PanIN-1A lesions are flat, tall columnar cells with basally located nuclei; PAN-1B lesions show papillary architecture; PanIN-2 lesions exhibit nuclear abnormalities, such as a loss of polarity or nuclear crowding; PanIN-3 lesions have marked nuclear and cytologic abnormalities and represent the lesions previously referred to as carcinoma in situ of the pancreas. Invasion through the basement membrane marks the transition from PanIN-3 to invasive carcinoma. PanIN lesions demonstrate a progressive accumulation of genetic aberrations that mirror their histologic progression. For example, certain changes like telomere length abnormalities and KRAS2 mutations are observed as early as in PanIN-1 lesions 92. p16 inactivation has often occurred by the PanIN-2 stage 93, while certain changes such as loss of function of SMAD4 94, BRCA2 95, and p53 96 tumor suppressor genes occur in advanced PanIN lesions.
Figure 4. .
Pancreatic intraepithelial neoplasia: PanIN-1A (flat), PanIN-1B (papillary), PanIN-2 (papillary with nuclear changes), and PanIN-3 (severely atypical with mitoses, budding, and luminal necrosis). Images appear on the Johns Hopkins Pancreas Cancer Web 91.
Mucinous cystic neoplasm
Some noninvasive pancreatic lesions can mimic PanINs and, analogous to the latter, can be associated with invasive pancreatic cancers in subsets of cases 90. Mucinous cystic neoplasms (MCN) of the pancreas are a heterogeneous group of cystic tumors that range from small, benign cysts to large cysts with infiltrating adenocarcinoma (Figure 5B) 97. The cysts contain a cloudy fluid that stains positive for mucin, and they are lined by tall, mucin-producing columnar epithelium. MCNs can be large (mean tumor diameter is 10.5 cm) and do not communicate with the main pancreatic duct. They occur more often in women then men, and are usually diagnosed in the fifth decade of life. The presence of dense ovarian stroma underneath the cyst epithelium is a defining characteristic of MCNs 97.
Figure 5. .
(A) Normal pancreatic tissue. (B) Mucinous cystic neoplasm. (C) Intraductal papillary-mucinous neoplasms (IPMNs). (D) Pancreatic ductal adenocarcinoma. Images appear on the Johns Hopkins Pancreas Cancer Web 91.
MCNs may be subcategorized into four groups based on the degree of cellular atypia (e.g. high nuclear-cytoplasmic ratios, pleomorphism, and loss of cell polarity) and the presence or absence of invasion. Thus, mucinous cystadenomas lack significant atypia and are lined by a single layer of epithelium, borderline mucinous cystic neoplasms exhibit papillae and have moderate nuclear atypia, while mucinous carcinoma in situ lesions have severe nuclear atypia. All these categories of mucinous cystic lesions are noninvasive and complete surgical resection is nearly always curative 98. In contrast, approximately a third of MCNs are associated with an invasive carcinoma (mucinous cystadenocarcinoma), which demonstrate invasion of tumor cells into the surrounding stroma, and are associated with a 33% 5-year survival post-resection 98.
IPMNs
Intraductal papillary-mucinous neoplasms (IPMNs) are similar in many aspects to MCNs (Figure 5C). For instance, IPMNs are a heterogeneous group of tumors that include both noninvasive (60%) and invasive (40%) neoplasms 99. Microscopically, they are composed of columnar, mucin-secreting cells and can develop a papillary architecture 97. Indeed, MCNs and IPMNs cannot be distinguished with an electron microscope. However, IPMNs are not associated with ovarian stroma, and they always communicate with the pancreatic duct system. The main pancreatic duct or major side branches are usually dilated and mucus-filled. Therefore, mucin may be seen oozing from the ampulla of Vater during endoscopic evaluation of an IPMN.
The genetic pattern in IPMNs is different from the genetics observed in PanIN lesions and ductal adenocarcinoma. Immunohistochemical studies reveal that 84% of IPMNs express the SMAD4 gene, compared with 45% of ductal adenocarcinomas of the pancreas 100. Furthermore, IPMNs express a different mucin (MUC2) than PanIN lesions and ductal adenocarcinoma (MUC1) 101.
The epithelium of IPMNs is subcategorized by a four-tiered histologic classification scheme, like the one described earlier for MCNs. Intraductal papillary-mucinous adenomas lack significant atypia; borderline IPMNs show an intermediate degree of atypia; and IPMN carcinoma in situ demonstrate severe atypia 102. ‘Colloid’ (mucinous non-cystic) carcinoma is identified in the majority of IPMNs associated with invasive carcinoma; a tubular/ductal type occurs in most of the remaining cases 99.
IPMNs occur equally in men and women, and they generally arise during the seventh decade of life 99. Demographic data suggest that the time interval between the development of an IPMN adenoma and progression to an IPMN with invasive cancer is over 5 years. Five-year survival rates for resected IPMNs without and with invasive cancer are 77% and 43%, respectively 99. Approximately 8% of patients with a resected benign IPMN develop a recurrence or a second metachronous IPMN in the pancreatic remnant 99.
Ductal adenocarcinoma
Ductal adenocarcinoma accounts for >80% of pancreatic cancers. Grossly, they are white-yellow and firm masses. Sixty percent of the cases arise in the pancreatic head, 15% in the body or tail, and 20% involve the gland diffusely 97. They are characterized microscopically by infiltrating small glands that are lined with low-columnar, mucin-containing cells (Figure 5D). Typically, a strong desmoplastic reaction occurs around the cancer 97,102. The nuclei often demonstrate pleomorphism, hyperchromasia, loss of polarity, and prominent nucleoli 97.
Ductal adenocarcinomas are highly aggressive cancers with frequent invasion of vascular, lymphatic, and perineural tissue. Approximately 80% of surgical specimens show disease in regional lymph nodes. There is clinically evident disease at distant organ sites in up to 80% of all patients who are discovered to have pancreatic cancer. The most common sites for distant metastases are the liver (80%), peritoneum (60%), lung and pleura (50–70%), and adrenal glands (25%) 97. The 5-year survival rate for all patients with pancreatic ductal adneocarcinoma is <5% 2. The 5-year survival rate is 15–25% in patients who undergo pancreaticoduoedenectomy, and 30–40% in patients who undergo pancreaticoduoedenectomy with small tumors and node-negative disease 103,104.
Other rare primary non-endocrine tumors of the pancreas include adenosquamous carcinoma, acinar cell carcinoma, giant cell carcinoma, giant cell carcinoma with osteoclast-like giant cells, pancreatoblastoma, serous cystadenoma/cystadenocarcinoma, and solid-pseudopapillary (Hamoudi) neoplasm 97. Table IV gives the AJCC TMN staging system for pancreatic cancer, last revised in 2002 105.
Table IV. TNM classification of exocrine pancreatic cancer.
| TNM definitions |
| Primary tumor (T) |
| TX: Primary tumor cannot be assessed |
| T0: No evidence of primary tumor |
| Tis: Carcinoma in situ |
| T1: Tumor limited to the pancreas, 2 cm or less in greatest dimension |
| T2: Tumor limited to the pancreas, more than 2 cm in greatest dimension |
| T3: Tumor extends beyond the pancreas but without involvement of the celiac axis or the superior mesenteric artery |
| T4: Tumor involves the celiac axis or the superior mesenteric artery (unresectable primary tumor) |
| Regional lymph nodes (N) |
| NX: Regional lymph nodes cannot be assessed |
| N0: No regional lymph node metastasis |
| N1: Regional lymph node metastasis |
| Distant metastasis (M) |
| MX: Distant metastasis cannot be assessed |
| M0: No distant metastasis |
| M1: Distant metastasis |
| AJCC stage groupings |
| Stage 0 |
| Tis, N0, M0 |
| Stage IA |
| T1, N0, M0 |
| Stage IB |
| T2, N0, M0 |
| Stage IIA |
| T3, N0, M0 |
| Stage IIB |
| T1, N1, M0 |
| T2, N1, M0 |
| T3, N1, M0 |
| Stage III |
| T4, any N, M0 |
| Stage IV |
| Any T, any N, M1 |
From AJCC Cancer Staging Manual105.
Conclusions
New insights into the molecular mechanisms that underlie pancreatic carcinogenesis have emerged during the past decade. There is a well supported model that outlines the genetic and histological progression of pancreatic neoplasia. Microarray analysis offers the opportunity to study genomic copy number and gene expression changes across the entire human genome and transcriptome, respectively. Consequently, the number of genes that are known, or suspected, to play a role in the development of pancreatic cancer has increased exponentially in recent years.
However, there is still much to learn. Although there are five well characterized hereditary tumor predisposition syndromes associated with pancreatic cancer, the genetic abnormalities responsible for the majority of cases of familial pancreatic cancer have not been discovered. The exact roles of putative tumor suppressor genes and oncogenes implicated by microarray techniques remain unclear. In addition, the contribution of complex developmental signaling pathways (e.g. Shh, Notch, and TGF-β) to pancreatic carcinogenesis is just beginning to be understood.
Improved patient outcomes in pancreatic cancer will require continued progress in cancer genetics. The molecular analysis of serum and gastrointestinal excreta to detect the presence of pancreatic cancer precursor lesions is one promising area of investigation. Berthelemy et al. demonstrated the potential of this approach 10 years ago when they discovered KRAS2 mutations in pancreatic juice from two patients suspected of having benign pancreatic disease. Both patients presented with symptoms that led to the diagnosis of pancreatic cancer >1 year after the study 106. KRAS2 mutations have also been identified in the stool of patients with pancreatic carcinoma and cholangiocarcinoma 107. However, recent studies show that screening pancreatic juice and stool for KRAS2 mutations (76% and 50% specific, respectively) fails to achieve an appropriate specificity for a relatively low prevalence disease like pancreatic cancer 108, mandating the ongoing search for new markers such as HIP/PAP and PAP-2 109. While ERCP may be an inappropriate screening test due to its invasiveness, EUS is a promising strategy to identify subtle parenchymal changes before they can be visualized by CT or MR. Continued efforts to elucidate the molecular causes of pancreatic cancer will hopefully translate into better diagnostic tests and therapies in the near future.
References
- 1.Jemal A, Murray T, Ward E, Samuels A, Tiwari RC, Ghafoor A, et al. Cancer statistics, 2005. CA Cancer J Clin. 2005;55:10–30. doi: 10.3322/canjclin.55.1.10. [DOI] [PubMed] [Google Scholar]
- 2.Ries LAG, Eisner MP, Kosary CL, Hankey BF, Miller BA, Clegg L, et al. SEER Cancer Statistics Review, 1975–2000. Bethesda: National Cancer Institute, 2003. http://seer.cancer.gov/csr/1975_2000 [Google Scholar]
- 3.Parkin DM, Bray F, Ferlay J, Pisani P. Estimating the world cancer burden: Globocan 2000. Int J Cancer. 2001;94:153–6. doi: 10.1002/ijc.1440. [DOI] [PubMed] [Google Scholar]
- 4.Hansel DE, Kern SE, Hruban RH. Molecular pathogenesis of pancreatic cancer. Annu Rev Genomics Hum Genet. 2003;4:237–56. doi: 10.1146/annurev.genom.4.070802.110341. [DOI] [PubMed] [Google Scholar]
- 5.Trask B. Human cytogenetics: 46 chromosomes, 46 years and counting. Nat Rev Genet. 2002;3:769–78. doi: 10.1038/nrg905. [DOI] [PubMed] [Google Scholar]
- 6.Kricka LJ, Fortina P. Microarray technology and applications: an all-language literature survey including books and patents. Clin Chem. 2001;47:1479–82. [PubMed] [Google Scholar]
- 7.Coe B, Antler B. Spot your genes – an overview of the microarray. BioTeach http://www.bioteach.ubc.ca/MolecularBiology/microarray/ [accessed 9 January 2005]. [Google Scholar]
- 8.Albertson DG, Pinkel D. Genomic microarrays in human genetic disease and cancer. Hum Mol Genet. 2003;12:145–52. doi: 10.1093/hmg/ddg261. [DOI] [PubMed] [Google Scholar]
- 9.Malkhosyan S, Yasuda J, Soto JL, Sekiya T, Yokota J, Perucho M. Molecular karyotype (amplotype) of metastatic colorectal cancer by unbiased arbitrarily primed PCR DNA fingerprinting. Proc Natl Acad Sci U S A. 1998;95:10170–5. doi: 10.1073/pnas.95.17.10170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Iacobuzio-Donahue CA, van der Heijden MS, Baumgartner MR, Troup WJ, Romm JM, Doheny K, et al. Large-scale allelotype of pancreaticobiliary carcinoma provides quantitative estimates of genome-wide allelic loss. Cancer Res. 2004;64:871–5. doi: 10.1158/0008-5472.can-03-2756. [DOI] [PubMed] [Google Scholar]
- 11.Novak U, Oppliger Leibundgut E, Hager J, Muhlematter D, Jotterand M, Besse C, et al. A high-resolution allelotype of B-cell chronic lymphocytic leukemia (B-CLL) Blood. 2002;100:1787–94. [PubMed] [Google Scholar]
- 12.Mahlamaki EH, Hoglund M, Gorunova L, Karhu R, Dawiskiba S, Andren-Sandberg A, et al. Comparative genomic hybridization reveals frequent gains of 20q, 8q, 11q, 12p, and 17q, and losses of 18q, 9p, and 15q in pancreatic cancer. Genes Chromosomes Cancer. 1997;20:383–91. doi: 10.1002/(sici)1098-2264(199712)20:4<383::aid-gcc10>3.0.co;2-o. [DOI] [PubMed] [Google Scholar]
- 13.Curtis LJ, Li Y, Gerbault-Seureau M, Kuick R, Dutrillaux AM, Goubin G, et al. Amplification of DNA sequences from chromosome 19q13.1 in human pancreatic cell lines. Genomics. 1998;53:42–55. doi: 10.1006/geno.1998.5405. [DOI] [PubMed] [Google Scholar]
- 14.Schleger C, Arens N, Zentgraf H, Bleyl U, Verbeke C. Identification of frequent chromosomal aberrations in ductal adenocarcinoma of the pancreas by comparative genomic hybridization (CGH) J Pathol. 2000;191:27–32. doi: 10.1002/(SICI)1096-9896(200005)191:1<27::AID-PATH582>3.0.CO;2-J. [DOI] [PubMed] [Google Scholar]
- 15.Ghadimi BM, Schrock E, Walker RL, Wangsa D, Jauho A, Meltzer PS, et al. Specific chromosomal aberrations and amplification of the AIB1 nuclear receptor coactivator gene in pancreatic carcinomas. Am J Pathol. 1999;154:525–36. doi: 10.1016/S0002-9440(10)65298-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Wilentz RE, Goggins M, Redston M, Marcus VA, Adsay NV, Sohn TA, et al. Genetic, immunohistochemical, and clinical features of medullary carcinoma of the pancreas: a newly described and characterized entity. Am J Pathol. 2000;156:1641–51. doi: 10.1016/S0002-9440(10)65035-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Boland CR. Hereditary nonpolyposis colorectal cancer, The genetic basis of human cancer In:. New York: McGraw-Hill, 1998:307–21. [Google Scholar]
- 18.Feinberg AP. The epigenetics of cancer etiology. Semin Cancer Biol. 2004;14:427–32. doi: 10.1016/j.semcancer.2004.06.005. [DOI] [PubMed] [Google Scholar]
- 19.Ueki T, Toyota M, Sohn T, Yeo CJ, Issa JP, Hruban RH, et al. Hypermethylation of multiple genes in pancreatic adenocarcinoma. Cancer Res. 2000;60:1835–9. [PubMed] [Google Scholar]
- 20.Ueki T, Walter KM, Skinner H, Jaffee E, Hruban RH, Goggins M. Aberrant CpG island methylation in cancer cell lines arises in the primary cancers from which they were derived. Oncogene. 2002;21:2114–17. doi: 10.1038/sj.onc.1205275. [DOI] [PubMed] [Google Scholar]
- 21.Sohn TA. The molecular genetics of pancreatic ductal carcinoma. Minerva Chir. 2002;57:561–74. [PubMed] [Google Scholar]
- 22.Caldas C, Hahn SA, da Costa LT, Redston MS, Schutte M, Seymour AB, 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]
- 23.Schutte M, Hruban RH, Geradts J, Maynard R, Hilgers W, Rabindran SK, et al. Abrogation of the Rb/p16 tumor-suppressive pathway in virtually all pancreatic carcinomas. Cancer Res. 1997;57:3126–30. [PubMed] [Google Scholar]
- 24.Morgan D. Alberts B, Johnson A, Lewis J, Raff M, Roberts K, Walter P. Garland Science; New York: 2002. The cell cycle and programmed cell death, Molecular biology of the cell; pp. 1007–8. [Google Scholar]
- 25.Redston MS, Caldas C, Seymour AB, Hruban RH, da Costa L, Yeo CJ, et al. p53 mutations in pancreatic carcinoma and evidence of common involvement of homocopolymer tracts in DNA microdeletions. Cancer Res. 1994;54:3025–33. [PubMed] [Google Scholar]
- 26.Pellegata NS, Sessa F, Renault B, Bonato M, Leone BE, Solcia E, et al. K-ras and p53 gene mutations in pancreatic cancer: ductal and nonductal tumors progress through different genetic lesions. Cancer Res. 1994;54:1556–60. [PubMed] [Google Scholar]
- 27.Taylor WR, Stark GR. Regulation of the G2/M transition by p53. Oncogene. 2005;20:1803–15. doi: 10.1038/sj.onc.1204252. [DOI] [PubMed] [Google Scholar]
- 28.Hahn SA, Schutte M, Hoque AT, Moskaluk CA, da Costa LT, Rozenblum E, et al. DPC4, a candidate tumor suppressor gene at human chromosome 18q21.1. Science. 1996;271:350–3. doi: 10.1126/science.271.5247.350. [DOI] [PubMed] [Google Scholar]
- 29.Yoo J, Ghiassi M, Jirmanova L, Balliet AG, Hoffman B, Fornace AJ, Jr, et al. Transforming growth factor-beta-induced apoptosis is mediated by Smad-dependent expression of GADD45b through p38 activation. J Biol Chem. 2003;278:43001–7. doi: 10.1074/jbc.M307869200. [DOI] [PubMed] [Google Scholar]
- 30.Goggins M, Shekher M, Turnacioglu K, Yeo CJ, Hruban RH, Kern SE. Genetic alterations of the transforming growth factor beta receptor genes in pancreatic and biliary adenocarcinomas. Cancer Res. 1998;58:5329–32. [PubMed] [Google Scholar]
- 31.Hempen PM, Zhang L, Bansal RK, Iacobuzio-Donahue CA, Murphy KM, Maitra A, et al. Evidence of selection for clones having genetic inactivation of the activin A type II receptor (ACVR2) gene in gastrointestinal cancers. Cancer Res. 2003;63:994–9. [PubMed] [Google Scholar]
- 32.Su GH, Hilgers W, Shekher MC, Tang DJ, Yeo CJ, Hruban RH, et al. Alterations in pancreatic, biliary, and breast carcinomas support MKK4 as a genetically targeted tumor suppressor gene. Cancer Res. 1998;58:2339–42. [PubMed] [Google Scholar]
- 33.Su GH, Hruban RH, Bansal RK, Bova GS, Tang DJ, Shekher MC, et al. Germline and somatic mutations of the STK11/LKB1 Peutz-Jeghers gene in pancreatic and biliary cancers. Am J Pathol. 1999;154:1835–40. doi: 10.1016/S0002-9440(10)65440-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Spicer J, Ashworth A. LKB1 kinase: master and commander of metabolism and polarity. Curr Biol. 2004;14:R383–5. doi: 10.1016/j.cub.2004.05.012. [DOI] [PubMed] [Google Scholar]
- 35.Goggins M, Schutte M, Lu J, Moskaluk CA, Weinstein CL, Petersen GM, et al. Germline BRCA2 gene mutations in patients with apparently sporadic pancreatic carcinomas. Cancer Res. 1996;56:5360–4. [PubMed] [Google Scholar]
- 36.Murphy KM, Brune KA, Griffin C, Sollenberger JE, Petersen GM, Bansal R, et al. Evaluation of candidate genes MAP2K4, MADH4, ACVR1B, and BRCA2 in familial pancreatic cancer: deleterious BRCA2 mutations in 17% Cancer Res. 2002;62:3789–93. [PubMed] [Google Scholar]
- 37.van der Heijden MS, Yeo CJ, Hruban RH, Kern SE. Fanconi anemia gene mutations in young-onset pancreatic cancer. Cancer Res. 2003;63:2585–8. [PubMed] [Google Scholar]
- 38.Almoguera C, Shibata D, Forrester K, Martin J, Arnheim N, Perucho M. Most human carcinomas of the exocrine pancreas contain mutant c-K-ras genes. Cell. 1988;53:549–54. doi: 10.1016/0092-8674(88)90571-5. [DOI] [PubMed] [Google Scholar]
- 39.Hruban RH, van Mansfeld AD, Offerhaus GJ, van Weering DH, Allison DC, Goodman SN, 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;143:545–54. [PMC free article] [PubMed] [Google Scholar]
- 40.Smit VT, Boot AJ, Smits AM, Fleuren GJ, Cornelisse CJ, Bos JL. KRAS codon 12 mutations occur very frequently in pancreatic adenocarcinomas. Nucleic Acids Res. 1988;16:7773–82. doi: 10.1093/nar/16.16.7773. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Kalthoff H, Schmiegel W, Roeder C, Kasche D, Schmidt A, Lauer G, et al. p53 and K-RAS alterations in pancreatic epithelial cell lesions. Oncogene. 1993;8:289–98. [PubMed] [Google Scholar]
- 42.Calhoun ES, Jones JB, Ashfaq R, Adsay V, Baker SJ, Valentine V, et al. BRAF and FBXW7 (CDC4, FBW7, AGO, SEL10) mutations in distinct subsets of pancreatic cancer: potential therapeutic targets. Am J Pathol. 2003;163:1255–60. doi: 10.1016/S0002-9440(10)63485-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Polyak K, Riggins GJ. Gene discovery using the serial analysis of gene expression technique: implications for cancer research. J Clin Oncol. 2001;19:2948–58. doi: 10.1200/JCO.2001.19.11.2948. [DOI] [PubMed] [Google Scholar]
- 44.Guo QM. DNA microarray and cancer. Curr Opin Oncol. 2003;15:36–43. doi: 10.1097/00001622-200301000-00005. [DOI] [PubMed] [Google Scholar]
- 45.Iacobuzio-Donahue CA, Ashfaq R, Maitra A, Adsay NV, Shen-Ong GL, Berg K, et al. Highly expressed genes in pancreatic ductal adenocarcinomas: a comprehensive characterization and comparison of the transcription profiles obtained from three major technologies. Cancer Res. 2003;63:8614–22. [PubMed] [Google Scholar]
- 46.Ryu B, Jones J, Blades NJ, Parmigiani G, Hollingsworth MA, Hruban RH, et al. Relationships and differentially expressed genes among pancreatic cancers examined by large-scale serial analysis of gene expression. Cancer Res. 2002;62:819–26. [PubMed] [Google Scholar]
- 47.Argani P, Rosty C, Reiter RE, Wilentz RE, Murugesan SR, Leach SD, et al. Discovery of new markers of cancer through serial analysis of gene expression: prostate stem cell antigen is overexpressed in pancreatic adenocarcinoma. Cancer Res. 2001;61:4320–4. [PubMed] [Google Scholar]
- 48.Argani P, Iacobuzio-Donahue C, Ryu B, Rosty C, Goggins M, Wilentz RE, et al. Mesothelin is overexpressed in the vast majority of ductal adenocarcinomas of the pancreas: identification of a new pancreatic cancer marker by serial analysis of gene expression (SAGE) Clin Cancer Res. 2001;7:3862–8. [PubMed] [Google Scholar]
- 49.Iacobuzio-Donahue CA, Maitra A, Shen-Ong GL, van Heek T, Ashfaq R, Meyer R, et al. Discovery of novel tumor markers of pancreatic cancer using global gene expression technology. Am J Pathol. 2002;160:1239–49. doi: 10.1016/S0002-9440(10)62551-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Han H, Bearss DJ, Browne LW, Calaluce R, Nagle RB, Von Hoff DD. Identification of differentially expressed genes in pancreatic cancer cells using cDNA microarray. Cancer Res. 2002;62:2890–6. [PubMed] [Google Scholar]
- 51.Crnogorac-Jurcevic T, Missiaglia E, Blaveri E, Gangeswaran R, Jones M, Terris B, et al. Molecular alterations in pancreatic carcinoma: expression profiling shows that dysregulated expression of S100 genes is highly prevalent. J Pathol. 2003;201:63–74. doi: 10.1002/path.1418. [DOI] [PubMed] [Google Scholar]
- 52.Crnogorac-Jurcevic T, Efthimiou E, Nielsen T, Loader J, Terris B, Stamp G, et al. Expression profiling of microdissected pancreatic adenocarcinomas. Oncogene. 2002;21:4587–94. doi: 10.1038/sj.onc.1205570. [DOI] [PubMed] [Google Scholar]
- 53.Iacobuzio-Donahue CA, Maitra A, Olsen M, Lowe AW, van Heek NT, Rosty C, et al. Exploration of global gene expression patterns in pancreatic adenocarcinoma using cDNA microarrays. Am J Pathol. 2003;162:1151–62. doi: 10.1016/S0002-9440(10)63911-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Real FX, Vila MR, Skoudy A, Ramaekers FC, Corominas JM. Intermediate filaments as differentiation markers of exocrine pancreas. II. Expression of cytokeratins of complex and stratified epithelia in normal pancreas and in pancreas cancer. Int J Cancer. 1993;54:720–7. doi: 10.1002/ijc.2910540503. [DOI] [PubMed] [Google Scholar]
- 55.Sinha P, Hutter G, Kottgen E, Dietel M, Schadendorf D, Lage H. Increased expression of epidermal fatty acid binding protein, cofilin, and 14-3-3-sigma (stratifin) detected by two-dimensional gel electrophoresis, mass spectrometry and microsequencing of drug-resistant human adenocarcinoma of the pancreas. Electrophoresis. 1999;20:2952–60. doi: 10.1002/(SICI)1522-2683(19991001)20:14<2952::AID-ELPS2952>3.0.CO;2-H. [DOI] [PubMed] [Google Scholar]
- 56.Elsasser HP, MacDonald R, Dienst M, Kern HF. Characterization of a transglutaminase expressed in human pancreatic adenocarcinoma cells. Eur J Cell Biol. 1993;61:321–8. [PubMed] [Google Scholar]
- 57.Hruban RH, Petersen GM, Goggins M, Tersmette AC, Offerhaus GJ, Falatko F, et al. Familial pancreatic cancerAnn Oncol1999;10:569–573. http://pathology2.jhu.edu/pancreas/nfptr.cfm [PubMed] [Google Scholar]
- 58.Tersmette AC, Petersen GM, Offerhaus GJ, Falatko FC, Brune KA, Goggins M, et al. Increased risk of incident pancreatic cancer among first-degree relatives of patients with familial pancreatic cancer. Clin Cancer Res. 2001;7:738–44. [PubMed] [Google Scholar]
- 59.Bartsch DK, Kress R, Sina-Frey M, Grutzmann R, Gerdes B, Pilarsky C, et al. Prevalence of familial pancreatic cancer in Germany. Int J Cancer. 2004;110:902–6. doi: 10.1002/ijc.20210. [DOI] [PubMed] [Google Scholar]
- 60.Rieder H, Bartsch D. Familial pancreatic cancer. Fam Cancer. 2004;3:69–74. doi: 10.1023/B:FAME.0000026822.67291.a1. [DOI] [PubMed] [Google Scholar]
- 61.Eberle MA, Pfutzer R, Pogue-Geile KL, Bronner MP, Crispin D, Kimmey MB, et al. A new susceptibility locus for autosomal dominant pancreatic cancer maps to chromosome 4q32-34. Am J Hum Genet. 2002;70:1044–8. doi: 10.1086/339692. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Rulyak SJ, Brentnall TA. Inherited pancreatic cancer: surveillance and treatment strategies for affected families. Pancreatology. 2001;1:477–85. doi: 10.1159/000055851. [DOI] [PubMed] [Google Scholar]
- 63.Canto MI, Goggins M, Yeo CJ, Griffin C, Axilbund JE, Brune K, et al. Screening for pancreatic neoplasia in high-risk individuals: an EUS-based approach. Clin Gastroenterol Hepatol. 2004;2:606–21. doi: 10.1016/s1542-3565(04)00244-7. [DOI] [PubMed] [Google Scholar]
- 64.Lynch HT, Deters CA, Lynch JF, Brand RE. Familial pancreatic carcinoma in Jews. Fam Cancer. 2004;3:233–40. doi: 10.1007/s10689-004-9549-8. [DOI] [PubMed] [Google Scholar]
- 65.Salovaara R, Loukola A, Kristo P, Kaariainen H, Ahtola H, Eskelinen M, et al. Population-based molecular detection of hereditary nonpolyposis colorectal cancer. J Clin Oncol. 2000;18:2193–200. doi: 10.1200/JCO.2000.18.11.2193. [DOI] [PubMed] [Google Scholar]
- 66.Peltomaki P, Gao X, Mecklin JP. Genotype and phenotype in hereditary nonpolyposis colon cancer: a study of families with different vs. shared predisposing mutations. Fam Cancer. 2001;1:9–15. doi: 10.1023/a:1011564720772. [DOI] [PubMed] [Google Scholar]
- 67.Ulrich CD.Growth factors, receptors, and molecular alterations in pancreatic cancer. Putting it all together Med Clin North Am 2000;84:697–705, xi–xii. [DOI] [PubMed] [Google Scholar]
- 68.Watson P, Lynch HT. Cancer risk in mismatch repair gene mutation carriers. Fam Cancer. 2001;1:57–60. doi: 10.1023/a:1011590617833. [DOI] [PubMed] [Google Scholar]
- 69.Giardiello FM, Welsh SB, Hamilton SR, Offerhaus GJ, Gittelsohn AM, Booker SV, et al. Increased risk of cancer in the Peutz-Jeghers syndrome. N Engl J Med. 1987;316:1511–14. doi: 10.1056/NEJM198706113162404. [DOI] [PubMed] [Google Scholar]
- 70.Vasen HF, Gruis NA, Frants RR, van Der Velden PA, Hille ET, Bergman W. Risk of developing pancreatic cancer in families with familial atypical multiple mole melanoma associated with a specific 19 deletion of p16 (p16-Leiden) Int J Cancer. 2000;87:809–11. [PubMed] [Google Scholar]
- 71.Thompson D, Easton DF. Breast Cancer Linkage Consortium. Cancer incidence in BRCA1 mutation carriers. J Natl Cancer Inst. 2002;94:1358–65. doi: 10.1093/jnci/94.18.1358. [DOI] [PubMed] [Google Scholar]
- 72.Figer A, Irmin L, Geva R, Flex D, Sulkes J, Sulkes A, et al. The rate of the 6174delT founder Jewish mutation in BRCA2 in patients with non-colonic gastrointestinal tract tumours in Israel. Br J Cancer. 2001;84:478–81. doi: 10.1054/bjoc.2000.1605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Breast Cancer Linkage Consortium. Cancer risks in BRCA2 mutation carriers. J Natl Cancer Inst 1999;91:1310–16. [DOI] [PubMed] [Google Scholar]
- 74.Ozcelik H, Schmocker B, Di Nicola N, Shi XH, Langer B, Moore M, et al. Germline BRCA2 6174delT mutations in Ashkenazi Jewish pancreatic cancer patients. Nat Genet. 1997;16:17–18. doi: 10.1038/ng0597-17. [DOI] [PubMed] [Google Scholar]
- 75.Whitcomb DC, Gorry MC, Preston RA, Furey W, Sossenheimer MJ, Ulrich CD, et al. Hereditary pancreatitis is caused by a mutation in the cationic trypsinogen gene. Nat Genet. 1996;14:141–5. doi: 10.1038/ng1096-141. [DOI] [PubMed] [Google Scholar]
- 76.Witt H, Luck W, Hennies HC, Classen M, Kage A, Lass U, et al. Mutations in the gene encoding the serine protease inhibitor, Kazal type 1 are associated with chronic pancreatitis. Nat Genet. 2000;25:213–16. doi: 10.1038/76088. [DOI] [PubMed] [Google Scholar]
- 77.Lowenfels AB, Maisonneuve P, DiMagno EP, Elitsur Y, Gates LK, Jr, Perrault J, et al. Hereditary pancreatitis and the risk of pancreatic cancer. International Hereditary Pancreatitis Study Group. J Natl Cancer Inst. 1997;89:442–6. doi: 10.1093/jnci/89.6.442. [DOI] [PubMed] [Google Scholar]
- 78.Lowenfels AB, Maisonneuve P. Epidemiologic and etiologic factors of pancreatic cancer. Hematol Oncol Clin North Am. 2002;16:1–16. doi: 10.1016/s0889-8588(01)00003-x. [DOI] [PubMed] [Google Scholar]
- 79.Meszoely IM, Means AL, Scoggins CR, Leach SD. Developmental aspects of early pancreatic cancer. Cancer J. 2001;7:242–50. [PubMed] [Google Scholar]
- 80.Sessa F, Bonato M, Frigerio B, Capella C, Solcia E, Prat M, et al. Ductal cancers of the pancreas frequently express markers of gastrointestinal epithelial cells. Gastroenterology. 1990;98:1655–65. doi: 10.1016/0016-5085(90)91104-e. [DOI] [PubMed] [Google Scholar]
- 81.Wagner M, Greten FR, Weber CK, Koschnick S, Mattfeldt T, Deppert W, et al. A murine tumor progression model for pancreatic cancer recapitulating the genetic alterations of the human disease. Genes Dev. 2001;15:286–93. doi: 10.1101/gad.184701. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Song SY, Gannon M, Washington MK, Scoggins CR, Meszoely IM, Goldenring JR, et al. Expansion of Pdx-1-expressing pancreatic epithelium and islet neogenesis in transgenic mice overexpressing transforming growth factor α. Gastroenterology. 1999;117:1416–26. doi: 10.1016/s0016-5085(99)70292-1. [DOI] [PubMed] [Google Scholar]
- 83.Miyamoto Y, Maitra A, Ghosh B, Zechner U, Argani P, Iacobuzio-Donahue CA, et al. Notch mediates TGFα-induced changes in epithelial differentiation during pancreatic tumorigenesis. Cancer Cell. 2003;3:565–76. doi: 10.1016/s1535-6108(03)00140-5. [DOI] [PubMed] [Google Scholar]
- 84.Heiser PW, Hebrok M. Development and cancer. Lessons learned in the pancreas. Cell Cycle. 2004;3:270–2. [PubMed] [Google Scholar]
- 85.Kim SK, Hebrok M. Intercellular signals regulating pancreas development and function. Genes Dev. 2001;15:111–27. doi: 10.1101/gad.859401. [DOI] [PubMed] [Google Scholar]
- 86.Thayer SP, Pasca di Maliano M, Heiser PW, Nielsen CM, Roberts DJ, Lauwers GY, et al. Hedgehog is an early and late mediator of pancreatic cancer tumorigenesis. Nature. 2003;425:851–6. doi: 10.1038/nature02009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Berman DM, Karhadkar SS, Maitra A, Montes De Oca R, Gerstenblith MR, Briggs K, et al. Widespread requirement for Hedgehog ligand stimulation in growth of digestive tract tumours. Nature. 2003;425:846–51. doi: 10.1038/nature01972. [DOI] [PubMed] [Google Scholar]
- 88.Means AL, Leach SD. Lineage commitment and cellular differentiation in exocrine pancreas. Pancreatology. 2001;1:587–96. doi: 10.1159/000055868. [DOI] [PubMed] [Google Scholar]
- 89.Hruban RH, Wilentz RE, Kern SE. Genetic progression in the pancreatic ducts. Am J Pathol. 2000;156:1821–5. doi: 10.1016/S0002-9440(10)65054-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Hruban RH, Adsay NV, Albores-Saavedra J, Compton C, Garrett ES, Goodman SN, et al. Pancreatic intraepithelial neoplasia. A new nomenclature and classification system for pancreatic duct lesions. Am J Surg Pathol. 2001;25:579–86. doi: 10.1097/00000478-200105000-00003. [DOI] [PubMed] [Google Scholar]
- 91.New Nomenclature for Classification of Duct Lesions in the Pancreas. Johns Hopkins Pancreas Cancer Web, 1999. Johns Hopkins University. http://pathology.jhu.edu/n.web?EP=N&FL=PANCREAS_PANIN [accessed 3 April 2005]. [Google Scholar]
- 92.Luttges J, Schlehe B, Menke MA, Vogel I, Henne-Bruns D, Kloppel G. The K-ras mutation pattern in pancreatic ductal adenocarcinoma usually is identical to that in associated normal, hyperplastic, and metaplastic ductal epithelium. Cancer. 1999;85:1703–10. [PubMed] [Google Scholar]
- 93.Moskaluk CA, Hruban RH, Kern SE. p16 and K-ras gene mutations in the intraductal precursors of human pancreatic adenocarcinoma. Cancer Res. 1997;57:2140–3. [PubMed] [Google Scholar]
- 94.Wilentz RE, Iacobuzio-Donahue CA, Argani P, McCarthy DM, Parsons JL, Yeo CJ, et al. Loss of expression of Dpc4 in pancreatic intraepithelial neoplasia: evidence that DPC4 inactivation occurs late in neoplastic progression. Cancer Res. 2000;60:2002–6. [PubMed] [Google Scholar]
- 95.Goggins M, Hruban RH, Kern SE. BRCA2 is inactivated late in the development ofpancreatic intraepithelial neoplasia. Am J Pathol. 2000;156:1767–71. doi: 10.1016/S0002-9440(10)65047-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.DiGiuseppe JA, Hruban RH, Goodman SN, Polak M, van den Berg FM, Allison DC, et al. Overexpression of p53 protein in adenocarcinoma of the pancreas. Am J Clin Pathol. 1994;101:684–8. doi: 10.1093/ajcp/101.6.684. [DOI] [PubMed] [Google Scholar]
- 97.Wilentz RE, Hruban RH. Pathology of cancer of the pancreas. Surg Oncol Clin North Am. 1998;7:43–65. [PubMed] [Google Scholar]
- 98.Wilentz RE, Albores-Saavedra J, Zahurak M, Talamini MA, Yeo CJ, Cameron JL, et al. Pathologic examination accurately predicts prognosis in mucinous cystic neoplasms of the pancreas. Am J Surg Pathol. 1999;23:1320–7. doi: 10.1097/00000478-199911000-00002. [DOI] [PubMed] [Google Scholar]
- 99.Sohn TA, Yeo CJ, Cameron JL, Hruban RH, Fukushima N, Campbell KA, et al. Intraductal papillary mucinous neoplasms of the pancreas: an updated experience Ann Surg 2004;239:788–97; discussion 797–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Sohn TA, Yeo CJ, Cameron JL, Iacobuzio-Donahue CA, Hruban RH, Lillemoe KD. Intraductal papillary mucinous neoplasms of the pancreas: an increasingly recognized clinicopathologic entity Ann Surg 2001;234:313–21; discussion 321–2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Adsay NV, Merati K, Basturk O, Iacobuzio-Donahue C, Levi E, Cheng JD, et al. Pathologically and biologically distinct types of epithelium in intraductal papillary mucinous neoplasms: delineation of an “intestinal” pathway of carcinogenesis in the pancreas. Am J Surg Pathol. 2004;28:839–48. doi: 10.1097/00000478-200407000-00001. [DOI] [PubMed] [Google Scholar]
- 102.Kloppel G, Solcia E, Longnecker DS, Capella S, Sobin LH. Springer; Berlin: 1996. Histological typing of tumours of the exocrine pancreas, World Health Organization international classification of tumors, 2nd edn; pp. 11–20. [Google Scholar]
- 103.Iacobuzio-Donahue CA, Ryu B, Hruban RH, Kern SE. Exploring the host desmoplastic response to pancreatic carcinoma: gene expression of stromal and neoplastic cells at the site of primary invasion. Am J Pathol. 2002;160:91–9. doi: 10.1016/S0002-9440(10)64353-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Yeo CJ, Sohn TA, Cameron JL, Hruban RH, Lillemoe KD, Pitt HA. Periampullary adenocarcinoma: analysis of 5-year survivors. Ann Surg. 1998;227:821–31. doi: 10.1097/00000658-199806000-00005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Exocrine pancreas. In: American Joint Committee on Cancer: AJCC Cancer Staging Manual, 6th edn New York: Springer, 2002:157–64. [Google Scholar]
- 106.Berthelemy P, Bouisson M, Escourrou J, Vaysse N, Rumeau JL, Pradayrol L. Identification of K-ras mutations in pancreatic juice in the early diagnosis of pancreatic cancer. Ann Intern Med. 1995;123:188–91. doi: 10.7326/0003-4819-123-3-199508010-00005. [DOI] [PubMed] [Google Scholar]
- 107.Caldas C, Hahn SA, Hruban RH, Redston MS, Yeo CJ, Kern SE. Detection of K-ras mutations in the stool of patients with pancreatic adenocarcinoma and pancreatic ductal hyperplasia. Cancer Res. 1994;54:3568–73. [PubMed] [Google Scholar]
- 108.Lu X, Xu T, Qian J, Wen X, Wu D. Detecting K-ras and p53 gene mutation from stool and pancreatic juice for diagnosis of early pancreatic cancer. Chin Med J (Engl) 2002;115:1632–6. [PubMed] [Google Scholar]
- 109.Gronborg M, Bunkenborg J, Kristiansen TZ, Jensen ON, Yeo CJ, Hruban RH, et al. Comprehensive proteomic analysis of human pancreatic juice. J Proteome Res. 2004;3:1042–55. doi: 10.1021/pr0499085. [DOI] [PubMed] [Google Scholar]





