Abstract
Context
The introduction of genome- and epigenome-wide screening techniques has dramatically improved our understanding of the molecular mechanisms underlying the development of pancreatic cancer. There are now 3 recognized histologic precursors of pancreatic cancer: pancreatic intraepithelial neoplasia, intraductal papillary mucinous neoplasm, and mucinous cystic neoplasm. Each of these precursor lesions is associated with specific molecular alterations.
Objective
To understand the molecular characteristics of pancreatic ductal adenocarcinoma and its precursor lesions.
Data Sources
PubMed (US National Library of Medicine).
Conclusions
In this review, we briefly summarize recent research findings on the genetics and epigenetics of pancreatic cancer. In addition, we characterize these molecular alterations in the context of the histologic subtypes of pancreatic cancer.
Pancreatic cancer is the fourth leading cause of cancer death in both men and women in the United States. In 2010, it is estimated that 43 140 Americans will be diagnosed and 36 800 patients will die of pancreatic cancer.1 Most pancreatic cancers are pancreatic ductal adenocarcinomas and the 5-year survival rate for patients with localized disease after surgical resection is 20% and for those with metastatic disease, the survival is only 2%.1 The poor survival rate is attributed to the late detection of pancreatic cancers; 85% of patients present with advanced disease that is unresectable. Although significant resources have been committed to improving the survival of patients with pancreatic cancer in the past decades, there has been no significant improvement in survival.1 Research into the molecular mechanisms of pancreatic cancer has revealed that the disease is due to both genetic and epigenetic changes. The introduction of genome- and epigenome-wide screening techniques has expanded the numbers of genes linked to pancreatic cancer.2–6 In this review, we briefly summarize recent research findings on genetics and epigenetics of pancreatic cancer in the context of histologic variants, precursor lesions, and familial pancreatic cancer.
GENETICS OF PANCREATIC CANCER
A recent comprehensive study of the pancreatic cancer genome profiled the genetic abnormalities of pancreatic ductal adenocarcinomas. In this study, Jones and colleagues7 sequenced 20 661 protein-coding genes in 24 ductal adenocarcinomas and demonstrated an average of 48 nonsilent mutations, 6 amplifications, and 8 homozygous deletions per pancreatic cancer. These mutations were associated with 12 core signaling pathways.7 Based on the frequency of genetically affected genes in pancreatic cancers, a genetic “topographic map” of the pancreatic cancers can be generated in which the most frequent mutations are represented as 4 “mountains” (high-frequency driver genes) involving KRAS2, CDKN2A/p16, SMAD4/DPC4, and TP53, with numerous “hills” (low-frequency driver genes) involving SMARC4A, CDH1, EPHA3, FBXW7, EGFR, IDH1, and NF1.7
1. Oncogenes and Pancreatic Cancer
The most frequently mutated oncogene in pancreatic cancers is KRAS2 (mutated in >95% of pancreatic cancers), which is activated by point mutations, most often in codon 12.7,8 The KRAS2 gene is located on chromosome arm 12p and encodes a membrane-bound guanosine triphosphate (GTP)–binding protein. This GTP-binding protein mediates various cellular functions, such as proliferation, cellular survival, motility, and cytoskeletal remodeling. Activating KRAS gene mutations abolish the regulated GTPase activity of the Kras protein, which results in constitutive signaling.9 Mutations in the KRAS2 gene are observed in the earliest _ENREF_10pancreatic intraepithelial neoplasia (PanIN) lesions and are considered to be one of the earliest genetic events in pancreatic tumorigenesis.7,10,11 Several additional signaling pathways downstream from KRAS2, including BRAF-MAPK and PI3K-AKT, may also be activated by mutations. The BRAF pathway is activated by a point mutation at V600E. BRAF gene mutations are observed in 5% of pancreatic cancers that do not possess a KRAS2 mutation.12 These cancers are often microsatellite unstable. Similarly, amplifications in the AKT2 gene are seen in 10% to 20% of pancreatic cancers.13,14 Amplifications of other oncogenes such as C-MYC,7,15 KRAS, and GATA6,16, 15 are less frequent.
2. Tumor Suppressor Genes in Pancreatic Cancer
Three tumor suppressor genes, CDKN2A/p16, TP53, and SMAD4/DPC4, are commonly inactivated in pancreatic cancers.7,17–20 CDKN2A/p16 on chromosome arm 9p is inactivated in more than 95% of pancreatic cancers by several different mechanisms, such as homozygous deletion of both alleles of the gene; intragenic mutation in 1 allele, coupled with loss of the other allele; or promoter hypermethylation.17,21,22 The p16 protein inhibits progression of the cell cycle at the G1-S checkpoint binding of cyclin-dependant kinases (CDKs), including CDK4 and CDK6.23 The TP53 gene on chromosome arm 17p is inactivated in 50% to 75% of pancreatic cancers.7,19,24,25
p53 proteins play several key roles including maintaining G2-M arrest, regulating G1-S checkpoint, inducing apoptosis, regulating senescence, repairing DNA, and changing cellular metabolism.26 Inactivation of the TP53 gene typically occurs through intragenic mutations of 1 allele, accompanied with loss of the other allele.19 Functional loss of the p53 protein enables cellular survival and division in the presence of DNA damage; this facilitates the accumulation of further genetic abnormalities.26
SMAD4/DPC4 on chromosome arm 18q is inactivated in 55% of pancreatic cancers.27,28 SMAD4/DPC4 is inactivated by homozygous deletion and by intragenic mutations accompanied by loss of the other allele.28,29 Smad4 (dpc4) protein has a critical function in the signal transduction cascade that involves transforming growth factor β (TGF-β) and multiple targets in the TGF-β pathway. Binding of the TGF-β ligand to its receptor triggers a series of reactions including binding of the transcription factor smad2/3 to smad4. Through multiple target genes, the TGF-β pathway normally regulates cellular growth. Loss of smad4 (dpc4) function abolishes the smad4-dependant TGF-β pathway and gives rise to unregulated cellular proliferation.30 Loss of smad4 nuclear labeling by immunohistochemistry is generally observed late in pancreatic carcinogenesis, such as in PanIN-3 precursor lesions and infiltrating adenocarcinomas.31 Both SMAD4/DPC4 mutation and loss of smad4 expression are markers of poor prognosis in pancreatic cancers.32,33 In contrast to SMAD4/DPC4 mutation, mutations in TP53 and CDKN2A/p16 have not been shown to predict survival.33 Loss of smad4 protein expression can also be used in the differential diagnosis of carcinomas of unknown primary tumor; SMAD4/DPC4 mutations with loss of nuclear smad4 labeling frequently occur in pancreatic adenocarcinomas, but not in extrapancreatic malignancies.34 SMAD4 mutations have recently been associated with poor prognosis and with the development of widespread metastases in pancreatic cancer.33,35
An additional tumor suppressor pathway that can be altered in pancreatic cancers involves STK11/LKB1 on chromosome arm 19p. Germline mutations of STK11/LKB1 are responsible for Peutz-Jeghers syndrome and are associated with intraductal papillary mucinous neoplasms (IPMNs) and invasive pancreatic cancer. In addition to germline mutations, somatic mutations of STK11/LKB1 are observed in 5% of patients with sporadic IPMNs and pancreatic cancers.36,37 Other tumor suppressor genes, including TGFBR2,38 MAP2K4/MKK4,39,40 FBXW7,12 and ACVR1B4, are inactivated in a small subset of pancreatic cancers. The genetically altered genes involved in pancreatic cancer are summarized in Table 1.
Table 1.
Gene Symbol | Gene Name | Genetic Alteration | Mechanism of Genetic Alteration | Chromosome Site | Known or Predicted Function | Alteration in Primary Pancreatic Cancer, % | Source, y |
---|---|---|---|---|---|---|---|
CDKN2A/p16 | Cyclin-dependent kinase inhibitor 2A | Inactivation | Homozygous deletion (41%), intragenic mutation (38%) | 9p21 | Cyclin-dependent kinase inhibitor | 95 | Caldas et al,17 1994 |
KRAS2 | v-Ki-ras2 Kirsten rat sarcoma viral oncogene homolog | Activation | Point mutation | 12p12.1 | Signal transduction, proliferation, cell survival, and motility | >90 | Hruban et al,8 1993 |
TP53 | Tumor protein p53 | Inactivation | Intragenic mutation in 1 allele and loss in the other allele | 17p13.1 | Cell cycle arrest, apoptosis, senescence, DNA repair, metabolism change | 50–70 | Redston et al,19 1994 Moore et al,24 2001 Scarpa et al,25 1993 |
SMAD4/DPC4 | Mothers against decapentaplegic, drosophila, homolog of, 4 | Inactivation | Homozygous deletion (50%), intragenic mutation in 1 allele and loss in the other allele (50%) | 18q21.1 | Signal transmission | 55 | Iacobuzio-Donahue et al,27 2004 Hahn et al,28 1996 |
AKT2 | v-akt murine thymoma viral oncogene homolog 2 | Activation | Amplification | 19q13.1-q13.2 | AKT pathway, hormone metabolism | 10–20 | Ruggeri et al,13 1998 Cheng et al,14 1996 |
MLH1 | mutL homolog 1, colon cancer, nonpolyposis type 2 (E. coli) | Inactivation | Heterozygous mutations | 3p21.3 | DNA mismatch repair | 3–15 | Goggins et al,59 1998 Wilentz et al,61 2000 |
BRCA2 | Breast cancer 2, early onset | Inactivation | Homozygous deletion | 13q12.3 | DNA repair, proliferation, differentiation | 7 | Goggins et al,155 1996 |
STK11/LKB1 | Serine/threonine kinase 11 | Inactivation | Homozygous deletion, intragenic mutation in 1 allele and loss in the other allele | 19p13.3 | Apoptosis regulation | 5 | Su et al,36 1999 |
BRAF | v-raf murine sarcoma viral oncogene homolog B1 | Activation | Point mutation | 7q34 | Signal transduction, cell growth | 5 | Calhoun et al,12 2003 |
TGFBR2 | Transforming growth factor, β receptor II (70/80 kDa) | Inactivation | Homozygous deletion, homozygous frameshift mutation | 3p22 | Signal transduction | 4 | Goggins et al,38 1998 |
MAP2K4 | Mitogen-activated protein kinase kinase 4 | Inactivation | Homozygous deletions, missense mutation | 17p11.2 | MAPK pathway | 2 | Su et al,39 1998 Teng et al,40 1997 |
Abbreviation: MAPK, mitogen-activated protein kinase.
3. Genetics of Precursor Lesions
There are 3 histologically recognized precursor lesions of pancreatic cancer: PanINs, IPMNs, and mucinous cystic neoplasms (MCNs).42–45 Pancreatic intraepithelial neoplasia lesions are microscopic papillary or flat noninvasive epithelial neoplasms (<0.5 mm) arising in pancreatic ducts characterized by mucin-containing cuboidal to columnar cells.
Pancreatic intraepithelial neoplasia lesions can be further classified according to the degree of cytologic and architectural atypia as PanIN-1, PanIN-2, and PanIN-3.43,44 Two distinct genetic events occur in early low-grade PanIN lesions (PanIN-1): telomere shortening and KRAS2 gene mutations.8,10,11,46,47 Activating point mutations of KRAS2 occur in approximately 45% of PanIN-1 lesions.8,10,11,47 Telomere shortening is found in approximately 90% of PanIN-1 lesions and may contribute to global chromosomal abnormalities in PanINs.46 Inactivating mutations of CDKN2A/p16 begin to occur in PanIN-2 lesions, while inactivation of TP53, SMAD4/DPC4, and BRCA2 are generally associated with higher-grade PanIN lesions(PanIN-3).31,48
Intraductal papillary mucinous neoplasms are mucin-producing epithelial neoplasms, usually with papillary architecture; they arise from the main pancreatic duct or branch ducts.44 These neoplasms are larger lesions than PanINs (≥1 cm) and therefore can be detected by imaging.44 Activating point mutations of KRAS2 occur in approximately 50% of IPMNs with low-grade dysplasia, and the prevalence of KRAS mutations increases with the degree of dysplasia.49–51 Inactivating mutations of CDKN2A/p16 and TP53 are found in IPMNs with high-grade dysplasia.52 Loss of smad4 expression is observed in only a small subset of IPMNs (3%), whereas smad4 loss in PanIN3 occurs in approximately 30% of cases.53 As described above, somatic mutations of STK11/LKB1, with loss of the wild-type allele and corresponding inactivation of stk11 protein, occur in a small proportion of IPMNs.36,37,53
Mucinous cystic neoplasms occur predominantly in women.42 In contrast to IPMNs, MCNs do not have a connection with the pancreatic duct. In addition, MCNs are unique among pancreatic precursor lesions because of an associated ovarian-type stroma.42 As compared to PanINs and IPMNs, the genetic alterations of MCNs have not been well defined. Studies of MCNs54–56 have reported a range in the prevalence of KRAS2 mutations and p53 overexpression, with the prevalence of abnormalities increasing with increasing degrees of dysplasia. One observation is that Smad4 mutation and loss of nuclear expression do not occur in most noninvasive MCNs. As with cancers arising from PanIN-3 lesions, smad4 expression is lost when infiltrating cancers arise from MCNs.29 This suggests that inactivation of SMAD4/DPC4 occurs in the late stages of neoplastic progression from MCNs.29
4. Genetics of Histologic Variants of Pancreatic Cancer
Several histologic variants of pancreatic cancer have been described, which include adenosquamous carcinoma, colloid carcinoma, medullary carcinoma, signet ring cell carcinoma, undifferentiated carcinoma, and undifferentiated carcinoma with osteoclast-like giant cells.42 Recognition of these variants is clinically important. Indeed, colloid and medullary carcinomas typically have better prognoses than the typical infiltrating ductal adenocarcinomas, and adenosquamous and undifferentiated carcinomas have worse prognoses than the typical ductal adenocarcinomas.57–59 Furthermore, medullary carcinomas have distinct mechanisms of pathogenesis. We will briefly describe the genetic characteristics of these histologic variants, but we recommend a more comprehensive review for more in-depth discussion.60
Adenosquamous carcinomas contain both glandular and squamous components.42 The squamous component, by definition, comprises at least 30% of the neoplasm. Adenosquamous carcinomas share similar genetic features with ductal adenocarcinomas, including KRAS2 mutations and inactivation of CDKN2A/p16, SMAD4/DPC4, and/or TP53.58 The squamous component expresses p63, which is a helpful finding for identifying squamous components. Recognition of adenosquamous carcinoma is clinically important because it is associated with worse survival than adenocarcinomas.58
Medullary carcinomas are characterized by well-defined pushing border, syncytial growth pattern, and poorly differentiated cancer cells.59,61,62 Similar to medullary carcinomas of the colorectum, medullary carcinomas of the pancreas are often microsatellite unstable; this is caused either by germline or somatic mutation of the mismatch repair genes MHL1 and MSH2 or by epigenetic silencing of MLH1 by promoter methylation.22,59,61,62 Medullary carcinomas are associated with a better prognosis than ductal adenocarcinomas. Medullary colorectal cancers (with microsatellite instability) respond poorly to 5-fluorouracil–based chemotherapy, but it is not known if this 5-fluorouracil resistance applies to medullary carcinoma of the pancreas.63
Colloid carcinomas are characterized by well-differentiated neoplastic cells floating in pools of extracellular mucin; by definition, the mucin pools should comprise at least 80% of the tumor.57 The neoplastic cells have intestinal differentiation and label with antibodies to MUC2 and/or CDX2.64,65 Colloid carcinomas are associated with a better prognosis than ductal adenocarcinomas.57
Undifferentiated carcinomas lack histologic features of differentiation.42,59–61 The median survival time for patients with undifferentiated pancreatic adenocarcinoma is only 5 months after surgical resection.66 Undifferentiated carcinomas are noncohesive cancers characterized by the loss of E-cadherin protein expression.67 The expression of L1CAM, COX2, and EGFR proteins in undifferentiated carcinomas have been noted as possible future targets of inhibitor-based treatments.68
Undifferentiated carcinomas with osteoclast-like giant cells are composed of cytologically benign, multinucleated, osteoclast-like giant cells admixed with atypical pleomorphic mononuclear cells.42 Frequently, undifferentiated carcinomas with osteoclast-like giant cells occur in association with noninvasive precursor lesions and share mutations with the associated noninvasive precursor lesions.69–73
5. Genetics of Familial Pancreatic Cancer
Up to 10% of pancreatic cancers have a familial basis.74 Several cohort and case-control studies75,76 report that individuals with first-degree relatives who have pancreatic cancer are at significantly greater risk for pancreatic cancer, a risk that increases with the number of affected relatives. Thus, the risk for pancreatic cancer in individuals with 1 first-degree relative with pancreatic cancer is 2-fold higher than that for an individual without an affected first-degree relative; persons with 2 affected first-degree relatives have a 6-fold increased risk; and persons with 3 or more affected first-degree relatives have a 14- to 32-fold increased risk for pancreatic cancer.75,76
Several genetic syndromes are linked to the development of familial pancreatic cancer. Hereditary breast and ovarian cancer syndrome is an autosomal, dominantly inherited disease characterized by early development of breast and ovarian cancer and germline mutation of BRCA2 and BRCA1.74 Germline mutation of BRCA2 increases risk for pancreatic cancer 3.5- to 10-fold.77–79 BRCA2 is a member of the Fanconi anemia gene family, and the function of the BRCA2 gene product is to repair DNA interstrand cross-links and double-strand breaks.80 Pancreatic cancer cells with BRCA2 mutation are hypersensitive to DNA interstrand cross-linking agents, including mitomycin C, cisplatin, and poly(ADP-ribose) polymerase inhibitors.81–83 Peutz-Jeghers syndrome is an autosomal, dominantly inherited disease characterized by hamartomatous polyps of the gastrointestinal tract and pigmented macules of the lips and buccal mucosa.84 Germline mutations of STK11/LKB1 are responsible for Peutz-Jeghers syndrome, and patients with this syndrome have a very high lifetime risk for pancreatic cancer (up to 132-fold).84,85 As we described above, pancreatic cancers from patients with Peutz-Jeghers syndrome develop as IPMNs.
Familial atypical multiple mole melanoma (FAMMM) is an autosomal, dominantly inherited disorder characterized by multiple nevi and atypical nevi and an increased risk for malignant melanoma.86,87 Germline mutations of CDKN2A/p16 cause FAMMM, and patients with FAMMM and mutated CDKN2A/p16 have a 47-fold increased risk for pancreatic cancer.88 Hereditary pancreatitis is characterized by recurrent attacks of pancreatitis at a young age. Germline mutations of PRSS1 are associated with a markedly increased risk for hereditary pancreatitis and a 53-fold increased risk for pancreatic cancer.89–92 Variants in SPINK1 are associated with a moderate increased risk for pancreatitis.
Hereditary nonpolyposis colorectal cancer syndrome (HNPCC) is an autosomal, dominantly inherited disease characterized by early onset of right-sided colon cancer as well as an increased risk for endometrial cancer and carcinomas of the small intestine, stomach, endometrium, ovary, bile duct, and kidney.93 Germline mutations of mismatch repair genes, including MLH1, MSH2, PMS1, PMS2, and MSH6, are associated with HNPCC. When pancreatic cancers arise in patients with HNPCC, they usually have a characteristic medullary phenotype.
Familial adenomatous polyposis (FAP) syndrome is an autosomal, dominantly inherited disease characterized by the presence of more than hundreds of polyps in the colon at an early age.94,95 Germline mutation of APC is linked with FAP. Patients with FAP have a 4-fold increased risk for pancreatic cancer.96
Genetic syndromes associated with familial pancreatic cancer are summarized in Table 2.
Table 2.
Genetic Syndrome | Gene Symbol | Relative Risk of Developing Pancreatic Cancer (Fold) | Histologic Feature of Pancreatic Neoplasm | Extrapancreatic Cancer | Source, y |
---|---|---|---|---|---|
No familial history | None | 1 | Ductal adenocarcinoma Intraductal papillary mucinous neoplasm, ductal adenocarcinoma, pancreatoblastoma | Unknown | |
Familial adenomatous polyposis | APC | 4 | Colorectum, small intestine, stomach | Giardiello et al,96 1993 | |
Familial atypical multiple mole melanoma | CDKN2A/p16 | 13–22 | Ductal adenocarcinoma | Melanoma | Gruis et al,86 1995 de Snoo et al,88 2008 |
Familial pancreatic cancer | Unknown | 2–32 | Ductal adenocarcinoma | Amundadottir et al,75 2004 Klein et al,76 2004 |
|
Hereditary breast and ovarian cancer | BRCA2, BRCA1, FANCC, FANCG, PALB2 | 3.5–10 | Ductal adenocarcinoma | Breast, ovary, prostate | Hruban et al,77 1999 Hahn et al,78 2003 van Asperen et al,79 2005 |
Hereditary pancreatitis | PRSS1, SPINK1 | 53 | Ductal adenocarcinoma | None | de las Heras-Castano et al,89 2009 Lowenfels et al,90 1997 Schneider et al,91 2002 Whitcomb et al,92 1996 |
Hereditary nonpolyposis colorectal cancer syndrome | MLH1, MSH2 | Increased | Medullary carcinoma | Colorectum, small intestine, endometrium | Wilentz et al,61 2000 |
Peutz-Jeghers syndrome | SKT11/LKB1 | 132 | Intraductal papillary mucinous neoplasm, ductal adenocarcinoma | Small intestine, colorectum, esophagus, stomach, bile duct, lung, breast, ovary, uterus | Zbuk & Eng,84 2007 Giardiello et al,85 2000 |
EPIGENETICS OF PANCREATIC CANCER
Epigenetics is defined as heritable changes in gene expression without accompanying changes in DNA sequence.97 The main epigenetic mechanisms that may affect gene expression include DNA methylation, histone modification, and microRNA expression.
1. DNA Methylation
DNA methylation is the covalent binding of a methyl group (CH3-) to the 5-carbon of cytosine residues. This methyl-group binding is catalyzed and maintained by a family of enzymes, DNA methyltransferases (DNMTs), including DNMT1, DNMT3A, and DNMT3B. DNMT1 is involved in preserving parental methylation patterns and transferring these patterns to offspring. DNMT3A and DNMT3B are involved in de novo methylation.98–100 Approximately 80% of pancreatic cancers overexpress dnmt1 protein.101
A major pattern of DNA methylation occurs in CpG islands. CpG islands are stretches of DNA with a high CG nucleotide content (>50%).102 The CpG islands are frequently located near the transcriptional start sites of genes. About 60% of human genes have associated CpG islands; for many years CpG islands were thought to be unmethylated except during genomic imprinting and X-chromosome inactivation,103 but more recent evidence indicates that some CpG islands are methylated in a tissue-specific manner,104 and CpG island methylation increases with age at many loci.105,106 Aberrant hypermethylation of promoter CpG islands is tightly associated with gene silencing and may be associated with loss of tumor suppressor function in cancer.107
Aberrant Hypermethylation in Pancreatic Cancer
Several classic tumor suppressor genes, as well as increasing numbers of functionally important genes, show aberrant promoter CpG island hypermethylation in a subset of pancreatic cancers. The first tumor suppressor gene that was shown to undergo promoter hypermethylation and silencing in pancreatic cancer was CDKN2A/p16.21 Other genetically inactivated tumor suppressor genes in pancreatic cancers, including TP53, MADH4/DPC4, and STK11/LKB1, have not been shown to undergo epigenetic silencing by DNA methylation.
MLH1 on chromosome arm 3p undergoes DNA methylation in pancreatic cancers and is associated with microsatellite instability in medullary carcinomas.22,108,109 The CDH1 gene on chromosome arm 16q, which encodes E-cadherin protein, shows aberrant methylation in a small fraction of pancreatic cancers.22
SPARC, located on chromosome arm 5q, encodes a calcium-binding protein that interacts with extracellular matrix.110 Sparc has effects on cellular migration, proliferation, angiogenesis during wound healing, cell-matrix adhesion, and tissue remodeling.110 In pancreatic and other cancers, Sparc expression is usually lost through abnormal DNA methylation.110 Pancreatic cancer–associated peritumoral fibroblasts often express Sparc, and patients with pancreatic cancer and sparc-expressing peritumoral fibroblasts were reported to have a poorer survival in 1 study.111
Other cancer-related genes that have been shown to undergo abnormal methylation and induced gene silencing include RELN,112 CCND2,105 TFPI2,113 RUNX3,114 SOCS-1,115 and TSLC1/IGSF4.116
Genome-wide screening has made it possible to identify epigenetic alterations in novel genes within the setting of pancreatic cancer. Ueki and colleagues4 used methylated CpG island amplification with representational difference analysis to identify differentially methylated CpG islands in pancreatic cancer. PENK (preproenkephalin) on chromosome arm 8q was one of the genes identified by this method, and more than 90% of pancreatic cancers in this study had aberrantly methylated PENK.4,117 Using oligo-nucleotide microarrays, Sato and colleagues5 identified a total of 475 candidate genes that were induced by a DNMT inhibitor (5-aza-2′-deoxycytidine) in 4 pancreatic cancer cell lines, but not in HPDE (a nonneoplastic pancreatic ductal epithelial cell line). Of these 475 genes, UCHL1 on chromosome arm 4p was methylated in all 42 pancreatic cancers studied.5 RPRM on chromosome arm 2q was methylated in 80% of pancreatic cancers studied and was associated with a worse prognosis.118 More recently, Omura and colleagues3 applied the methylated CpG island amplification technique to an Agilent 44K promoter microarray (Agilent Technologies, Santa Clara, California) and identified 606 differentially methylated genes in pancreatic cancer cell lines compared to normal pancreas.
A selected list of genes that are aberrantly hypermethylated in pancreatic cancer is summarized in Table 3.
Table 3.
Gene Symbol | Gene Name | Epigenetic Alteration | Chromosome Site | Known or Predicted Function | Methylation in Pancreatic Cancer Cell Lines, No. (%) | Methylation in Primary or Xenografted Pancreatic Cancer, No. (%) | Source, y |
---|---|---|---|---|---|---|---|
PENK | Preproenkephalin | Hypermethylation | 8q23-q24 | Neuropeptide precursor | 11/11 (100) | 43/47 (91) | Ueki et al,4 2001 Fukushima et al,117 2002 |
UCHL1 | Ubiquitin carboxyl-terminal esterase L1 (ubiquitin thiolesterase) | Hypermethylation | 4p14 | Ubiquitin hydroxylase | 22/22 (100) | 42/42 (100) | Sato et al,5 2003 |
MDF-1 | MAD (yeast Mitosis Arrest DeFicient) related | Hypermethylation | 11q13 | Glycogen metabolism | 45/47 (96) | Not determined | Omura et al,3 2008 |
NPTX2 | Neuronal pentraxin II | Hypermethylation | 7q21.3-q22.1 | Neuronal transport | 21/22 (95) | 20/20 (100) | Sato et al,5 2003 |
SPARC/ON | Secreted protein, acidic, cysteine-rich (osteonectin) | Hypermethylation | 5q31.3-q32 | Cell-cycle progression inhibition, cell-matrix interaction | 16/17 (94) | 21/24 (88) | Sato et al,110 2003 |
RPRM | Reprimo, TP53-dependent G2 arrest mediator candidate | Hypermethylation | 2q23.3 | P53-induced G2/M cell-cycle arrest | 20/22 (91) | 16/20 (80) | Sato et al,118 2006 |
BNIP3 | BCL2/adenovirus E1B 19 kDa interacting protein 3 | Hypermethylation | 10q26.3 | Hypoxia-induced cell death | 9/10 (90) | 8/10 (80) | Okami et al,156 2004 |
miR9-1 | MicroRNA 9-1 | Hypermethylation | 1q22 | miRNA translation control | 42/47 (89) | Not determined | Omura et al,3 2008 |
SERPINB5 | Serpin peptidase inhibitor, clade B, member 5 (maspin) | Hypomethylation | 18q21.3 | Regulation of cell motility and cell death | 20/23 (87) | 32/34 (94) | Sato et al,132 2003 Fitzgerald et al,157 2003 Ohike et al,158 2003 |
CCND2 | Cyclin D2 | Hypermethylation | 12p13 | Cell-cycle control | 19/22 (86) | 71/109 (65) | Matsubayashi et al,105 2003 |
ZNF415 | Zinc finger protein 415 | Hypermethylation | 19q13.42 | 40/47 (86) | Not determined | Omura et al,3 2008 | |
CLDN4 | Claudin-4 | Hypomethylation | 7q11.23 | Cell adhesion/invasion | 17/20 (85) | 33/37 (89) | Sato et al,132 2003 |
SFN | Stratifin (14-3-3 σ) | Hypomethylation | 1p35 | P53-induced G2/M cell-cycle arrest | 17/20 (85) | 36/37 (97) | Sato et al,132 2003 Iacobuzio-Donahue et al,159 2003 |
LCN2 | Lipocalin-2 | Hypomethylation | 9q34 | Epithelial differentiation | 17/20 (85) | 34/37 (92) | Sato et al,132 2003 |
TFPI2 | Tissue factor pathway inhibitor 2 | Hypermethylation | 7q22 | Serine protease inhibitor | 14/17 (82) | 102/140 (73) | Sato et al,113 2005 |
CNTNAP2 | Contactin-associated protein-like 2 | Hypermethylation | 7q35-q36 | Higher cortical function | 39/47 (82) | Not determined | Omura et al,3 2008 |
CDKN1C/p57 | Cyclin-dependent kinase inhibitor 1C | Hypermethylation | 11p15.5 | Cyclin-dependent kinase inhibitor | 7/9 (78) | Not determined | Sato et al,119 2005 |
SIP1 | Survival of motor neuron protein-interacting protein 1 | Hypermethylation | 14q13-q21 | Assembly of spliceosomal snRNP | 11/15 (73) | 34/35 (97) | Li et al,152 2010 |
ELOVL4 | Elongation of very-long-chain fatty acids (FEN1/Elo2, SUR4/Elo3, yeast)–like 4 | Hypermethylation | 6q14 | Fatty acid synthesis | 32/47 (68) | Not determined | Omura et al,3 2008 |
TFF2 | Trefoil factor 2 | Hypomethylation | 21q22.3 | Secretory polypeptide/epithelial repair | 13/20 (65) | 31/37 (84) | Sato et al,132 2003 |
FOXE1 | Forkhead box E1 (thyroid transcription factor 2) | Hypermethylation | 9q22 | Thyroid transcription factor | 14/22 (64) | 15/20 (75) | Sato et al,5 2003 |
S100P | S100 calcium-binding protein P | Hypomethylation | 4p16 | Cell-cycle progression and differentiation | 13/23 (57) | 30/34 (88) | Sato et al,132 2003 |
RARB | Retinoic acid receptor, β | Hypermethylation | 3p24 | Cell-growth control | 5/9 (56) | 4/36 (11) | Ueki et al,22 2000 |
S100A4 | S100 calcium-binding protein A4 | Hypomethylation | 1q21 | Motility, invasion, tubulin polymerization | 10/20 (50) | 28/37 (76) | Rosty et al,131 2002 Sato et al,132 2003 |
CDKN2A/p16 | Cyclin-dependent kinase inhibitor 2A | Hypermethylation | 9P21 | Cyclin-dependent kinase inhibitor | 3/9 (33) | 5/36 (14) | Schutte et al,21 1997 Ueki et al,22 2000 |
MSLN | Mesothelin | Hypomethylation | 16p13.3 | Cell surface antigen/cell adhesion | 8/20 (40) | 34/37 (29) | Sato et al,132 2003 |
SOCS1 | Suppressor of cytokine signaling 1 | Hypermethylation | 16p13.13 | Inhibitor of JAK/STAT pathway | 6/19 (32) | 13/60 (22) | Fukushima et al,115 2003 |
PSCA | Prostate stem cell antigen | Hypomethylation | 8q24.2 | Cell surface antigen/cell differentiation | 6/20 (30) | 20/37 (54) | Sato et al,132 2003 |
CADM1/TSLC1 | Cell adhesion molecule 1 | Hypermethylation | 11q23.2 | Cell-cell, cell-matrix interaction | 4/17 (24) | 25/91 (27) | Jansen et al,116 2002 |
Abbreviations: JAK/STAT, Janus kinase/signal transducer and activator of transcription; miRNA, microRNA; snRNP, small nuclear ribonucleoprotein.
Aberrant Methylation in Precursor Lesions
The discovery of abnormal methylation in pancreatic cancer has been followed by the investigation of methylation in precursor lesions. Many genes that are epigenetically silenced in pancreatic cancers also are silenced or have reduced expression in precursor lesions of pancreatic cancer. For example, global gene expression profiles of IPMN were compared with those of normal pancreatic ductal epithelial samples.119 CDKN1C/p57KIP2 on chromosome arm 11p codes for an inhibitor of cyclin/CDK complexes and negative regulator of cellular proliferation.120,121 Partial methylation of the CDKN1C/p57KIP2 promoter CpG islands in IPMNs and pancreatic cancer cell lines was correlated with a corresponding decrease in cdkn1c protein expression.119
Other genes identified in precursor lesions include PENK, CDKN2A/p16, STK11/LKB1, SPARC, SFRP1/SARP2 (chromosome arm 8p), TSLC1, RELN (chromosome arm 7q), TFPI2, CLDN5 (chromosome arm 22q), and UCHL1 in IPMNs37,122,123; PENK, CDKN2A/p16, CLDN5, NPTX2, RPRM, SFRP1/SARP2, and LHX1 (chromosome arm 11p) in PanINs117,118,124; and CDKN2A/p16 in mucinous cystic neoplasms.56 A selected list of genes that are aberrantly hypermethylated in pancreatic precursor lesions is summarized in Table 4.
Table 4.
Gene Symbol | Gene Name | Precursor Lesions | Methylation in Low-Grade Dysplasia (PanIN-1 or Low-Grade Dysplasia of IPMN or MCN), No. (%) | Methylation in Moderate-Grade Dysplasia (PanIN-2 or Moderate-Grade Dysplasia of IPMN or MCN), No. (%) | Methylation in High-Grade Dysplasia (PanIN-3 or High-Grade Dysplasia of IPMN or MCN), No. (%) | Methylation in Precursor in Total, No. (%) | Source, y |
---|---|---|---|---|---|---|---|
PENK | Preproenkephalin | IPMN | 1/6 (17) | 4/12 (33) | 27/32 (84) | 32/50 (64) | Sato et al,122 2002 |
PanIN | 5/67 (7) | 5/22 (23) | 6/13 (46) | 16/108 (15) | Fukushima et al,117 2002 | ||
PanIN | 1/38 (3) | 1/14 (7) | 7/12 (58) | 9/64 (14) | Sato et al,124 2008 | ||
CDKN2A/p16 | Cyclin-dependent kinase inhibitor 2A | IPMN | 0/6 (0) | 0/12 (0) | 7/32 (22) | 7/50 (14) | Sato et al,122 2002 |
PanIN | 4/63 (6) | 1/22 (5) | 3/14 (21) | 8/99 (8) | Fukushima et al,117 2002 | ||
PanIN | 3/38 (8) | 1/15 (7) | 3/11 (27) | 7/64 (11) | Sato et al,124 2008 | ||
MCN | 1/10 (10) | 1/4 (25) | NA | 2/14 (14) | Kim et al,56 2003 | ||
SPARC/ON | Secreted protein, acidic, cysteine-rich (osteonectin) | IPMN | 7/12 (58) | 7/12 (58) | 16/22 (73) | 30/48 (63) | Hong et al,123 2008 |
PanIN | 10/36 (21) | 3/14 (21) | 3/10 (30) | 16/60 (27) | Sato et al,124 2008 | ||
SFRP1/SARP2 | Secreted frizzled– related protein 1 | IPMN | 6/12 (50) | 8/12 (67) | 21/23 (91) | 35/57 (61) | Hong et al,123 2008 |
PanIN | 2/37 (5) | 3/15 (20) | 10/12 (83) | 15/64 (23) | Sato et al,124 2008 | ||
NPTX2 | Neuronal pentraxin 2 | PanIN | 2/35 (8) | 6/13 (46) | 4/12 (33) | 12/60 (20) | Sato et al,124 2008 |
CADM1/TSLC1 | Cell adhesion molecule 1 | IPMN | 6/12 (50) | 8/12 (67) | 21/23 (91) | 35/57 (61) | Hong et al,123 2008 |
RELN | Reelin | IPMN | 3/12 (25) | 4/12 (33) | 11/23 (48) | 18/57 (32) | Hong et al,123 2008 |
TFPI2 | Tissue factor pathway inhibitor 2 | IPMN | 3/12 (25) | 5/12 (42) | 20/23 (87) | 28/57 (49) | Hong et al,123 2008 |
CLDN5 | Claudin-5 | IPMN | 4/12 (33) | 5/12 (42) | 15/23 (65) | 24/57 (42) | Hong et al,123 2008 |
PanIN | 3/35 (9) | 1/15 (7) | 4/11 (36) | 8/61 (13) | Sato et al,124 2008 | ||
UCHL1 | Ubiquitin carboxyl-terminal esterase L1 (ubiquitin thiolesterase) | IPMN | 7/12 (58) | 10/12 (83) | 21/23 (19) | 38/57 (67) | Hong et al,123 2008 |
RPRM | Reprimo, TP53-dependent G2 arrest mediator candidate | PanIN | 8/36 (22) | 3/15 (20) | 8/12 (67) | 19/63 (30) | Sato et al,118 2006 |
LHX1 | LIM homeobox 1 | PanIN | 3/37 (8) | 1/15 (7) | 5/12 (42) | 9/64 (14) | Sato et al,124 2008 |
Abbreviations: IPMN, intraductal papillary mucinous neoplasm; MCN, mucinous cystic neoplasm; NA; PanIN, pancreatic intraepithelial neoplasia.
The degree of methylation for these genes positively correlates with the degree of cytologic and architectural atypia. These findings suggest that aberrant CpG island methylation begins in the earliest stages of precursor lesions, such as PanINs, IPMNs, and MCNs, and their prevalence progressively increases during pancreatic carcinogenesis.
Aberrant Hypomethylation in Pancreatic Cancer
In addition to hypermethylation as a mechanism of carcinogenesis, aberrant loss of methylation (hypomethylation of DNA) is also common in pancreatic adenocarcinomas. Hypomethylation can be detected at the genomic scale (global hypomethylation) and at the sequence-specific level (regional hypomethylation). Although global DNA hypomethylation associated with cancer was firstly described in the early 1980s,125,126 its significance is not known, but it may contribute to genomic instability. Folate and vitamin B12 deficiency can cause global DNA hypomethylation, which is associated with decreased levels of the methyl-group donor S-adenosylmethionine. Decreased DNA methylation results in decreased thymidine synthesis from uracil.127 Misplacement of uracil into thymidine leads to an imbalance of nucleotide pools and an increased frequency of DNA strand breaks; this can lead to genomic instability that can promote the development of cancer.128,129 Pancreatic cancers with defective methylenetetrahydrofolate reductase genotypes have more DNA hypomethylation, which is associated with increased chromosomal loss and genomic instability.130
DNA hypomethylation occurs at the 5′ regions of certain genes in pancreatic cancer and is associated with overexpression of the encoded protein. Thus, whereas hypermethylation results in overregulation and silencing of gene and protein expression, hypomethylation can result in loss of regulation and the promotion of gene and protein expression. S100A4 is linked with hypomethylation at specific CpG sites within the first intron and is associated with protein overexpression.131,132 Other frequently hypomethylated genes, including CLDN4 (chromosome arm 7q, encoding claudin-4), LCN2 (chromosome arm 9q, encoding lipocalin-2), SFN/14-3-3σ (chromosome arm 18q), TFF2 (chromosome arm 21q, encoding trefoil factor 2), MSLN (chromosome arm 16p, encoding mesothelin), and PSCA (8q, encoding prostate stem cell antigen), are overexpressed in pancreatic cancer cells in comparison with normal pancreatic duct.132 With oligo-nucleotide microarray technologies, 2 additional genes, S100P (chromosome arm 4p) and SERPINB5 (chromosome arm 18q, encoding maspin), have been identified as being hypomethylated and are overexpressed.6 A selected list of genes that are aberrantly hypomethylated in pancreatic cancer is summarized in Table 2.
2. MicroRNAs
Aberrant MicroRNA Expression in Pancreatic Cancers and Precursor Lesions
MicroRNAs (miRNAs) are a recently described family of small, nonprotein-coding RNA molecules (18 to 24 nucleotides) that regulate transcription of target messenger RNAs.133 More than 400 miRNAs in the human genome have been described and many are implicated in the regulation of cellular differentiation, proliferation, and apoptosis.23 Aberrant miRNA expression has been described in many types of cancers.134,135 Several mechanisms are involved in aberrant miRNA expression, including genetic (amplification and deletion)136–138 and epigenetic (chromatin modification, DNA methylation) alterations139–141 and transcription factor regulation.142,143
Pancreatic ductal adenocarcinomas have been shown to aberrantly express numerous miRNAs, including miR-200, miR-34, miR-21, miR-155, miR-221, and miR-222.144–151 For example, Li and colleagues152 have demonstrated hypomethylation and overexpression of miR-200a and miR-200b. Aberrant expression of some of these miRNAs is evident in PanINs. For example, miR-155 overexpression is evident in PanIN-2 lesions and aberrant miR-21 expression is evident in PanIN-3 lesions.153 Similarly, Habbe et al154 have reported abnormal miR-21 and miR-155 expression in IPMN lesions.
CONCLUSIONS
Pancreatic ductal adenocarinoma continues to be a fatal cancer that is difficult to treat. In the past decade, major advances have been made in the understanding of the earliest histologic and molecular changes that occur in precursor lesions and cancers of the pancreas. Subclassification of pancreatic adenocarcinomas according to its histologic features and molecular alterations could have important therapeutic and prognostic importance. In addition, the identification of molecular signatures that identify the earliest changes of carcinogenesis may lead to the earlier detection of pancreatic cancer. The survival data for pancreatic cancer clearly illustrate that patients do much better with earlier detection and surgical resection regardless of adjuvant chemotherapy or radiotherapy intervention. Understanding the signature of molecular alterations that occur before the development of invasive pancreatic cancer may lead to improved detection and survival in pancreatic cancer.
Acknowledgments
This work was supported by National Institutes of Health (NIH) grants (P50-CA62924, R01-CA120432, RO1-CA97075) and the Michael Rolfe Foundation.
Footnotes
The authors have no relevant financial interest in the products or companies described in this article.
Presented at the 9th Spring Seminar of the Korean Pathologists Association of North America (KOPANA), Washington, District of Columbia, March 18–20, 2010, in conjunction with the 99th Annual Meeting of the United States and Canadian Academy of Pathology (USCAP).
References
- 1.Jemal A, Siegel R, Xu J, Ward E. Cancer statistics, 2010. CA Cancer J Clin. 2010;60:277–300. doi: 10.3322/caac.20073. [DOI] [PubMed] [Google Scholar]
- 2.Jones S, Hruban RH, Kamiyama M, et al. Exomic sequencing identifies PALB2 as a pancreatic cancer susceptibility gene. Science. 2009;324(5924):217. doi: 10.1126/science.1171202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Omura N, Li CP, Li A, et al. Genome-wide profiling of methylated promoters in pancreatic adenocarcinoma. Cancer Biol Ther. 2008;7(7):1146–1156. doi: 10.4161/cbt.7.7.6208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Ueki T, Toyota M, Skinner H, et al. Identification and characterization of differentially methylated CpG islands in pancreatic carcinoma. Cancer Res. 2001;61(23):8540–8546. [PubMed] [Google Scholar]
- 5.Sato N, Fukushima N, Maitra A, et al. Discovery of novel targets for aberrant methylation in pancreatic carcinoma using high-throughput microarrays. Cancer Res. 2003;63(13):3735–3742. [PubMed] [Google Scholar]
- 6.Sato N, Fukushima N, Matsubayashi H, Goggins M. Identification of maspin and S100P as novel hypomethylation targets in pancreatic cancer using global gene expression profiling. Oncogene. 2004;23(8):1531–1538. doi: 10.1038/sj.onc.1207269. [DOI] [PubMed] [Google Scholar]
- 7.Jones S, Zhang X, Parsons DW, et al. Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science. 2008;321(5897):1801–1806. doi: 10.1126/science.1164368. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.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(2):545–554. [PMC free article] [PubMed] [Google Scholar]
- 9.Hingorani SR, Tuveson DA. Ras redux: rethinking how and where Ras acts. Curr Opin Genet Dev. 2003;13(1):6–13. doi: 10.1016/s0959-437x(02)00017-5. [DOI] [PubMed] [Google Scholar]
- 10.Tada M, Ohashi M, Shiratori Y, et al. Analysis of K-ras gene mutation in hyperplastic duct cells of the pancreas without pancreatic disease. Gastroenterology. 1996;110(1):227–231. doi: 10.1053/gast.1996.v110.pm8536861. [DOI] [PubMed] [Google Scholar]
- 11.Moskaluk CA, Hruban RH, Kern SE. p16 and K-ras gene mutations in the intraductal precursors of human pancreatic adenocarcinoma. Cancer Res. 1997;57(11):2140–2143. [PubMed] [Google Scholar]
- 12.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(4):1255–1260. doi: 10.1016/S0002-9440(10)63485-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Ruggeri BA, Huang L, Wood M, Cheng JQ, Testa JR. Amplification and overexpression of the AKT2 oncogene in a subset of human pancreatic ductal adenocarcinomas. Mol Carcinog. 1998;21(2):81–86. [PubMed] [Google Scholar]
- 14.Cheng JQ, Ruggeri B, Klein WM, et al. Amplification of AKT2 in human pancreatic cells and inhibition of AKT2 expression and tumorigenicity by antisense RNA. Proc Natl Acad Sci U S A. 1996;93(8):3636–3641. doi: 10.1073/pnas.93.8.3636. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Fu B, Luo M, Lakkur S, Lucito R, Iacobuzio-Donahue CA. Frequent genomic copy number gain and overexpression of GATA-6 in pancreatic carcinoma. Cancer Biol Ther. 2008;7(10):1593–1601. doi: 10.4161/cbt.7.10.6565. [DOI] [PubMed] [Google Scholar]
- 16.Kwei KA, Bashyam MD, Kao J, et al. Genomic profiling identifies GATA6 as a candidate oncogene amplified in pancreatobiliary cancer. PLoS Genet. 2008;4(5):e1000081. doi: 10.1371/journal.pgen.1000081. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.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(1):27–32. doi: 10.1038/ng0994-27. [DOI] [PubMed] [Google Scholar]
- 18.Shiota K, Yanagimachi R. Epigenetics by DNA methylation for development of normal and cloned animals. Differentiation. 2002;69(4–5):162–166. doi: 10.1046/j.1432-0436.2002.690406.x. [DOI] [PubMed] [Google Scholar]
- 19.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(11):3025–3033. [PubMed] [Google Scholar]
- 20.Wilentz RE, Su GH, Dai JL, et al. Immunohistochemical labeling for dpc4 mirrors genetic status in pancreatic adenocarcinomas: a new marker of DPC4 inactivation. Am J Pathol. 2000;156(1):37–43. doi: 10.1016/S0002-9440(10)64703-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.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(15):3126–3130. [PubMed] [Google Scholar]
- 22.Ueki T, Toyota M, Sohn T, et al. Hypermethylation of multiple genes in pancreatic adenocarcinoma. Cancer Res. 2000;60(7):1835–1839. [PubMed] [Google Scholar]
- 23.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]
- 24.Moore PS, Sipos B, Orlandini S, et al. Genetic profile of 22 pancreatic carcinoma cell lines: analysis of K-ras, p53, p16 and DPC4/Smad4. Virchows Arch. 2001;439(6):798–802. doi: 10.1007/s004280100474. [DOI] [PubMed] [Google Scholar]
- 25.Scarpa A, Capelli P, Mukai K, et al. Pancreatic adenocarcinomas frequently show p53 gene mutations. Am J Pathol. 1993;142(5):1534–1543. [PMC free article] [PubMed] [Google Scholar]
- 26.Vogelstein B, Kinzler KW. Cancer genes and the pathways they control. Nat Med. 2004;10(8):789–799. doi: 10.1038/nm1087. [DOI] [PubMed] [Google Scholar]
- 27.Iacobuzio-Donahue CA, Song J, Parmiagiani G, Yeo CJ, Hruban RH, Kern SE. Missense mutations of MADH4: characterization of the mutational hot spot and functional consequences in human tumors. Clin Cancer Res. 2004;10(5):1597–1604. doi: 10.1158/1078-0432.ccr-1121-3. [DOI] [PubMed] [Google Scholar]
- 28.Hahn SA, Schutte M, Hoque AT, et al. DPC4, a candidate tumor suppressor gene at human chromosome 18q21.1. Science. 1996;271(5247):350–353. doi: 10.1126/science.271.5247.350. [DOI] [PubMed] [Google Scholar]
- 29.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(11):1544–1548. doi: 10.1097/00000478-200011000-00011. [DOI] [PubMed] [Google Scholar]
- 30.Siegel PM, Massague J. Cytostatic and apoptotic actions of TGF-beta in homeostasis and cancer. Nat Rev Cancer. 2003;3(11):807–821. doi: 10.1038/nrc1208. [DOI] [PubMed] [Google Scholar]
- 31.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(7):2002–2006. [PubMed] [Google Scholar]
- 32.Tascilar M, Skinner HG, Rosty C, et al. The SMAD4 protein and prognosis of pancreatic ductal adenocarcinoma. Clin Cancer Res. 2001;7(12):4115–4121. [PubMed] [Google Scholar]
- 33.Blackford A, Serrano OK, Wolfgang CL, et al. SMAD4 gene mutations are associated with poor prognosis in pancreatic cancer. Clin Cancer Res. 2009;15(14):4674–4679. doi: 10.1158/1078-0432.CCR-09-0227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Schutte M, Hruban RH, Hedrick L, et al. DPC4 gene in various tumor types. Cancer Res. 1996;56(11):2527–2530. [PubMed] [Google Scholar]
- 35.Iacobuzio-Donahue CA, Fu B, Yachida S, et al. DPC4 gene status of the primary carcinoma correlates with patterns of failure in patients with pancreatic cancer. J Clin Oncol. 2009;27(11):1806–1813. doi: 10.1200/JCO.2008.17.7188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Su GH, Hruban RH, Bansal RK, et al. Germline and somatic mutations of the STK11/LKB1 Peutz-Jeghers gene in pancreatic and biliary cancers. Am J Pathol. 1999;154(6):1835–1840. doi: 10.1016/S0002-9440(10)65440-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.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 Dec. 2001;159(6):2017–2022. doi: 10.1016/S0002-9440(10)63053-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.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(23):5329–5332. [PubMed] [Google Scholar]
- 39.Su GH, Hilgers W, Shekher MC, et al. Alterations in pancreatic, biliary, and breast carcinomas support MKK4 as a genetically targeted tumor suppressor gene. Cancer Res. 1998;58(11):2339–2342. [PubMed] [Google Scholar]
- 40.Teng DH, Perry WL, III, Hogan JK, et al. Human mitogen-activated protein kinase kinase 4 as a candidate tumor suppressor. Cancer Res. 1997;57(19):4177–4182. [PubMed] [Google Scholar]
- 41.Su GH, Bansal R, Murphy KM, et al. ACVR1B (ALK4, activin receptor type 1B) gene mutations in pancreatic carcinoma. Proc Natl Acad Sci U S A. 2001;98(6):3254–3257. doi: 10.1073/pnas.051484398. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Hruban RH, Pitman MB, Klimstra DS. Atlas of Tumor Pathology. Washington, DC: Armed Forces Institute of Pathology; 2007. Tumors of the Pancreas. 4th series. [Google Scholar]
- 43.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(5):579–586. doi: 10.1097/00000478-200105000-00003. [DOI] [PubMed] [Google Scholar]
- 44.Rogers CD, van der Heijden MS, Brune K, et al. The genetics of FANCC and FANCG in familial pancreatic cancer. Cancer Biol Ther. 2004;3(2):167–169. doi: 10.4161/cbt.3.2.609. [DOI] [PubMed] [Google Scholar]
- 45.Maitra A, Fukushima N, Takaori K, Hruban RH. Precursors to invasive pancreatic cancer. Adv Anat Pathol. 2005;12(2):81–91. doi: 10.1097/01.pap.0000155055.14238.25. [DOI] [PubMed] [Google Scholar]
- 46.van Heek NT, Meeker AK, Kern SE, et al. Telomere shortening is nearly universal in pancreatic intraepithelial neoplasia. Am J Pathol. 2002;161(5):1541–1547. doi: 10.1016/S0002-9440(10)64432-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Hruban RH, Goggins M, Parsons J, Kern SE. Progression model for pancreatic cancer. Clin Cancer Res. 2000;6(8):2969–2972. [PubMed] [Google Scholar]
- 48.Hruban RH, Maitra A, Goggins M. Update on pancreatic intraepithelial neoplasia. Int J Clin Exp Pathol. 2008;1(4):306–316. [PMC free article] [PubMed] [Google Scholar]
- 49.Schonleben F, Qiu W, Remotti HE, Hohenberger W, Su GH. PIK3CA, KRAS, and BRAF mutations in intraductal papillary mucinous neoplasm/carcinoma (IPMN/C) of the pancreas. Langenbecks Arch Surg. 2008;393(3):289–296. doi: 10.1007/s00423-008-0285-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Sessa F, Solcia E, Capella C, et al. Intraductal papillary-mucinous tumours represent a distinct group of pancreatic neoplasms: an investigation of tumour cell differentiation and K-ras, p53 and c-erbB-2 abnormalities in 26 patients. Virchows Arch. 1994;425(4):357–367. doi: 10.1007/BF00189573. [DOI] [PubMed] [Google Scholar]
- 51.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 [discussion in Ann Surg. 1997;226(4):498–500] Ann Surg. 1997;226(4):491–498. doi: 10.1097/00000658-199710000-00010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Abe T, Fukushima N, Brune K, et al. Genome-wide allelotypes of familial pancreatic adenocarcinomas and familial and sporadic intraductal papillary mucinous neoplasms. Clin Cancer Res. 2007;13(20):6019–6025. doi: 10.1158/1078-0432.CCR-07-0471. [DOI] [PubMed] [Google Scholar]
- 53.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(3):755–761. doi: 10.1016/S0002-9440(10)64589-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.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 [discussion in Ann Surg. 1999;230(4):509–511] Ann Surg. 1999;230(4):501–509. doi: 10.1097/00000658-199910000-00006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Thompson LD, Becker RC, Przygodzki RM, Adair CF, Heffess CS. Mucinous cystic neoplasm (mucinous cystadenocarcinoma of low-grade malignant potential) of the pancreas: a clinicopathologic study of 130 cases. Am J Surg Pathol. 1999;23(1):1–16. doi: 10.1097/00000478-199901000-00001. [DOI] [PubMed] [Google Scholar]
- 56.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(11):1086–1094. doi: 10.1097/01.MP.0000094088.37888.A6. [DOI] [PubMed] [Google Scholar]
- 57.Adsay NV, Pierson C, Sarkar F, et al. Colloid (mucinous noncystic) carcinoma of the pancreas. Am J Surg Pathol. 2001;25(1):26–42. doi: 10.1097/00000478-200101000-00003. [DOI] [PubMed] [Google Scholar]
- 58.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(5):651–659. doi: 10.1038/modpathol.2009.15. [DOI] [PubMed] [Google Scholar]
- 59.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(6):1501–1507. [PMC free article] [PubMed] [Google Scholar]
- 60.Hruban RH, Adsay NV. Molecular classification of neoplasms of the pancreas. Hum Pathol. 2009;40(5):612–623. doi: 10.1016/j.humpath.2009.01.008. [DOI] [PubMed] [Google Scholar]
- 61.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(5):1641–1651. doi: 10.1016/S0002-9440(10)65035-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Nakata B, Wang YQ, Yashiro M, et al. Negative hMSH2 protein expression in pancreatic carcinoma may predict a better prognosis of patients. Oncol Rep. 2003;10(4):997–1000. [PubMed] [Google Scholar]
- 63.Ribic CM, Sargent DJ, Moore MJ, et al. Tumor microsatellite-instability status as a predictor of benefit from fluorouracil-based adjuvant chemotherapy for colon cancer. N Engl J Med. 2003;349(3):247–257. doi: 10.1056/NEJMoa022289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Adsay NV, Merati K, Basturk O, 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(7):839–848. doi: 10.1097/00000478-200407000-00001. [DOI] [PubMed] [Google Scholar]
- 65.Adsay NV, Merati K, Nassar H, et al. Pathogenesis of colloid (pure mucinous) carcinoma of exocrine organs: coupling of gel-forming mucin (MUC2) production with altered cell polarity and abnormal cell-stroma interaction may be the key factor in the morphogenesis and indolent behavior of colloid carcinoma in the breast and pancreas. Am J Surg Pathol. 2003;27(5):571–578. doi: 10.1097/00000478-200305000-00002. [DOI] [PubMed] [Google Scholar]
- 66.Hoorens A, Prenzel K, Lemoine NR, Kloppel G. Undifferentiated carcinoma of the pancreas: analysis of intermediate filament profile and Ki-ras mutations provides evidence of a ductal origin. J Pathol. 1998;185(1):53–60. doi: 10.1002/(SICI)1096-9896(199805)185:1<53::AID-PATH45>3.0.CO;2-F. [DOI] [PubMed] [Google Scholar]
- 67.Winter JM, Ting AH, Vilardell F, et al. Absence of E-cadherin expression distinguishes noncohesive from cohesive pancreatic cancer. Clin Cancer Res. 2008;14(2):412–418. doi: 10.1158/1078-0432.CCR-07-0487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Bergmann F, Moldenhauer G, Herpel E, et al. Expression of L1CAM, COX-2, EGFR, c-KIT and Her2/neu in anaplastic pancreatic cancer: putative therapeutic targets? Histopathology. 2010;56(4):440–448. doi: 10.1111/j.1365-2559.2010.03499.x. [DOI] [PubMed] [Google Scholar]
- 69.Koorstra JB, Maitra A, Morsink FH, et al. Undifferentiated carcinoma with osteoclastic giant cells (UCOCGC) of the pancreas associated with the familial atypical multiple mole melanoma syndrome (FAMMM) Am J Surg Pathol. 2008;32(12):1905–1909. doi: 10.1097/PAS.0b013e31818371cd. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Kopreski MS, Benko FA, Kwee C, et al. Detection of mutant K-ras DNA in plasma or serum of patients with colorectal cancer. Br J Cancer. 1997;76(10):1293–1299. doi: 10.1038/bjc.1997.551. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Molberg KH, Heffess C, Delgado R, Albores-Saavedra J. Undifferentiated carcinoma with osteoclast-like giant cells of the pancreas and periampullary region. Cancer. 1998;82(7):1279–1287. doi: 10.1002/(sici)1097-0142(19980401)82:7<1279::aid-cncr10>3.0.co;2-3. [DOI] [PubMed] [Google Scholar]
- 72.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(10):1223–1229. doi: 10.1053/hupa.2000.18491. [DOI] [PubMed] [Google Scholar]
- 73.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(10):1247–1254. doi: 10.1097/00000478-199810000-00010. [DOI] [PubMed] [Google Scholar]
- 74.Shi C, Hruban RH, Klein AP. Familial pancreatic cancer. Arch Pathol Lab Med. 2009;133(3):365–374. doi: 10.5858/133.3.365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Amundadottir LT, Thorvaldsson S, Gudbjartsson DF, et al. Cancer as a complex phenotype: pattern of cancer distribution within and beyond the nuclear family. PLoS Med. 2004;1(3):e65. doi: 10.1371/journal.pmed.0010065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Klein AP, Brune KA, Petersen GM, et al. Prospective risk of pancreatic cancer in familial pancreatic cancer kindreds. Cancer Res. 2004;64(7):2634–2638. doi: 10.1158/0008-5472.can-03-3823. [DOI] [PubMed] [Google Scholar]
- 77.Hruban RH, Wilentz RE, Goggins M, Offerhaus GJ, Yeo CJ, Kern SE. Pathology of incipient pancreatic cancer. Ann Oncol. 1999;10(suppl 4):9–11. [PubMed] [Google Scholar]
- 78.Hahn SA, Greenhalf B, Ellis I, et al. BRCA2 germline mutations in familial pancreatic carcinoma. J Natl Cancer Inst. 2003;95(3):214–221. doi: 10.1093/jnci/95.3.214. [DOI] [PubMed] [Google Scholar]
- 79.van Asperen CJ, Brohet RM, Meijers-Heijboer EJ, et al. Cancer risks in BRCA2 families: estimates for sites other than breast and ovary. J Med Genet. 2005;42(9):711–719. doi: 10.1136/jmg.2004.028829. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Gallmeier E, Kern SE. Targeting Fanconi anemia/BRCA2 pathway defects in cancer: the significance of preclinical pharmacogenomic models. Clin Cancer Res. 2007;13(1):4–10. doi: 10.1158/1078-0432.CCR-06-1637. [DOI] [PubMed] [Google Scholar]
- 81.Bryant HE, Schultz N, Thomas HD, et al. Specific killing of BRCA2-deficient tumours with inhibitors of poly(ADP-ribose) polymerase. Nature. 2005;434(7035):913–917. doi: 10.1038/nature03443. [DOI] [PubMed] [Google Scholar]
- 82.McCabe N, Lord CJ, Tutt AN, Martin NM, Smith GC, Ashworth A. BRCA2-deficient CAPAN-1 cells are extremely sensitive to the inhibition of Poly (ADP-Ribose) polymerase: an issue of potency. Cancer Biol Ther. 2005;4(9):934–936. doi: 10.4161/cbt.4.9.2141. [DOI] [PubMed] [Google Scholar]
- 83.van der Heijden MS, Brody JR, Dezentje DA, et al. In vivo therapeutic responses contingent on Fanconi anemia/BRCA2 status of the tumor. Clin Cancer Res. 2005;11(20):7508–7515. doi: 10.1158/1078-0432.CCR-05-1048. [DOI] [PubMed] [Google Scholar]
- 84.Zbuk KM, Eng C. Hamartomatous polyposis syndromes. Nat Clin Pract Gastroenterol Hepatol. 2007;4(9):492–502. doi: 10.1038/ncpgasthep0902. [DOI] [PubMed] [Google Scholar]
- 85.Giardiello FM, Brensinger JD, Tersmette AC, et al. Very high risk of cancer in familial Peutz-Jeghers syndrome. Gastroenterology. 2000;119(6):1447–1453. doi: 10.1053/gast.2000.20228. [DOI] [PubMed] [Google Scholar]
- 86.Gruis NA, Sandkuijl LA, van der Velden PA, Bergman W, Frants RR. CDKN2 explains part of the clinical phenotype in Dutch familial atypical multiple-mole melanoma (FAMMM) syndrome families. Melanoma Res. 1995;5(3):169–177. doi: 10.1097/00008390-199506000-00005. [DOI] [PubMed] [Google Scholar]
- 87.Lynch HT, Fusaro RM. Pancreatic cancer and the familial atypical multiple mole melanoma (FAMMM) syndrome. Pancreas. 1991;6(2):127–131. doi: 10.1097/00006676-199103000-00001. [DOI] [PubMed] [Google Scholar]
- 88.de Snoo FA, Bishop DT, Bergman W, et al. Increased risk of cancer other than melanoma in CDKN2A founder mutation (p16-Leiden)-positive melanoma families. Clin Cancer Res. 2008;14(21):7151–7157. doi: 10.1158/1078-0432.CCR-08-0403. [DOI] [PubMed] [Google Scholar]
- 89.de las Heras-Castano G, Castro-Senosiain B, Fontalba A, Lopez-Hoyos M, Sanchez-Juan P. Hereditary pancreatitis: clinical features and inheritance characteristics of the R122C mutation in the cationic trypsinogen gene (PRSS1) in six Spanish families. JOP. 2009;10(3):249–255. [PubMed] [Google Scholar]
- 90.Lowenfels AB, Maisonneuve P, DiMagno EP, et al. Hereditary pancreatitis and the risk of pancreatic cancer: International Hereditary Pancreatitis Study Group. J Natl Cancer Inst. 1997;89(6):442–446. doi: 10.1093/jnci/89.6.442. [DOI] [PubMed] [Google Scholar]
- 91.Schneider A, Suman A, Rossi L, et al. SPINK1/PSTI mutations are associated with tropical pancreatitis and type II diabetes mellitus in Bangladesh. Gastroenterology. 2002;123(4):1026–1030. doi: 10.1053/gast.2002.36059. [DOI] [PubMed] [Google Scholar]
- 92.Whitcomb DC, Gorry MC, Preston RA, et al. Hereditary pancreatitis is caused by a mutation in the cationic trypsinogen gene. Nat Genet. 1996;14(2):141–145. doi: 10.1038/ng1096-141. [DOI] [PubMed] [Google Scholar]
- 93.Rustgi AK. The genetics of hereditary colon cancer. Genes Dev. 2007;21(20):2525–2538. doi: 10.1101/gad.1593107. [DOI] [PubMed] [Google Scholar]
- 94.Groden J, Thliveris A, Samowitz W, et al. Identification and characterization of the familial adenomatous polyposis coli gene. Cell. 1991;66(3):589–600. doi: 10.1016/0092-8674(81)90021-0. [DOI] [PubMed] [Google Scholar]
- 95.Kinzler KW, Nilbert MC, Su LK, et al. Identification of FAP locus genes from chromosome 5q21. Science. 1991;253(5020):661–665. doi: 10.1126/science.1651562. [DOI] [PubMed] [Google Scholar]
- 96.Giardiello FM, Offerhaus GJ, Lee DH, et al. Increased risk of thyroid and pancreatic carcinoma in familial adenomatous polyposis. Gut. 1993;34(10):1394–1396. doi: 10.1136/gut.34.10.1394. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Jones PA, Baylin SB. The epigenomics of cancer. Cell. 2007;128(4):683–692. doi: 10.1016/j.cell.2007.01.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Bestor T, Laudano A, Mattaliano R, Ingram V. Cloning and sequencing of a cDNA encoding DNA methyltransferase of mouse cells: the carboxyl-terminal domain of the mammalian enzymes is related to bacterial restriction methyl-transferases. J Mol Biol. 1988;203(4):971–983. doi: 10.1016/0022-2836(88)90122-2. [DOI] [PubMed] [Google Scholar]
- 99.Okano M, Xie S, Li E. Cloning and characterization of a family of novel mammalian DNA (cytosine-5) methyltransferases. Nat Genet. 1998;19(3):219–220. doi: 10.1038/890. [DOI] [PubMed] [Google Scholar]
- 100.Yen RW, Vertino PM, Nelkin BD, et al. Isolation and characterization of the cDNA encoding human DNA methyltransferase. Nucleic Acids Res. 1992;20(9):2287–2291. doi: 10.1093/nar/20.9.2287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Li A, Omura N, Hong SM, Goggins M. Pancreatic cancer DNMT1 expression and sensitivity to DNMT1 inhibitors. Cancer Biol Ther. 2010;9(4) doi: 10.4161/cbt.9.4.10750. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Bird AP. CpG-rich islands and the function of DNA methylation. Nature. 1986;321(6067):209–213. doi: 10.1038/321209a0. [DOI] [PubMed] [Google Scholar]
- 103.Antequera F, Bird A. Number of CpG islands and genes in human and mouse. Proc Natl Acad Sci U S A. 1993;90(24):11995–11999. doi: 10.1073/pnas.90.24.11995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Shen L, Kondo Y, Guo Y, et al. Genome-wide profiling of DNA methylation reveals a class of normally methylated CpG island promoters. PLoS Genet. 2007;3(10):2023–2036. doi: 10.1371/journal.pgen.0030181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Matsubayashi H, Sato N, Fukushima N, et al. Methylation of cyclin D2 is observed frequently in pancreatic cancer but is also an age-related phenomenon in gastrointestinal tissues. Clin Cancer Res. 2003;9(4):1446–1452. [PubMed] [Google Scholar]
- 106.Matsubayashi H, Sato N, Brune K, et al. Age- and disease-related methylation of multiple genes in nonneoplastic duodenum and in duodenal juice. Clin Cancer Res. 2005;11(2 pt 1):573–583. [PubMed] [Google Scholar]
- 107.Jones PA, Baylin SB. The fundamental role of epigenetic events in cancer. Nat Rev Genet. 2002;3(6):415–428. doi: 10.1038/nrg816. [DOI] [PubMed] [Google Scholar]
- 108.Nakata B, Wang YQ, Yashiro M, et al. Prognostic value of microsatellite instability in resectable pancreatic cancer. Clin Cancer Res. 2002;8(8):2536–2540. [PubMed] [Google Scholar]
- 109.Yamamoto H, Itoh F, Nakamura H, et al. Genetic and clinical features of human pancreatic ductal adenocarcinomas with widespread microsatellite instability. Cancer Res. 2001;61(7):3139–3144. [PubMed] [Google Scholar]
- 110.Sato N, Fukushima N, Maehara N, et al. SPARC/osteonectin is a frequent target for aberrant methylation in pancreatic adenocarcinoma and a mediator of tumor-stromal interactions. Oncogene. 2003;22(32):5021–5030. doi: 10.1038/sj.onc.1206807. [DOI] [PubMed] [Google Scholar]
- 111.Infante JR, Matsubayashi H, Sato N, et al. Peritumoral fibroblast SPARC expression and patient outcome with resectable pancreatic adenocarcinoma. J Clin Oncol. 2007;25(3):319–325. doi: 10.1200/JCO.2006.07.8824. [DOI] [PubMed] [Google Scholar]
- 112.Sato N, Fukushima N, Chang R, Matsubayashi H, Goggins M. Differential and epigenetic gene expression profiling identifies frequent disruption of the RELN pathway in pancreatic cancers. Gastroenterology. 2006;130(2):548–565. doi: 10.1053/j.gastro.2005.11.008. [DOI] [PubMed] [Google Scholar]
- 113.Sato N, Parker AR, Fukushima N, et al. Epigenetic inactivation of TFPI-2 as a common mechanism associated with growth and invasion of pancreatic ductal adenocarcinoma. Oncogene. 2005;24(5):850–858. doi: 10.1038/sj.onc.1208050. [DOI] [PubMed] [Google Scholar]
- 114.Wada M, Yazumi S, Takaishi S, et al. Frequent loss of RUNX3 gene expression in human bile duct and pancreatic cancer cell lines. Oncogene. 2004;23(13):2401–2407. doi: 10.1038/sj.onc.1207395. [DOI] [PubMed] [Google Scholar]
- 115.Fukushima N, Sato N, Sahin F, Su GH, Hruban RH, Goggins M. Aberrant methylation of suppressor of cytokine signalling-1 (SOCS-1) gene in pancreatic ductal neoplasms. Br J Cancer. 2003;89(2):338–343. doi: 10.1038/sj.bjc.6601039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Jansen M, Fukushima N, Rosty C, et al. Aberrant methylation of the 5′ CpG island of TSLC1 is common in pancreatic ductal adenocarcinoma and is first manifest in high-grade PanlNs. Cancer Biol Ther. 2002;1(3):293–296. doi: 10.4161/cbt.84. [DOI] [PubMed] [Google Scholar]
- 117.Fukushima N, Sato N, Ueki T, et al. Aberrant methylation of preproenkephalin and p16 genes in pancreatic intraepithelial neoplasia and pancreatic ductal adenocarcinoma. Am J Pathol. 2002;160(5):1573–1581. doi: 10.1016/S0002-9440(10)61104-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Sato N, Fukushima N, Matsubayashi H, Iacobuzio-Donahue CA, Yeo CJ, Goggins M. Aberrant methylation of Reprimo correlates with genetic instability and predicts poor prognosis in pancreatic ductal adenocarcinoma. Cancer. 2006;107(2):251–257. doi: 10.1002/cncr.21977. [DOI] [PubMed] [Google Scholar]
- 119.Sato N, Matsubayashi H, Abe T, Fukushima N, Goggins M. Epigenetic down-regulation of CDKN1C/p57KIP2 in pancreatic ductal neoplasms identified by gene expression profiling. Clin Cancer Res. 2005;11(13):4681–4688. doi: 10.1158/1078-0432.CCR-04-2471. [DOI] [PubMed] [Google Scholar]
- 120.Lee MH, Reynisdottir I, Massague J. Cloning of p57KIP2, a cyclin-dependent kinase inhibitor with unique domain structure and tissue distribution. Genes Dev. 1995;9(6):639–649. doi: 10.1101/gad.9.6.639. [DOI] [PubMed] [Google Scholar]
- 121.Matsuoka S, Edwards MC, Bai C, et al. p57KIP2, a structurally distinct member of the p21CIP1 Cdk inhibitor family, is a candidate tumor suppressor gene. Genes Dev. 1995;9(6):650–662. doi: 10.1101/gad.9.6.650. [DOI] [PubMed] [Google Scholar]
- 122.Sato N, Ueki T, Fukushima N, et al. Aberrant methylation of CpG islands in intraductal papillary mucinous neoplasms of the pancreas. Gastroenterology. 2002;123(1):365–372. doi: 10.1053/gast.2002.34160. [DOI] [PubMed] [Google Scholar]
- 123.Hong SM, Kelly D, Griffith M, et al. Multiple genes are hypermethylated in intraductal papillary mucinous neoplasms of the pancreas. Mod Pathol. 2008;21(12):1499–1507. doi: 10.1038/modpathol.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Sato N, Fukushima N, Hruban RH, Goggins M. CpG island methylation profile of pancreatic intraepithelial neoplasia. Mod Pathol. 2008;21(3):238–244. doi: 10.1038/modpathol.3800991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Feinberg AP, Vogelstein B. Hypomethylation distinguishes genes of some human cancers from their normal counterparts. Nature. 1983;301(5895):89–92. doi: 10.1038/301089a0. [DOI] [PubMed] [Google Scholar]
- 126.Gama-Sosa MA, Slagel VA, Trewyn RW, et al. The 5-methylcytosine content of DNA from human tumors. Nucleic Acids Res. 1983;11(19):6883–6894. doi: 10.1093/nar/11.19.6883. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.van der Put NM, Gabreels F, Stevens EM, et al. A second common mutation in the methylenetetrahydrofolate reductase gene: an additional risk factor for neural-tube defects? Am J Hum Genet. 1998;62(5):1044–1051. doi: 10.1086/301825. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Blount BC, Mack MM, Wehr CM, et al. Folate deficiency causes uracil misincorporation into human DNA and chromosome breakage: implications for cancer and neuronal damage. Proc Natl Acad Sci U S A. 1997;94(7):3290–3295. doi: 10.1073/pnas.94.7.3290. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Pogribny IP, Basnakian AG, Miller BJ, Lopatina NG, Poirier LA, James SJ. Breaks in genomic DNA and within the p53 gene are associated with hypomethylation in livers of folate/methyl-deficient rats. Cancer Res. 1995;55(9):1894–1901. [PubMed] [Google Scholar]
- 130.Matsubayashi H, Skinner HG, Iacobuzio-Donahue C, et al. Pancreaticobiliary cancers with deficient methylenetetrahydrofolate reductase genotypes. Clin Gastroenterol Hepatol. 2005;3(8):752–760. doi: 10.1016/s1542-3565(05)00359-9. [DOI] [PubMed] [Google Scholar]
- 131.Rosty C, Ueki T, Argani P, et al. Overexpression of S100A4 in pancreatic ductal adenocarcinomas is associated with poor differentiation and DNA hypomethylation. Am J Pathol. 2002;160(1):45–50. doi: 10.1016/S0002-9440(10)64347-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Sato N, Maitra A, Fukushima N, et al. Frequent hypomethylation of multiple genes overexpressed in pancreatic ductal adenocarcinoma. Cancer Res. 2003;63(14):4158–4166. [PubMed] [Google Scholar]
- 133.Hwang HW, Mendell JT. MicroRNAs in cell proliferation, cell death, and tumorigenesis. Br J Cancer. 2006;94(6):776–780. doi: 10.1038/sj.bjc.6603023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Zhang B, Pan X, Cobb GP, Anderson TA. microRNAs as oncogenes and tumor suppressors. Dev Biol. 2007;302(1):1–12. doi: 10.1016/j.ydbio.2006.08.028. [DOI] [PubMed] [Google Scholar]
- 135.Krutzfeldt J, Rajewsky N, Braich R, et al. Silencing of microRNAs in vivo with ‘antagomirs’. Nature. 2005;438(7068):685–689. doi: 10.1038/nature04303. [DOI] [PubMed] [Google Scholar]
- 136.Calin GA, Dumitru CD, Shimizu M, et al. Frequent deletions and down-regulation of micro-RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia. Proc Natl Acad Sci U S A. 2002;99(24):15524–15529. doi: 10.1073/pnas.242606799. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Hayashita Y, Osada H, Tatematsu Y, et al. A polycistronic microRNA cluster, miR-17-92, is overexpressed in human lung cancers and enhances cell proliferation. Cancer Res. 2005;65(21):9628–9632. doi: 10.1158/0008-5472.CAN-05-2352. [DOI] [PubMed] [Google Scholar]
- 138.Rinaldi A, Poretti G, Kwee I, et al. Concomitant MYC and microRNA cluster miR-17-92 (C13orf25) amplification in human mantle cell lymphoma. Leuk Lymphoma. 2007;48(2):410–412. doi: 10.1080/10428190601059738. [DOI] [PubMed] [Google Scholar]
- 139.Brueckner B, Stresemann C, Kuner R, et al. The human let-7a-3 locus contains an epigenetically regulated microRNA gene with oncogenic function. Cancer Res. 2007;67(4):1419–1423. doi: 10.1158/0008-5472.CAN-06-4074. [DOI] [PubMed] [Google Scholar]
- 140.Saito Y, Liang G, Egger G, et al. Specific activation of microRNA-127 with downregulation of the proto-oncogene BCL6 by chromatin-modifying drugs in human cancer cells. Cancer Cell. 2006;9(6):435–443. doi: 10.1016/j.ccr.2006.04.020. [DOI] [PubMed] [Google Scholar]
- 141.Han L, Witmer PD, Casey E, Valle D, Sukumar S. DNA methylation regulates MicroRNA expression. Cancer Biol Ther. 2007;6(8):1284–1288. doi: 10.4161/cbt.6.8.4486. [DOI] [PubMed] [Google Scholar]
- 142.Chang TC, Yu D, Lee YS, et al. Widespread microRNA repression by Myc contributes to tumorigenesis. Nat Genet. 2008;40(1):43–50. doi: 10.1038/ng.2007.30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.O’Donnell KA, Wentzel EA, Zeller KI, Dang CV, Mendell JT. c-Myc-regulated microRNAs modulate E2F1 expression. Nature. 2005;435(7043):839–843. doi: 10.1038/nature03677. [DOI] [PubMed] [Google Scholar]
- 144.Bloomston M, Frankel WL, Petrocca F, et al. MicroRNA expression patterns to differentiate pancreatic adenocarcinoma from normal pancreas and chronic pancreatitis. JAMA. 2007;297(17):1901–1908. doi: 10.1001/jama.297.17.1901. [DOI] [PubMed] [Google Scholar]
- 145.Dillhoff M, Liu J, Frankel W, Croce C, Bloomston M. MicroRNA-21 is overexpressed in pancreatic cancer and a potential predictor of survival. J Gastrointest Surg. 2008;12(12):2171–2176. doi: 10.1007/s11605-008-0584-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Lee EJ, Gusev Y, Jiang J, et al. Expression profiling identifies microRNA signature in pancreatic cancer. Int J Cancer. 2007;120(5):1046–1054. doi: 10.1002/ijc.22394. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.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(30):4442–4452. doi: 10.1038/sj.onc.1210228. [DOI] [PubMed] [Google Scholar]
- 148.Volinia S, Calin GA, Liu CG, et al. A microRNA expression signature of human solid tumors defines cancer gene targets. Proc Natl Acad Sci U S A. 2006;103(7):2257–2261. doi: 10.1073/pnas.0510565103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Zhang Y, Li M, Wang H, et al. Profiling of 95 microRNAs in pancreatic cancer cell lines and surgical specimens by real-time PCR analysis. World J Surg. 2009;33(4):698–709. doi: 10.1007/s00268-008-9833-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Chang TC, Wentzel EA, Kent OA, et al. Transactivation of miR-34a by p53 broadly influences gene expression and promotes apoptosis. Mol Cell. 2007;26(5):745–752. doi: 10.1016/j.molcel.2007.05.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Kent OA, Mullendore M, Wentzel EA, et al. A resource for analysis of microRNA expression and function in pancreatic ductal adenocarcinoma cells. Cancer Biol Ther. 2009;8(21):2013–2024. doi: 10.4161/cbt.8.21.9685. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Li A, Omura N, Hong SM, et al. Pancreatic cancers epigenetically silence SIP1 and hypomethylate and overexpress miR-200a/200b in association with elevated circulating miR-200a and miR-200b levels. Cancer Res. 2010;70(13):5226–5237. doi: 10.1158/0008-5472.CAN-09-4227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Ryu JK, Hong SM, Karikari CA, Hruban RH, Goggins MG, Maitra A. Aberrant MicroRNA-155 expression is an early event in the multistep progression of pancreatic adenocarcinoma. Pancreatology. 2010;10(1):66–73. doi: 10.1159/000231984. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Habbe N, Koorstra JB, Mendell JT, et al. MicroRNA miR-155 is a biomarker of early pancreatic neoplasia. Cancer Biol Ther. 2009;8(4):340–346. doi: 10.4161/cbt.8.4.7338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Goggins M, Schutte M, Lu J, et al. Germline BRCA2 gene mutations in patients with apparently sporadic pancreatic carcinomas. Cancer Res. 1996;56(23):5360–5364. [PubMed] [Google Scholar]
- 156.Okami J, Simeone DM, Logsdon CD. Silencing of the hypoxia-inducible cell death protein BNIP3 in pancreatic cancer. Cancer Res. 2004;64(15):5338–5346. doi: 10.1158/0008-5472.CAN-04-0089. [DOI] [PubMed] [Google Scholar]
- 157.Fitzgerald M, Oshiro M, Holtan N, et al. Human pancreatic carcinoma cells activate maspin expression through loss of epigenetic control. Neoplasia. 2003;5(5):427–436. doi: 10.1016/s1476-5586(03)80045-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Ohike N, Maass N, Mundhenke C, et al. Clinicopathological significance and molecular regulation of maspin expression in ductal adenocarcinoma of the pancreas. Cancer Lett. 2003;199(2):193–200. doi: 10.1016/s0304-3835(03)00390-2. [DOI] [PubMed] [Google Scholar]
- 159.Iacobuzio-Donahue CA, Maitra A, Olsen M, et al. Exploration of global gene expression patterns in pancreatic adenocarcinoma using cDNA micro-arrays. Am J Pathol. 2003;162(4):1151–1162. doi: 10.1016/S0002-9440(10)63911-9. [DOI] [PMC free article] [PubMed] [Google Scholar]