Abstract
Pancreatic cancer (PC) is a leading cause of cancer related deaths in United States. The lack of early symptoms results in late-stage detection and a high mortality rate. Currently, the only potentially curative approach for PC is surgical resection, which is often unsuccessful because the invasive and metastatic nature of the tumor masses makes their complete removal difficult. Consequently, patients suffer relapses from remaining cancer stem cells or drug resistance that eventually lead to death. To improve the survival rate, the early detection of PC is critical.
Current biomarker research in PC indicates that a serum carbohydrate antigen, CA 19-9, is the only available biomarker with approximately 90% specificity to PC. However, the efficacy of CA 19-9 for assessing prognosis and monitoring patients with PC remains contentious. Thus, advances in technology and the detection of new biomarkers with high specificity to PC are needed to reduce the mortality rate of pancreatic cancer.
Keywords: Pancreatic cancer, biomarker, tumor progression, risk factor, CA 19-9
INTRODUCTION
With a mortality rate of nearly 100 %, pancreatic cancer (PC) is one of the most lethal malignancies [1] and it is the fourth most prominent cause for cancer-related deaths in both men and women in the United States [2–3]. Even patients treated with current state-of-the-art therapies have an overall 5-year survival rate of only 3% [4]. The main reason is that approximately 80% of newly diagnosed patients are already in the metastatic stage of the disease, for which no curative therapy is currently available [5–7].
Most patients with PC have no distinct clinical manifestations in their early stages of the disease. Commonly described symptoms include pain in the upper abdominal region, significant weight loss, heartburn, altered bowel habits, and obstructive jaundice [8]. Obstructive jaundice is the only symptom with some specificity for PC [9–12]. The diagnosis of PC relies on biopsy and computed tomography (CT) and/or magnetic resonance imaging (MRI) [13–14]. The diagnosis could be improved by using endoscopic ultrasound (EUS) and positron emission tomography (PET) techniques [15–17]. Advanced technologies such as magnetic resonance cholangiopancreatography (MRCP) and endoscopic retrograde cholangiopancreatography (ERCP) facilitate better detection of tumors at an early stage by allowing a comprehensive and introspective analysis of the morphological changes of the gland [18–20]. Other techniques like fine-needle aspiration (FNA) are useful for pinpointing and retrieving biopsied specimens [21–22]. However, these techniques are not only costly but also require access to highly advanced, specialized facilities (Table 1), and therefore may not be readily available for detecting the disease at an early stage.
Table 1.
| Diagnostic Tool | Applications and Advantages | Limitations |
|---|---|---|
| Transabdominal ultrasound | Visualization of bile duct and liver | Poor visualization of pancreas |
| Computed tomography (CT) | Diagnosis and staging | Nephrotoxicity (Iodine-contrast agent); potential side effects from exposure to radiation |
| Magnetic resonance imaging (MRI) | Superior imaging of pancreas and bile duct Iodine-free contrast agent No radiation |
Expensive Less availability |
| Positron emission tomography (PET) | Metastatic disease assessment, Monitoring recurrence and response to adjuvant therapy | Expensive Less availability |
| Endoscopic ultrasound (EUS) | Imaging of pancreatic masses and lymph nodes | Expensive Less availability |
| EUS-FNA (Fine needle) | Specific detection of PC Safe and less invasive technology Imaging of lesions from 3–4 mm upwards Puncturing of lymph nodes greater than 5 mm |
Access only to posterior mediastinum |
| Magnetic resonance cholangiopancreatography (MRCP) | Imaging of fluid in the pancreatic ducts in a non-invasive manner | Lower resolution No biopsy Expensive |
| Endoscopic retrograde cholangiopancreatography (ERCP) | Imaging strictures | Complications like bleeding and infection |
| Cholangiopancreatoscopy | Site-directed biopsies of strictures | Fragile equipment |
At present, no curative therapies are available for patients with PC; the mainstay of treatment with the potential for cure is the complete, eradicative surgical resection of the primary carcinoma. Due to the lack of specific early-stage clinical symptoms and the highly deleterious, aggressive nature of the evolving disease, patients are usually diagnosed at the unresectable stages [23–24]. Consequently, the average survival of the resected patients is approximately 12 to 20 months with a high probability of relapse [25–26]. Use of gemcitabine (Gemzar®) or the combinations of gemcitabine with other cancer drugs, such as oxalipatin, cisplatin or erlotinib has demonstrated efficacy in the treatment of PC [27–30]. The role of radiotherapy in PC treatment remains controversial [31], although radiochemotherapy is increasingly accepted as a treatment modality for unresectable, nonmetastatic PC [32–33]. Despite these advances, PC remains one of the most deadly malignancies.
TUMOR ORIGIN AND PROGRESSION
Although the exocrine pancreas consists of a very small ductal system, 90% of pancreatic neoplasms (of which 80% are invasive adenocarcinomas) are ductal in origin, and the remaining 10% are islet-cell and cystic tumors [1]. The etiology of PC remains unclear, but several risk factors have been identified (Table 2). In general, individuals diagnosed with diabetes or with a history of pancreatitis or chronic pancreatitis are at a risk for developing PC [1, 34–35]. A history of smoking and/or alcoholism also increases the risk of PC [36–38]. Recently, Peutz-Jeghers syndrome, single nucleotide polymorphisms (SNPs), hereditary pancreatitis, and a 5–10% family history of PC have also been added as PC risk factors [16, 35, 39].
Table 2.
High Risk Factors for PC
| Risk factors | References |
|---|---|
| Obesity (Body Mass Index) ≥ 30 | [188–190] |
| Having symptoms indicative of PC for ≥ 3 years | [1] |
| Smokers & smokers specifically with polymorphism at Arg188His | [34–35, 38, 191–192] |
| Obstructive jaundice | [1, 159] |
| Onset of type II diabetes for ≥ 3 years | [1, 193–194] |
| Chronic pancreatitis & pancreatitis history | [195–196] |
| Peutz-Jeghers syndrome | [197] |
| Hypermethylation of p14 and p16 + K-ras mutation | [198] |
| ABO blood group | [199–201] |
More than 90% of PC patients acquire the disease sporadically; it is unclear how the normal tissue is transformed to PC. Fig. (1) illustrates a Genetic Progression Model based on the oncogenic markers in tumor tissues [1, 40–41]. In the model, normal pancreatic cells are transformed to pre-malignant lesions termed pancreatic intraepithelial neoplasia (PanINs) in concomitance with a multitude of genetic changes and morphological dysplasia, named PanIN-1a, PanIN-1b, PanIN-2, and PanIN-3 that collectively may be regarded as molecular and structural antecedents to the eventual establishment of invasive adenocarcinoma [10,11]. Some of the most frequently detected genetic changes in the initial stages of PC include the activation of mutated K-ras [42–44], the hypermethylation of preproenkephalin (ppENK) [45–46], the over-expression of EGF-family ligands and receptors such as ERBB2 and EGFR [47–49], the robust increase in the level of carcinoembryonic antigen-related cell adhesion molecule 1 (biliary glycoprotein) (CEACAM1) [50], and the hyperphosphorylation of Retinoblastoma protein (Rb) [51]. Most notably, some researchers hypothesize that the activation of K-ras mutation signaling, leading to the overexpression of EGF-family ligands and receptors such as ERBB2 and EGFR, plays a pivotal role in the initiation of PC [47–49]. During disease progression, the loss of tumor suppressor genes (e.g. p16, p53, SMAD4/DPC4, BRCA2) has been identified at later stages of PanIN [52–58]. The over-expression of a negative regulator of p53 called MDM2 occurs in coordination with the malignant proliferation of PC [59–60]. These molecular changes and events as a whole promulgate the hyperphosphorylation of Rb protein, which promotes cell cycle phase transition and leads to uncontrolled cell division [51]. Along with tumor progression, the expression of CXCR4, a chemokine receptor, and miR-155 also increases in PC especially in the PanIN-3 lesions [41, 61–62]. Further, the level of macrophage stimulating 1 receptor (MST1R), also termed RON, is increasingly elevated in premalignant PanIN stages [63]. Additionally progressive methylation in the promoter regions of LHX1, a transcriptional regulator and secreted apoptosis related protein (SARP2), has also been reported in PanIN lesions [64–66]. The aggressive stage of the tumor is also characterized by the appearance of abnormally shortened telomere [67] and the abnormal fusion of chromosome ends, concurrent with neoplastic progression of pancreatic cells [68].
Fig. (1).
ADVANCES IN PC BIOMARKER RESEARCH
Improving the PC mortality rate necessitates the discovery of new tools for the early detection, diagnosis, and monitoring of therapeutic efficacy. To provide a context, we provide an overview of technological developments relevant to biomarker research and biomarker identification in the early stages of PC.
TECHNOLOGIES FOR BIOMARKER DISCOVERY AND IDENTIFICATION
Important technological innovations have been made in the past several decades to facilitate biomarker identification. Microarray and quantitative RT-PCR or PCR are frequently used to analyze the variations of RNA/microRNA or DNA in the tumor tissues and other types of specimens from patients and healthy individuals [69–71]. Methylation-specific PCR (MSP) and bisulfite-sequencing PCR (BSP) [72] have been used to analyze DNA from different samples [73]. Gene expression profiling and tissue microarray (TMA) have been used to identify novel cell-surface targets [36]. New equipment and techniques have been developed to systematically search for protein markers in specimens, including differential in-gel electrophoresis (DIGE), tandem mass spectrometry (MS/MS), electron transfer dissociation mass spectroscopy [74], surface-enhanced laser desorption/ionization (SELDI) [75–76], reverse-phase protein array [74], enzyme linked immuno sorbent assay (ELISA), matrix assisted laser desorption/ionization time-of-flight mass spectroscopy (MALDI-TOF-MS), and quantitative proteomics [77–78]. Advances in these technologies have provided significant leads in search of potential biomarkers for PC (Table 3).
Table 3.
| Technology | Applications and Advantages | Limitations | |
|---|---|---|---|
| Microarray | Chip-based concurrent analysis of thousands of genes | Frequent identification of false positives | |
| RT-PCR/PCR/ MSP | Multiple target analysis in a single, multiplexed reaction | Sensitive, can generate false positive results | |
| PROTEOMICS | Two-dimensional gel electrophoresis (2DE) | Separation of proteins by isoelectric point (IP) and molecular weight (MW) | Permits detection of very basic or acidic proteins only |
| Differential in-gel electrophoresis (DIGE) | Co-separation of multiple samples and visualization on one 2-D gel | Changes in labeling can markedly alter position of migration of specific proteins | |
| Multi-dimensional protein identification technology (MudPIT) | Non-gel approach for the identification of proteins from complex mixtures | Inability to directly measure the relative abundance of proteins | |
| Isotope coded affinity tag (ICAT) | Measurement of relative abundance of proteins labeled at cysteine residues | Suitable only for proteins with labeling-accessible cysteines | |
| Isobaric tags for relative and absolute quantification (iTRAQ) | Identification and quantification of proteins by labeling both whole and subcellular proteomes | Unlikely to reveal basic information on proteins of interest | |
| Matrix-assisted laser desorption/ionization (MALDI) | Identification of proteins isolated through gel electrophoresis | Quantitation difficult | |
| Surface-enhanced laser desorption/ionization (SELDI) | Analysis of protein mixtures by mass spectroscopy ionization method | Low reproducibility Low resolution |
|
SOURCE OF BIOSPECIMENS IN BIOMARKER ANALYSIS
Sample source is important in the search for biomarkers to detect and diagnose early PC. Pancreatic biopsy samples are reliable sources for diagnosis, and pancreatic fluid (known as juice) from the pancreatic duct has recently been identified as an alternative source for biomarker discovery [72, 79] because the juice contains PC-specific markers (DNA/RNA/proteins and cancer cells) that are shed into the ductal lumen [79] (Table 4). Furthermore, plasma/serum DNA/RNA/protein, disseminated tumor cells (DTC) and circulating tumor cells (CTC) in the blood are also frequently used for biomarker detection. In addition, saliva has also been exploited as a source of PC biomarkers and genes have been detected with potential for classification of resectable PC [80]. A recent study has suggested that detection of genetic markers such as K-ras and p53 in stool samples might enable the detection of early PC [81]. Potential biomarkers for PC using various source materials and discovered thus far are summarized in Table 4.
Table 4.
Various Tissue Types Used for PC Biomarker Research
| Sample Source | Detection Method | Markers | References |
|---|---|---|---|
| Tissue | Immunohistochemistry, PCR-based techniques, and Microarray | K-ras, TP53, SMAD4, p16, Plectin-1, Caveolin-1, hENT1, miR-216, miR-217, miR-196, miR-196a, miR-186, miR-222, miR-200b, miR-15b, miR-95, FosB, KLF6, ATP4A, NFKBIZ, GSG1, SIGLEC11 | [22, 43, 84–85, 100, 125, 131, 213–220] |
| Pancreatic juice | Quantitative proteomics | AGR2, S100A6, MMP-9, DJ-1, A1BG, MMP-7, miRNA-21, miRNA-155, Twist, PRSS2, PLRP1, hTERT | [72, 166, 221–226] |
| Serum | Quantitative proteomics, ELISA, | CA 19-9, CA 125, CEACAM1, MMP-7, REG4, HSP-27, IGF, IGFR, cyclin I, GDI2, PF4, SAA, fibrinogen γ | [37, 50, 227–236] |
| Plasma | Functional genomics | TNC, TFP1, TGFBI, SEL-1L, LICAM, WWTR1, CDC4BPA, CK 18 | [101, 237] |
| CTC | RT-PCR | C-MET, h-TERT, CK20, ELC, PlGF | [238–239] |
| Saliva | Human genome array | K-ras, MBD3L2, ACRV1, DPM1 | [80] |
| Stool | PCR | K-ras, TP53 | [81, 240] |
BIOMARKERS
With the aid of new technologies and using a variety of sample sources, a wide array of potential biomarkers for PC has been identified (Table 5).
Table 5.
Most Studied Markers in PC
| Biomarkers | Detection Method | References | |
|---|---|---|---|
| Molecular biomarkers | |||
| K-ras | PCR | [22, 43, 87, 241–243] | |
| p16 | PCR and immunohistochemistry | [22, 90, 213, 244–246] | |
| P53 | PCR and immunohistochemistry | [43, 87, 90, 242] | |
| DPC4 | PCR and immunohistochemistry | [93, 247] | |
| Epigenetic biomarkers | |||
| ppENK | Methylation specific PCR | [45, 99, 107, 241] | |
| RASSF1A | Methylation specific PCR | [108–111] | |
| Cyclin D2 | Methylation specific PCR | [99, 112, 248] | |
| MicroRNA biomarkers | |||
| miR-155 | Microarray and qRT-PCR | [41, 218, 224, 249–251] | |
| miR-21 | RT-PCR | [128, 130, 252] | |
| miR-196a | Immunohistochemistry | [126, 131, 216, 249] | |
| Other biomarkers | |||
| CA 19-9 | ELISA | [132, 253–256] | |
| CEACAM1 | ELISA | [50] | |
| AGR2 | RT-PCR, ELISA and Western blotting | [166, 257] | |
| CEA | Immunohistochemistry | [236, 253, 258–260] | |
| CA 125 | Immunoradiometric assay and Immunohistochemistry | [260–261] | |
| CA242 | ELISA | [236, 253, 262–264] | |
GENETIC BIOMARKERS
Based on the tumor progression model, multiple genetic alterations are involved in a step-wise manner in the development of PC and in most cases, genetic changes in the four genes – K-ras, CDKN2A/p16, TP53, and SMAD4/DPC4 were detected in the PC patients [5, 22, 82–86]. Among these genetic changes, the K-ras mutation was most frequently detected with 45%–100% frequency in the tumor tissue and a 77% frequency in the serum of patients with PC or pancreatic adenocarcinoma (PAC) [22, 43, 87–88]. 50–75% patients with PAC have mutations in TP53 [43, 89]. In addition to K-ras and TP53 mutations, the loss of p16 and DPC4 was also frequently detected in tumor tissue of PC patients and correlated with its clinical relevance [84, 90–93]. The K-ras mutation was detected in the premalignant stage, which suggested that the oncogenic gain of mutation in K-ras could be a potential marker used for the early detection of cancer [94–96]. Other genetic alterations described in PC occurring at a much lower frequency include BRCA2, TGFBR1, BAX, RB1, STK11, hMLH1, hCDC4, MKK4, FancC, and AKT2 [94, 97–99]. Another biomarker Plectin-1 that was highly expressed in the PanIN-3 tissues may be used to discriminate the primary from metastatic PAC [100]. Studies using a functional genomics approach identified seven genes from plasma – TNC, TFP1, TGFBI, SEL-IL, LICAM, WWTR1, and CDC4BPA – as belonging to a category that was differentially regulated in PC versus normal samples. Studies also suggested that tissue factor pathway inhibitor (TFPI) may have the potential to be a candidate plasma PC biomarker [101–102]. It is worth noting that although genetic alterations in oncogenes - K-ras, TP53, p16 can be detected in PC [103] and may have diagnostic value in PC, they are not specific to PC and hence not suitable for the early detection of this deadly malignant disease.
EPIGENETIC BIOMARKERS
Epigenetic factors such as aberrations in DNA methylation, histone post-translational modification, and chromatin remodeling could also be considered as tumorigenic biomarkers [104]. Global methylation profiling on 150 CpG sites across 807 genes revealed that 289 CpG sites were differentially methylated between normal and PC, raising the possibility that they have diagnostic and therapeutic values and implications [105]. Genome-wide profiling of methylated promoters in 57 PC and 34 normal pancreatic samples identified excessive methylation on the promoters of several genes, including MDF1, hsa-miR-9-1, ZNF415, CNTAP2 and ELOVL4 in cancerous tissues [106]. Further, frequent epigenetic changes have also been demonstrated in PC in RASSF1A, cyclin D2, ppENK, TFPI2, CDH13 and secreted apoptosis related protein 2 (SARP2) [45, 64, 99, 107–115], suggesting that epigenetic profiling of selected genes in tissue specimens could be used for PC diagnosis and prognosis. A similar approach applied to the analysis of pancreatic juice showed that over 1% in the methylation status of cyclin D2, ppENK, tissue factor pathway inhibitor 2(TFPI2), neuronal pentraxin-2(NPTX2) and forkhead box protein (E1FOXE1) could predict PC with 82% sensitivity and 100% specificity [99]. The hypermethylation of NPTX2 and SARP2 in PC patients was recently confirmed [115–116]; similarly, methylation profiling analysis of circulating plasma DNA in PC patients detected cyclin D2 and a few other markers in PC patients [117]. Other epigenetic modifications potentially useful as biomarkers for poor survival in resected PAC patients include low cellular levels of histone H3 lysine 4 dimethylation (H3K4me2), histone H3 lysine 9 dimethylation (H3K9me2), and histone H3 lysine 18 acetylation (H3K18ac) [118]. Epigenetic silencing of SIP1 and hypermethylation and over-expression of miR-200a/200b in association with elevated circulating miRs have been reported in PC [119].
MIRNA BIOMARKERS
miRNAs are short non-coding RNAs of approximately 22 nucleotides that have been shown to regulate the expression of both oncogenes or tumor suppressor genes, as part of the posttranscriptional gene silencing (PTGS) mechanism [120–124]. Recently, it was found that the levels of certain tissue-specific miR-NAs, such as miR-216 and -217, were markedly down-regulated in tumor tissues compared to matched normal controls [125]. Conversely, the expression of miR-196 and -196a was significantly elevated in tumor tissues derived from PAC patients compared to normal pancreatic specimens [125]. Moreover, miR-196a was detected in the serum of PAC, and thus may have predictive potential regarding the survival of PAC patients [126]. Other miRNA biomarker candidates include elevation in circulating miR-210 for PC [127], miR-21 as an integral link to the precursor lesions of PAC [128–130], and upregulation in an eight-miRNA panel (miR196a, miR-190, miR-186, miR-221, miR-222, miR-200b, miR-15b, and miR-95) based on an RT-PCR analysis involving 5 PC tissues and 3 PC cell lines spanning a total of 95 microRNAs [131].
OTHER BIOMARKERS
Beyond targeting genetic, epigenetic and miRNA markers in PC using primarily tumor specimens, with some less well defined and specific changes detected in the serum and other body fluids, several other PC biomarkers have been identified and are listed in Tables 5 & 6. Among these, perhaps the one considered to have the most definitive diagnostic, prognostic, and predictive value is carbohydrate antigen 19-9 (CA 19-9), which shows an 81% sensitivity and 90% specificity [132–137].
Table 6.
Most Promising Protein Markers for PC in the Serum
| Markers | Screening | Diagno- sis/Prognosis |
healthy Individual |
Patient | Sensitivity | Specificity | References |
|---|---|---|---|---|---|---|---|
| CA 19-9 | No | Yes | < 37 U/ml | > 37 U/ml | 81% | 90% | [1, 134, 158, 170, 265–266] |
| CEA | No | Yes | < 5 U/ml | > 50 U/ml | 45% | 75% | [158, 161, 236, 266] |
| CA 125 | No | Yes | < 35 U/ml | > 35 U/ml | 60% | 80% | [260, 267] |
| CA 242 | No | Yes | <25 U/ml | > 25 U/ml | 60% | 80% | [170, 236, 253, 264, 268–269] |
CA 19-9
Although first described as a tumor-associated carbohydrate antigen in 1979 by Koprowski et al., [138–139], CA 19-9 is now known to be a sialylated Lea blood group antigen [140]. The nomenclature of CA 19-9 was compiled from the mucinous glycoprotein complex (such as H-CanAg, L-CanAg, and MUC1) [133, 141] based on recognition of sialylated lacto-N-fucopentaose II epitope by the moloclonal antibody (1116-NS-19-9). The levels of CA 19-9 in the serum varied according to the patient's secretor status and Lewis genotype [139–140, 142–143]. Individuals who are Lewis blood type negative (Lea-b-) do not express the CA 19-9; hence, for those diagnosed with PC in this group (about 5% of the PC population), elevation in CA19-9 is not observed because of deficiency of a fucosyltransferase essential for the biosynthesis of CA19-9 and the Lewis antigen [144].
CA 19-9 was initially detected in colorectal cancer cell line SW1116 and colorectal tissues [139] but its occurrence was subsequently extended to other organs such as pancreas, stomach, and biliary epithelium [145]. Based on reports showing that the sialyl Lewisa structure (NeuNAcα2-3Ga1β1-3Glc [4-Fucα1] NAcβl-3Galβl-4Glc) acts as a ligand for E-selectin [140, 146–148], Kannagi proposed that three factors should be taken into consideration when predicting the risk of hematogenous metastasis in cancer patients: the degree of sialyl Lewis a/x expression on cancer cells, the degree of E-selectin expression on vessel walls, and the number of CTC in the bloodstream [147]. The concentration of CA 19-9 can be quantitatively determined by CA 19-9 ELISA [101]. Elevated levels (>37 U/ml) of CA 19-9 have been associated with gastrointestinal carcinomas, particularly in PAC [144, 149–151], and may be considered as a well characterized biomarker for PC (reviewed by Duffy et al.) [152], and it is possible to predict the operability of pancreatic tumor in patients. For instance, elevated levels of CA 19-9 (1000 U/ml) are correlated with patients showing a tumor size >5cm in diameter [153]; only 5% of those patients have resectable tumor [134, 153–154]. Other studies have shown that serum CA 19-9 concentrations greater than 300 U/ml are indicative of unresectable PC tumors [155–156].
An increase in CA 19-9 levels may also be used to predict cancer progression while its decrease is suggestive of favorable response to neodjuvant irradiation and chemotherapy [157]. Steinberg compared CA 19-9 levels in 24 studies and found that CA 19-9 showed 81% sensitivity and 90% specificity for PC with a 37 U/ml cut-off [134]. Similar conclusions were reached by Goonnetilleke and Siriwardena [158]. Taken together, therefore, the immuno quantitation of CA 19-9 antigen is regarded to be the gold standard for PC detection [152, 159]. Reservation in using CA 19-9 alone for PC diagnosis has been raised by European Group of Tumor Markers (EGTM) [152, 160], and by the National academy of Clinical Biochemistry (NACB) [152, 161]. Some noted caveats include: elevated CA 19-9 can be similarly detected in non-malignant patients diagnosed with acute and chronic pancreatitis, obstructive jaundice, and liver cirrhosis [134, 152, 162], and in gastrointestinal carcinomas [134, 158, 162–163]; furthermore, false positive results may result based on detection of a low molecular weight antigen, non-CA 19-9 [164]. Thus, the European Group of Tumor Markers (EGTM) [152, 160] the National academy of Clinical Biochemistry (NACB) recommend that CA 19-9 should not be the only indicator used for diagnosing PC.
Other potential PC biomarkers include (1) elevation in MMP-7 in plasma and pancreatic juice to discriminate between benign disease and carcinoma when combined with CA 19-9 [165], (2) a 2–10 fold increase in AGR2 in pancreatic juice, for the early detection of PC [166], (3) changes in S100A6 in pancreatic juice, for early detection of the disease [167], (4) elevated telomerase activity in pancreatic juice [168], (5) serine proteinase-2 (PRSS2) pre-protein and pancreatic lipase related protein-1 (PLRP1) in pancreatic juice of PC patients [72], and perineural invasion in PC [169]. Recently, a few studies have investigated whether use of biomarker panels may enhance the specificity and improve the diagnosis of PC [170–171]. The results showed that grouped biomarkers are comprised of CA 19-9, MCSF, CEA, SAA, Haptoglobin, TSGF, CA 242, and HSP27 only showed comparable sensitivity and specificity to single serum markers [170]; in contrast, combined assessment of CA 19-9, ICAM-1, and OPG gave 78/94% sensitivity/specificity and represented a significant improvement over individual CA 19-9 measurements for differentiating PC patients from healthy controls and subjects diagnosed with benign disease [171].
CANCER STEM CELLS AND CTC
The prevention of tumor relapse in PC has not been successful [172–173]. The underlying mechanisms are multi-faceted and may be attributed to drug resistance and insensitivity to repeated treatments, as well as the presence of pancreatic cancer stem cells (CSC) [174] which provide gain-of-function via their ability to undergo self-renewal, differentiation [175], and metastasis [172]. Accordingly, several recent studies have confirmed the presence of CTC in the blood stream and utilized CTC in the tumor tissue as a novel approach to develop biomarkers for PC. Experiments have shown that the presence of CTC might prove useful for prognosis in PC patients [176–178]. Although preliminary results from studies of CSC and CTC are encouraging, the technologies required for the capture of CSC or CTC are still at an early stage for routine application in PC diagnosis.
Likewise, mixed lymphocytes have shown promise for predicting patient’s outcome in PC. For instance, an overall survival of 2.4 months was predicted in PC patients based on an elevated neutrophil to lymphocyte ratio [179]; the elevated levels of micronuclei in peripheral lymphocytes may have an association with PC risk [180]. Experimentally, identity of mixed lymphocytes may be validated by the RT-PCR amplification of CK-19 in blood [181]. However, the specificity of these measurements may be confounded by innumerable factors that affect the lymphocyte populations in the blood stream.
FUTURE PERSPECTIVES
In conclusion, the establishment of early diagnostic biomarkers is critical to increase survival of PC patients. So far, no single marker has been identified with the specificity and reliability needed for screening PC during the early stages of the disease, even though a wide array of serum and tissue-based candidate markers have been identified. Biomarkers in body fluids have considerable potential for clinical diagnosis and prognosis; however, their application is limited by their low concentration compared to what might be available in the tumor tissue. The possibility that CA 19-9 may prove beneficial in diagnosing PC is supported by its 90% specificity to PC, but its reliability is still open to debate. Furthermore, more basic studies using cell lines, animal model experiment systems, and patient samples will be required to better understand the PC biology as a necessary requisite to identify more specific biomarkers for PC. The development of new methods to detect potential biomarkers with higher specificity and accuracy even for low concentrations in the body fluids would further advance biomarker research.
Promising new area for PC biomarker research and application lies in an in-depth analysis of CTCs and CSCs. New technologies and approaches must be developed for isolating these cells, keeping them in culture without changing their genotype or phenotype, and understanding the roles which they might play in the genetic evolution of PC, will go a long way in facilitating PC diagnosis and developing novel management and treatment strategies.
ACKNOWLEDGMENTS
We wish to extend our thanks to Mary Pull (North Dakota State University) for her thoughtful reading of the manuscript. We have attempted to include the most relevant materials in the field, so we apologize to authors whose work we have inadvertently overlooked. This project was supported by ND EPSCoR funds (EW), North Dakota State University Faculty Funds (EW) and Pilot Project Grant (EW) from the Centers of Biomedical Research Excellence (COBRE) grant NIH P20 RR020151 from the National Center for Research Resources (NCRR). NCRR is a component of the National Institutes of Health (NIH). The contents of this report are solely the responsibility of the authors and do not necessarily reflect the official views of the NIH or NCRR. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
ABBREVIATIONS
- PC
Pancreatic cancer
- CT
Computed tomography
- MRI
Magnetic resonance imaging
- EUS
Endoscopic ultrasound
- PET
Positron emission tomography
- MRCP
Magnetic resonance cholangiopancreatography
- ERCP
Endoscopic retrograde cholangiopancreatography
- FNA
Fine-needle aspiration
- PanIN
Pancreatic intraepithelial neoplasia
- SNP
Single nucleotide polymorphism
- MSP
Methylation specific PCR
- BSP
Bisulfite-sequencing PCR
- TMA
Tissue microarray
- DIGE
Differential in-gel electrophoresis
- MS
Mass spectrometry
- SELDI
Surface enhanced laser desortion/ionization
- ELISA
Enzyme linked immune sorbent assay
- MALDI
Matrix assisted laser desorption/ionization
- DTC
Disseminated tumor cells
- CTC
Circulating tumor cells
- PAC
Pancreatic adenocarcinoma
- PTGS
Post transcriptional gene silencing
- CA 19-9
Carbohydrate antigen 19-9
- EGTM
European group of tumor markers
- NACB
National academy of clinical biochemistry
- CSC
Cancer stem cells
- 2DE
2 dimensional electrophoresis
- MudPIT
Multi-dimensional protein identification technology
- ICAT
Isotope coded affinity tag
- iTRAQ
Isobaric tags for relative and absolute quantification
REFERENCES
- 1.Chakraborty S, Baine MJ, Sasson AR, Batra SK. Current status of molecular markers for early detection of sporadic pancreatic cancer. Biochim Biophys Acta. 2011;1815(1):44–64. doi: 10.1016/j.bbcan.2010.09.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Verma M. Pancreatic Cancer Biomarkers and Their Implication in Cancer Diagnosis and Epidemiology. Cancers. 2010;2:1830–1837. doi: 10.3390/cancers2041830. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Siegel R, Ward E, Brawley O, Jemal A. Cancer statistics, 2011: the impact of eliminating socioeconomic and racial disparities on pre-mature cancer deaths. CA Cancer J Clin. 2011;61(4):212–236. doi: 10.3322/caac.20121. [DOI] [PubMed] [Google Scholar]
- 4.Fry LC, Monkemuller K, Malfertheiner P. Molecular markers of pancreatic cancer: development and clinical relevance. Langenbecks Arch Surg. 2008;393:883–890. doi: 10.1007/s00423-007-0276-0. [DOI] [PubMed] [Google Scholar]
- 5.Jimeno A, Hidalgo M. Molecular biomarkers: their increasing role in the diagnosis, characterization, and therapy guidance in pancreatic cancer. Mol Cancer Ther. 2006;5:787–796. doi: 10.1158/1535-7163.MCT-06-0005. [DOI] [PubMed] [Google Scholar]
- 6.Strimpakos AS, Syrigos KN, Saif MW. Translational research in pancreatic cancer. JOP; Highlights from the "2010 ASCO Gastrointestinal Cancers Symposium"; January 22–24, 2010; Orlando, FL, USA. pp. 124–127. [PubMed] [Google Scholar]
- 7.Stathis A, Moore MJ. Advanced pancreatic carcinoma: current treatment and future challenges. Nat Rev Clin Oncol. 2010;7:163–172. doi: 10.1038/nrclinonc.2009.236. [DOI] [PubMed] [Google Scholar]
- 8.Holly EA, Chaliha I, Bracci PM, Gautam M. Signs and symptoms of pancreatic cancer: a population-based case-control study in the San Francisco Bay area. Clin Gastroenterol Hepatol. 2004;2:510–517. doi: 10.1016/s1542-3565(04)00171-5. [DOI] [PubMed] [Google Scholar]
- 9.Schmassmann A. Pancreatic carcinoma: conservative and adjuvant therapy. Ther Umsch. 1996;53:387–393. [PubMed] [Google Scholar]
- 10.Isozaki H, Okajima K, Morita S, et al. Review of pancreatic cancer. Bull Osaka Med Coll. 1990;36:1–11. [PubMed] [Google Scholar]
- 11.Costamagna G, Cotroneo AR, Mutignani M, Di Stasi C, Perri V. Carcinoma of the pancreatic head area. Therapy: nonoperative biliary drainage for palliation. Rays. 1995;20:326–337. [PubMed] [Google Scholar]
- 12.el-Kamar FG, Grossbard ML, Kozuch PS. Metastatic pancreatic cancer: emerging strategies in chemotherapy and palliative care. Oncologist. 2003;8:18–34. doi: 10.1634/theoncologist.8-1-18. [DOI] [PubMed] [Google Scholar]
- 13.Robinson PA. The role of MRI in pancreatic cancer. Eur Radiol. 2002;12:267–269. doi: 10.1007/s00330-001-1148-6. [DOI] [PubMed] [Google Scholar]
- 14.Stroszczynski C, Grutzmann R, Kittner T. CT and MR imaging of pancreatic cancer. Recent Results Cancer Res. 2008;177:5–14. doi: 10.1007/978-3-540-71279-4_2. [DOI] [PubMed] [Google Scholar]
- 15.Ardengh JC, de Paulo GA, Ferrari AP. Pancreatic carcinomas smaller than 3.0 cm: endosonography (EUS) in diagnosis, staging and prediction of resectability. HPB (Oxford) 2003;5:226–230. doi: 10.1080/13651820310001342. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Klapman J, Malafa MP. Early detection of pancreatic cancer: why, who, and how to screen. Cancer Control. 2008;15:280–287. doi: 10.1177/107327480801500402. [DOI] [PubMed] [Google Scholar]
- 17.Serrano OK, Chaudhry MA, Leach SD. The role of PET scanning in pancreatic cancer. Adv Surg. 2010;44:313–325. doi: 10.1016/j.yasu.2010.05.007. [DOI] [PubMed] [Google Scholar]
- 18.Adamek HE, Albert J, Breer H, Weitz M, Schilling D, Riemann JF. Pancreatic cancer detection with magnetic resonance cholangiopancreatography and endoscopic retrograde cholangiopancreatography: a prospective controlled study. Lancet. 2000;356:190–193. doi: 10.1016/S0140-6736(00)02479-X. [DOI] [PubMed] [Google Scholar]
- 19.Erickson RA. ERCP and pancreatic cancer. Annals of Surgical Oncology. 2004;11:555–557. doi: 10.1245/ASO.2004.03.904. [DOI] [PubMed] [Google Scholar]
- 20.Ku YM, Shin SS, Lee CH, Semelka RC. Magnetic resonance imaging of cystic and endocrine pancreatic neoplasms. Top Magn Reson Imaging. 2009;20:11–18. doi: 10.1097/RMR.0b013e3181b4812c. [DOI] [PubMed] [Google Scholar]
- 21.Bournet B, Souque A, Senesse P, et al. Endoscopic ultrasound-guided fine-needle aspiration biopsy coupled with KRAS mutation assay to distinguish pancreatic cancer from pseudotumoral chronic pancreatitis. Endoscopy. 2009;41:552–557. doi: 10.1055/s-0029-1214717. [DOI] [PubMed] [Google Scholar]
- 22.Salek C, Benesova L, Zavoral M, et al. Evaluation of clinical relevance of examining K-ras, p16 and p53 mutations along with allelic losses at 9p and 18q in EUS-guided fine needle aspiration samples of patients with chronic pancreatitis and pancreatic cancer. World J Gastroenterol. 2007;13:3714–3720. doi: 10.3748/wjg.v13.i27.3714. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.American gastroenterological association medical position statement: epidemiology, diagnosis, and treatment of pancreatic ductal adenocarcinoma. Gastroenterology. 1999;117:1463–1484. doi: 10.1016/s0016-5085(99)70297-0. [DOI] [PubMed] [Google Scholar]
- 24.Pitt HA. Curative treatment for pancreatic neoplasms. Standard resection. Surg Clin North Am. 1995;75:891–904. doi: 10.1016/s0039-6109(16)46734-4. [DOI] [PubMed] [Google Scholar]
- 25.Wray CJ, Ahmad SA, Matthews JB, Lowy AM. Surgery for pancreatic cancer: recent controversies and current practice. Gastroenterology. 2005;128:1626–1641. doi: 10.1053/j.gastro.2005.03.035. [DOI] [PubMed] [Google Scholar]
- 26.Zervos EE, Rosemurgy AS, Al-Saif O, Durkin AJ. Surgical management of early-stage pancreatic cancer. Cancer Control. 2004;11:23–31. doi: 10.1177/107327480401100104. [DOI] [PubMed] [Google Scholar]
- 27.Heinemann V. Gemcitabine: progress in the treatment of pancreatic cancer. Oncology. 2001;60:8–18. doi: 10.1159/000055290. [DOI] [PubMed] [Google Scholar]
- 28.Mamon HJ, Niedzwiecki D, Hollis D, et al. A phase 2 trial of gemcitabine, 5-florouracil, and radiation therapy in locally advanced nonmetastatic pancreatic adenocarcinoma: cancer and Leukemia Group B (CALGB) 80003. Cancer. 2010;117:2620–2628. doi: 10.1002/cncr.25742. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Kim MP, Gallick GE. Gemcitabine resistance in pancreatic cancer: picking the key players. Clin Cancer Res. 2008;14:1284–1285. doi: 10.1158/1078-0432.CCR-07-2247. [DOI] [PubMed] [Google Scholar]
- 30.Hidalgo M. Pancreatic cancer. N Engl J Med. 2010;362:1605–1617. doi: 10.1056/NEJMra0901557. [DOI] [PubMed] [Google Scholar]
- 31.Hazard L. The role of radiation therapy in pancreas cancer. Gastrointest Cancer Res. 2009;3:20–28. [PMC free article] [PubMed] [Google Scholar]
- 32.Le Scodan R, Mornex F, Girard N, et al. Preoperative chemoradiation in potentially resectable pancreatic adenocarcinoma: feasibility, treatment effect evaluation and prognostic factors, analysis of the SFRO-FFCD 9704 trial and literature review. Ann Oncol. 2009;20:1387–1396. doi: 10.1093/annonc/mdp015. [DOI] [PubMed] [Google Scholar]
- 33.Cooke EW, Hazard L. Curative radiation therapy for pancreatic malignancies. Surg Clin North Am. 2010;90:341–354. doi: 10.1016/j.suc.2009.12.002. [DOI] [PubMed] [Google Scholar]
- 34.Lowenfels AB, Maisonneuve P. Epidemiology and risk factors for pancreatic cancer. Best Pract Res Clin Gastroenterol. 2006;20:197–209. doi: 10.1016/j.bpg.2005.10.001. [DOI] [PubMed] [Google Scholar]
- 35.Lowenfels AB, Maisonneuve P. Risk factors for pancreatic cancer. J Cell Biochem. 2005;95:649–656. doi: 10.1002/jcb.20461. [DOI] [PubMed] [Google Scholar]
- 36.Morse DL, Balagurunathan Y, Hostetter G, et al. Identification of novel pancreatic adenocarcinoma cell-surface targets by gene expression profiling and tissue microarray. Biochem Pharmacol. 2010;80:748–754. doi: 10.1016/j.bcp.2010.05.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Takayama R, Nakagawa H, Sawaki A, et al. Serum tumor antigen REG4 as a diagnostic biomarker in pancreatic ductal adenocarcinoma. J Gastroenterol. 2010;45:52–59. doi: 10.1007/s00535-009-0114-y. [DOI] [PubMed] [Google Scholar]
- 38.Talamini G, Bassi C, Falconi M, et al. Alcohol and smoking as risk factors in chronic pancreatitis and pancreatic cancer. Digestive Diseases Sci. 1999;44:1303–1311. doi: 10.1023/a:1026670911955. [DOI] [PubMed] [Google Scholar]
- 39.Harada T, Chelala C, Bhakta V, et al. Genome-wide DNA copy number analysis in pancreatic cancer using high-density single nucleotide polymorphism arrays. Oncogene. 2008;27:1951–1960. doi: 10.1038/sj.onc.1210832. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Bardeesy N, DePinho RA. Pancreatic cancer biology and genetics. Nat Rev Cancer. 2002;2:897–909. doi: 10.1038/nrc949. [DOI] [PubMed] [Google Scholar]
- 41.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;10:66–73. doi: 10.1159/000231984. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Klimstra DS, Longnecker DS. K-ras mutations in pancreatic ductal proliferative lesions. Am J Pathol. 1994;145:1547–1550. [PMC free article] [PubMed] [Google Scholar]
- 43.Pellegata NS, Sessa F, Renault B, et al. K-ras and p53 gene mutations in pancreatic cancer: ductal and nonductal tumors progress through different genetic lesions. Cancer Res. 1994;54:1556–1560. [PubMed] [Google Scholar]
- 44.Ghaneh P, Costello E, Neoptolemos JP. Biology and management of pancreatic cancer. Postgrad Med J. 2008;84:478–497. doi: 10.1136/gut.2006.103333. [DOI] [PubMed] [Google Scholar]
- 45.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;160:1573–1581. doi: 10.1016/S0002-9440(10)61104-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Ueki T, Toyota M, Skinner H, et al. Identification and characterization of differentially methylated CpG islands in pancreatic carcinoma. Cancer Res. 2001;61:8540–8546. [PubMed] [Google Scholar]
- 47.Korc M, Chandrasekar B, Yamanaka Y, Friess H, Buchier M, Beger HG. Overexpression of the epidermal growth factor receptor in human pancreatic cancer is associated with concomitant increases in the levels of epidermal growth factor and transforming growth factor alpha. J Clin Invest. 1992;90:1352–1360. doi: 10.1172/JCI116001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Barton CM, Hall PA, Hughes CM, Gullick WJ, Lemoine NR. Transforming growth factor alpha and epidermal growth factor in human pancreatic cancer. J Pathol. 1991;163:111–116. doi: 10.1002/path.1711630206. [DOI] [PubMed] [Google Scholar]
- 49.Friess H, Berberat P, Schilling M, Kunz J, Korc M, Buchler MW. Pancreatic cancer: the potential clinical relevance of alterations in growth factors and their receptors. J Mol Med. 1996;74:35–42. doi: 10.1007/BF00202070. [DOI] [PubMed] [Google Scholar]
- 50.Simeone DM, Ji B, Banerjee M, et al. CEACAM1, a novel serum biomarker for pancreatic cancer. Pancreas. 2007;34:436–443. doi: 10.1097/MPA.0b013e3180333ae3. [DOI] [PubMed] [Google Scholar]
- 51.Schutte M, Hruban RH, Geradts J, et al. Abrogation of the Rb/p16 tumor-suppressive pathway in virtually all pancreatic carcinomas. Cancer Res. 1997;57:3126–3130. [PubMed] [Google Scholar]
- 52.Yamano M, Fujii H, Takagaki T, Kadowaki N, Watanabe H, Shirai T. Genetic progression and divergence in pancreatic carcinoma. Am J Pathol. 2000;156:2123–2133. doi: 10.1016/S0002-9440(10)65083-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Boschman CR, Stryker S, Reddy JK, Rao MS. Expression of P53 Protein in Precursor Lesions and Adenocarcinoma of Human Pancreas. Am J Pathol. 1994;145:1291–1295. [PMC free article] [PubMed] [Google Scholar]
- 54.Zindy F, Quelle DE, Roussel MF, Sherr CJ. Expression of the p16(INK4a) tumor suppressor versus other INK4 family members during mouse development and aging. Oncogene. 1997;15:203–211. doi: 10.1038/sj.onc.1201178. [DOI] [PubMed] [Google Scholar]
- 55.Liu L, Dilworth D, Gao LZ, et al. Mutation of the CDKN2A 5 ' UTR creates an aberrant initiation codon and predisposes to melanoma. Nat Genetics. 1999;21:128–132. doi: 10.1038/5082. [DOI] [PubMed] [Google Scholar]
- 56.DiGiuseppe JA, Hruban RH, Goodman SN, et al. Overexpression of p53 protein in adenocarcinoma of the pancreas. Am J Clin Pathol. 1994;101:684–688. doi: 10.1093/ajcp/101.6.684. [DOI] [PubMed] [Google Scholar]
- 57.Goggins M, Hruban RH, Kern SE. BRCA2 is inactivated late in the development of pancreatic intraepithelial neoplasia - Evidence and implications. Am J Pathol. 2000;156:1767–1771. doi: 10.1016/S0002-9440(10)65047-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Wilentz RE, Iacobuzio-Donahue CA, Argani P, et al. Loss of expression of Dpc4 in pancreatic intraepithelial neoplasia: Evidence that DPC4 inactivation occurs late in neoplastic progression. Cancer Res. 2000;60:2002–2006. [PubMed] [Google Scholar]
- 59.Asomaning K, Reid AE, Zhou W, et al. MDM2 promoter polymorphism and pancreatic cancer risk and prognosis. Clinical Cancer Res. 2008;14:4010–4015. doi: 10.1158/1078-0432.CCR-07-4187. [DOI] [PubMed] [Google Scholar]
- 60.Dong M, Ma G, Tu W, Guo KJ, Tian YL, Dong YT. Clinicopathological significance of p53 and mdm2 protein expression in human pancreatic cancer. World J Gastroenterol. 2005;11:2162–2165. doi: 10.3748/wjg.v11.i14.2162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Wehler T, Wolfert F, Schimanski CC, et al. Strong expression of chemokine receptor CXCR4 by pancreatic cancer correlates with advanced disease. Oncology Reports. 2006;16:1159–1164. [PubMed] [Google Scholar]
- 62.Thomas RM, Kim J, Revelo-Penafiel MP, Angel R, Dawson DW, Lowy AM. The chemokine receptor CXCR4 is expressed in pancreatic intraepithelial neoplasia. Gut. 2008;57:1555–1560. doi: 10.1136/gut.2007.143941. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Thomas RM, Toney K, Fenoglio-Preiser C, et al. The RON receptor tyrosine kinase mediates oncogenic phenotypes in pancreatic cancer cells and is increasingly expressed during pancreatic cancer progression. Cancer Res. 2007;67:6075–6082. doi: 10.1158/0008-5472.CAN-06-4128. [DOI] [PubMed] [Google Scholar]
- 64.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;63:3735–3742. [PubMed] [Google Scholar]
- 65.Selaru FM, David S, Meltzer SJ, Hamilton JP. Epigenetic Events in Gastrointestinal Cancer. Am J Gastroenterol. 2009;104:1910–1912. doi: 10.1038/ajg.2008.145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Koorstra JBM, Feldmann G, Habbe N, Maitra A. Morphogenesis of pancreatic cancer: role of pancreatic intraepithelial neoplasia (PanINs) Langenbecks Archives Surgery. 2008;393:561–570. doi: 10.1007/s00423-008-0282-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.van Heek NT, Meeker AK, Kern SE, et al. Telomere shortening is nearly universal in pancreatic intraepithelial neoplasia. Am J Pathol. 2002;161:1541–1547. doi: 10.1016/S0002-9440(10)64432-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Gisselsson D, Jonson T, Petersen A, et al. Telomere dysfunction triggers extensive DNA fragmentation and evolution of complex chromosome abnormalities in human malignant tumors. Proceed Nat Academy Sci USA. 2001;98:12683–12688. doi: 10.1073/pnas.211357798. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Lopez-Casas PP, Lopez-Fernandez LA. Gene-expression profiling in pancreatic cancer. Expert Rev Mol Diagn. 2010;10:591–601. doi: 10.1586/erm.10.43. [DOI] [PubMed] [Google Scholar]
- 70.Laurell H, Bouisson M, Berthelemy P, et al. Identification of biomarkers of human pancreatic adenocarcinomas by expression profiling and validation with gene expression analysis in endoscopic ultrasound-guided fine needle aspiration samples. World J Gastroenterol. 2006;12:3344–3351. doi: 10.3748/wjg.v12.i21.3344. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Liang JJ, Kimchi ET, Staveley-O'Carroll KF, Tan D. Diagnostic and prognostic biomarkers in pancreatic carcinoma. Int J Clin Exp Pathol. 2009;2:1–10. [PMC free article] [PubMed] [Google Scholar]
- 72.Gao J, Zhu F, Lv S, Li Z, Ling Z, Gong Y, et al. Identification of pancreatic juice proteins as biomarkers of pancreatic cancer. Oncol Rep. 2010;23:1683–1692. doi: 10.3892/or_00000812. [DOI] [PubMed] [Google Scholar]
- 73.Iacobuzio-Donahue CA, Maitra A, Olsen M, et al. Exploration of global gene expression patterns in pancreatic adenocarcinoma using cDNA microarrays. Am J Pathol. 2003;162:1151–1162. doi: 10.1016/S0002-9440(10)63911-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Wong SCC, Chan CML, Ma BBY, et al. Advanced proteomic technologies for cancer biomarker discovery. Expert Rev Proteomics. 2009;6:123–134. doi: 10.1586/epr.09.1. [DOI] [PubMed] [Google Scholar]
- 75.Ma N, Ge C-l, Luan F-m, et al. Serum protein fingerprint of patients with pancreatic cancer by SELDI technology. Chinese J Cancer Res. 2008;20:171–176. [Google Scholar]
- 76.Xue A, Scarlett CJ, Chung L, et al. Discovery of serum biomarkers for pancreatic adenocarcinoma using proteomic analysis. British J Cancer. 2010;103:391–400. doi: 10.1038/sj.bjc.6605764. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Yu KH, Rustgi AK, Blair IA. Characterization of proteins in human pancreatic cancer serum using differential gel electrophoresis and tandem mass spectrometry. J Proteome Res. 2005;4:1742–1751. doi: 10.1021/pr050174l. [DOI] [PubMed] [Google Scholar]
- 78.Tambor V, Fucikova A, Lenco J, et al. Application of Proteomics in Biomarker Discovery: a Primer for the Clinician. Physiological Res. 2010;59:471–497. doi: 10.33549/physiolres.931758. [DOI] [PubMed] [Google Scholar]
- 79.Chen R, Pan S, Yi EC, et al. Quantitative proteomic profiling of pancreatic cancer juice. Proteomics. 2006;6:3871–3879. doi: 10.1002/pmic.200500702. [DOI] [PubMed] [Google Scholar]
- 80.Zhang L, Farrell JJ, Zhou H, et al. Salivary transcriptomic biomarkers for detection of resectable pancreatic cancer. Gastroenterology. 2010;138:949–957. e1–e7. doi: 10.1053/j.gastro.2009.11.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Haug U, Wente MN, Seiler CM, Jesnowski R, Brenner H. Stool testing for the early detection of pancreatic cancer: rationale and current evidence. Expert Rev Mol Diagn. 2008;8:753–759. doi: 10.1586/14737159.8.6.753. [DOI] [PubMed] [Google Scholar]
- 82.Brody JR, Costantino CL, Potoczek M, et al. Adenosquamous carcinoma of the pancreas harbors KRAS2, DPC4 and TP53 molecular alterations similar to pancreatic ductal adenocarcinoma. Mod Pathol. 2009;22:651–659. doi: 10.1038/modpathol.2009.15. [DOI] [PubMed] [Google Scholar]
- 83.Liu F. SMAD4/DPC4 and pancreatic cancer survival. Commentary re: M. Tascilar et al., The SMAD4 protein and prognosis of pancreatic ductal adenocarcinoma. Clin Cancer Res. 2001;7:4115–4121. Clin Cancer Res 2001; 7: 3853-6. [PubMed] [Google Scholar]
- 84.Biankin AV, Morey AL, Lee CS, et al. DPC4/Smad4 expression and outcome in pancreatic ductal adenocarcinoma. J Clin Oncol. 2002;20:4531–4542. doi: 10.1200/JCO.2002.12.063. [DOI] [PubMed] [Google Scholar]
- 85.Gu LJ, Chen J, Lu ZH, Li L, Zhou WX, Luo YF. [Expression of DPC4/Smad4, p21wafI, and p16 in human pancreatic cancer] Ai Zheng. 2002;21:132–137. [PubMed] [Google Scholar]
- 86.Kipp BR, Fritcher EG, Clayton AC, et al. Comparison of KRAS mutation analysis and FISH for detecting pancreatobiliary tract cancer in cytology specimens collected during endoscopic retrograde cholangiopancreatography. J Mol Diagn. 2010;12:780–786. doi: 10.2353/jmoldx.2010.100016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Wu X, Lu XH, Xu T, et al. Evaluation of the diagnostic value of serum tumor markers, and fecal k-ras and p53 gene mutations for pancreatic cancer. Chin J Dig Dis. 2006;7:170–174. doi: 10.1111/j.1443-9573.2006.00263.x. [DOI] [PubMed] [Google Scholar]
- 88.Zinsky R, Bolukbas S, Bartsch H, Schirren J, Fisseler-Eckhoff A. Analysis of KRAS Mutations of Exon 2 Codons 12 and 13 by SNaPshot Analysis in Comparison to Common DNA Sequencing. Gastroenterol Res Pract. 2010 doi: 10.1155/2010/789363. 789363. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Morton JP, Timpson P, Karim SA, et al. Mutant p53 drives metastasis and overcomes growth arrest/senescence in pancreatic cancer. Proc Natl Acad Sci USA. 2009;107:246–251. doi: 10.1073/pnas.0908428107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Jeong J, Park YN, Park JS, Yoon DS, Chi HS, Kim BR. Clinical significance of p16 protein expression loss and aberrant p53 protein expression in pancreatic cancer. Yonsei Med J. 2005;46:519–525. doi: 10.3349/ymj.2005.46.4.519. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Gerdes B, Ramaswamy A, Kersting M, et al. p16(INK4a) alterations in chronic pancreatitis-indicator for high-risk lesions for pancreatic cancer. Surgery. 2001;129:490–497. doi: 10.1067/msy.2001.112071. [DOI] [PubMed] [Google Scholar]
- 92.Maitra A, Molberg K, Albores-Saavedra J, Lindberg G. Loss of Dpc4 expression in colonic adenocarcinomas correlates with the presence of metastatic disease. Am J Pathol. 2000;157:1105–1111. doi: 10.1016/S0002-9440(10)64625-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.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;27:1806–1813. doi: 10.1200/JCO.2008.17.7188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Maitra A, Kern SE, Hruban RH. Molecular pathogenesis of pancreatic cancer. Best Pract Res Clin Gastroenterol. 2006;20:211–226. doi: 10.1016/j.bpg.2005.10.002. [DOI] [PubMed] [Google Scholar]
- 95.Jiang Y, Mackley H, Cheng H, Ajani JA. Use of K-Ras as a predictive biomarker for selecting anti-EGF receptor/pathway treatment. Biomark Med. 2010;4:535–541. doi: 10.2217/bmm.10.74. [DOI] [PubMed] [Google Scholar]
- 96.Morris JP, Wang SC, Hebrok M. KRAS, Hedgehog, Wnt and the twisted developmental biology of pancreatic ductal adenocarcinoma. Nature Rev Cancer. 2010;10:683–695. doi: 10.1038/nrc2899. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Rustgi AK. The molecular pathogenesis of pancreatic cancer: clarifying a complex circuitry. Genes & Development. 2006;20:3049–3053. doi: 10.1101/gad.1501106. [DOI] [PubMed] [Google Scholar]
- 98.Slater EP, Langer P, Fendrich V, et al. Prevalence of BRCA2 and CDKN2a mutations in German familial pancreatic cancer families. Fam Cancer. 2010;9:335–343. doi: 10.1007/s10689-010-9329-6. [DOI] [PubMed] [Google Scholar]
- 99.Matsubayashi H, Canto M, Sato N, et al. DNA methylation alterations in the pancreatic juice of patients with suspected pancreatic disease. Cancer Res. 2006;66:1208–1217. doi: 10.1158/0008-5472.CAN-05-2664. [DOI] [PubMed] [Google Scholar]
- 100.Bausch D, Thomas S, Mino-Kenudson M, et al. Plectin-1 as a novel biomarker for pancreatic cancer. Clin Cancer Res. 2010;17:302–309. doi: 10.1158/1078-0432.CCR-10-0999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Balasenthil S, Chen N, Lott ST, et al. A migration signature and plasma biomarker panel for pancreatic adenocarcinoma. Cancer Prev Res (Phila) 2010;4:137–149. doi: 10.1158/1940-6207.CAPR-10-0025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Kurtova A, Sivina M, Quiroga MP, Wierda WG, Keating MJ, Burger JA. The Immunophenotype Signature CD49d(+)CD38(+) Identifies Chronic Lymphocytic Leukemia Cases with a Higher Potential for Migration Beneath Marrow Stromal Cells. Blood. 2009;114:150–151. [Google Scholar]
- 103.Sakorafas GH, Tsiotou AG, Tsiotos GG. Molecular biology of pancreatic cancer; oncogenes, tumour suppressor genes, growth factors, and their receptors from a clinical perspective. Cancer Treat Rev. 2000;26:29–52. doi: 10.1053/ctrv.1999.0144. [DOI] [PubMed] [Google Scholar]
- 104.Mulero-Navarro S, Esteller M. Epigenetic biomarkers for human cancer: The time is now. Critical Rev Oncol Hematol. 2008;68:1–11. doi: 10.1016/j.critrevonc.2008.03.001. [DOI] [PubMed] [Google Scholar]
- 105.Tan AC, Jimeno A, Lin SH, et al. Characterizing DNA methylation patterns in pancreatic cancer genome. Molecular Oncology. 2009;3:425–438. doi: 10.1016/j.molonc.2009.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Omura N, Li CP, Li A, et al. Genome-wide profiling of methylated promoters in pancreatic adenocarcinoma. Cancer Biol Therapy. 2008;7:1146–1156. doi: 10.4161/cbt.7.7.6208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Ohtsubo K, Watanabe H, Yao F, et al. Preproenkephalin hypermethylation in the pure pancreatic juice compared with p53 mutation in the diagnosis of pancreatic carcinoma. J Gastroenterol. 2006;41:791–797. doi: 10.1007/s00535-006-1857-3. [DOI] [PubMed] [Google Scholar]
- 108.Shimizu K, Itsuzaki Y, Fujii H, Honoki K, Tsujiuchi T. Reduced expression of the Rassf1a gene and its aberrant DNA methylation in pancreatic duct adenocarcinomas induced by N-nitrosobis(2-oxopropyl)amine in hamsters. Mol Carcinog. 2008;47:80–87. doi: 10.1002/mc.20250. [DOI] [PubMed] [Google Scholar]
- 109.Pizzi S, Azzoni C, Bottarelli L, et al. RASSF1A promoter methylation and 3p21.3 loss of heterozygosity are features of foregut, but not midgut and hindgut, malignant endocrine tumours. J Pathol. 2005;206:409–416. doi: 10.1002/path.1784. [DOI] [PubMed] [Google Scholar]
- 110.House MG, Herman JG, Guo MZ, et al. Aberrant hypermethylation of tumor suppressor genes in pancreatic endocrine neoplasms. Ann Surg. 2003;238:423–431. doi: 10.1097/01.sla.0000086659.49569.9e. discussion 31-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Dammann R, Schagdarsurengin U, Liu LM, et al. Frequent RASSF1A promoter hypermethylation and K-ras mutations in pancreatic carcinoma. Oncogene. 2003;22:3806–3812. doi: 10.1038/sj.onc.1206582. [DOI] [PubMed] [Google Scholar]
- 112.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;9:1446–1452. [PubMed] [Google Scholar]
- 113.Virmani A, Rathi A, Heda S, et al. Aberrant methylation of the Cyclin D2 promoter in primary small cell, nonsmall cell lung and breast cancers. Int J Cancer. 2003;107:341–345. doi: 10.1002/ijc.11393. [DOI] [PubMed] [Google Scholar]
- 114.Sakai M, Hibi K, Koshikawa K, et al. Frequent promoter methylation and gene silencing of CDH13 in pancreatic cancer. Cancer Sci. 2004;95:588–591. doi: 10.1111/j.1349-7006.2004.tb02491.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Watanabe H, Okada G, Ohtsubo K, et al. Aberrant methylation of secreted apoptosis-related protein 2 (SARP2) in pure pancreatic juice in diagnosis of pancreatic neoplasms. Pancreas. 2006;32:382–389. doi: 10.1097/01.mpa.0000221617.89376.38. [DOI] [PubMed] [Google Scholar]
- 116.Park JK, Ryu JK, Lee KH, et al. Quantitative analysis of NPTX2 hypermethylation is a promising molecular diagnostic marker for pancreatic cancer. Pancreas. 2007;35:e9–e15. doi: 10.1097/MPA.0b013e318153fa42. [DOI] [PubMed] [Google Scholar]
- 117.Melnikov AA, Scholtens D, Talamonti MS, Bentrem DJ, Levenson VV. Methylation Profile of Circulating Plasma DNA in Patients With Pancreatic Cancer. J Surgical Oncol. 2009;99:119–122. doi: 10.1002/jso.21208. [DOI] [PubMed] [Google Scholar]
- 118.Manuyakorn A, Paulus R, Farrell J, et al. Cellular Histone Modification Patterns Predict Prognosis and Treatment Response in Resectable Pancreatic Adenocarcinoma: Results From RTOG 9704. J Clinical Oncol. 2010;28:1358–1365. doi: 10.1200/JCO.2009.24.5639. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.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;70:5226–5237. doi: 10.1158/0008-5472.CAN-09-4227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Nimmo RA, Slack FJ. An elegant miRror: microRNAs in stem cells, developmental timing and cancer. Chromosoma. 2009;118:405–418. doi: 10.1007/s00412-009-0210-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Croce CM. Causes and consequences of microRNA dysregulation in cancer. Nat Rev Genet. 2009;10:704–714. doi: 10.1038/nrg2634. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Rosenfeld N, Aharonov R, Meiri E, et al. MicroRNAs accurately identify cancer tissue origin. Nat Biotechnol. 2008;26:462–469. doi: 10.1038/nbt1392. [DOI] [PubMed] [Google Scholar]
- 123.Esquela-Kerscher A, Slack FJ. Oncomirs - microRNAs with a role in cancer. Nat Rev Cancer. 2006;6:259–269. doi: 10.1038/nrc1840. [DOI] [PubMed] [Google Scholar]
- 124.Bartels CL, Tsongalis GJ. [MicroRNAs: novel biomarkers for human cancer] Ann Biol Clin (Paris) 2009;68:263–272. doi: 10.1684/abc.2010.0429. [DOI] [PubMed] [Google Scholar]
- 125.Szafranska AE, Davison TS, John J, et al. MicroRNA expression alterations are linked to tumorigenesis and non-neoplastic processes in pancreatic ductal adenocarcinoma. Oncogene. 2007;26:4442–4452. doi: 10.1038/sj.onc.1210228. [DOI] [PubMed] [Google Scholar]
- 126.Kong X, Du Y, Wang G, et al. Erratum to: Detection of Differentially Expressed microRNAs in Serum of Pancreatic Ductal Adenocarcinoma Patients: miR-196a Could Be a Potential Marker for Poor Prognosis. Dig Dis Sci. 2011;56:602–609. doi: 10.1007/s10620-010-1285-3. [DOI] [PubMed] [Google Scholar]
- 127.Ho AS, Huang X, Cao H, et al. Circulating miR-210 as a Novel Hypoxia Marker in Pancreatic Cancer. Transl Oncol. 2010;3:109–113. doi: 10.1593/tlo.09256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Giovannetti E, Funel N, Peters GJ, et al. MicroRNA-21 in pancreatic cancer: correlation with clinical outcome and pharmacologic aspects underlying its role in the modulation of gemcitabine activity. Cancer Res. 2010;70:4528–4538. doi: 10.1158/0008-5472.CAN-09-4467. [DOI] [PubMed] [Google Scholar]
- 129.Hwang J-H, Voortman J, Giovannetti E, et al. Identification of MicroRNA-21 as a Biomarker for Chemoresistance and Clinical Outcome Following Adjuvant Therapy in Resectable Pancreatic Cancer. PLoS One. 2010;5:e10630. doi: 10.1371/journal.pone.0010630. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.du Rieu MC, Torrisani J, Selves J, et al. MicroRNA-21 is induced early in pancreatic ductal adenocarcinoma precursor lesions. Clin Chem. 2010;56:603–612. doi: 10.1373/clinchem.2009.137364. [DOI] [PubMed] [Google Scholar]
- 131.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;33:698–709. doi: 10.1007/s00268-008-9833-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Haas M, Laubender RP, Stieber P, et al. Prognostic relevance of CA 19-9, CEA, CRP, and LDH kinetics in patients treated with palliative second-line therapy for advanced pancreatic cancer. Tumour Biol. 2010;31:351–357. doi: 10.1007/s13277-010-0044-6. [DOI] [PubMed] [Google Scholar]
- 133.Ho JJ, Siddiki B, Kim YS. Association of sialyl-Lewis(a) and sialyl-Lewis(x) with MUC-1 apomucin ina pancreatic cancer cell line. Cancer Res. 1995;55:3659–3663. [PubMed] [Google Scholar]
- 134.Steinberg W. The clinical utility of the CA 19-9 tumor-associated antigen. Am J Gastroenterol. 1990;85:350–355. [PubMed] [Google Scholar]
- 135.Andren-Sandberg A. CA 50 and CA 19-9 in serum as tumor markers for pancreatic cancer: a review of the literature. Acta Chir Scand Suppl. 1989;549:75–81. [PubMed] [Google Scholar]
- 136.Kokhanenko N, Ignashov AM, Varga EV, et al. [Role of the tumor markers CA 19-9 and carcinoembryonic antigen (CEA) in diagnosis, treatment and prognosis of pancreatic cancer] Vopr Onkol. 2001;47:294–297. [PubMed] [Google Scholar]
- 137.Baeckstrom D, Hansson GC, Nilsson O, Johansson C, Gendler SJ, Lindholm L. Purification and characterization of a membrane-bound and a secreted mucin-type glycoprotein carrying the carcinoma-associated sialyl-Lea epitope on distinct core proteins. J Biol Chem. 1991;266:21537–21547. [PubMed] [Google Scholar]
- 138.Koprowski H, Herlyn M, Steplewski Z, Sears HF. Specific antigen in serum of patients with colon carcinoma. Science. 1981;212:53–55. doi: 10.1126/science.6163212. [DOI] [PubMed] [Google Scholar]
- 139.Koprowski H, Steplewski Z, Mitchell K, Herlyn M, Herlyn D, Fuhrer P. Colorectal carcinoma antigens detected by hybridoma antibodies. Somatic Cell Genet. 1979;5:957–971. doi: 10.1007/BF01542654. [DOI] [PubMed] [Google Scholar]
- 140.Magnani JL, Nilsson B, Brockhaus M, et al. A monoclonal antibody-defined antigen associated with gastrointestinal cancer is a ganglioside containing sialylated lacto-N-fucopentaose II. J Biol Chem. 1982;257:14365–14369. [PubMed] [Google Scholar]
- 141.Ugorski M, Laskowska A. Sialyl Lewis(a): a tumor-associated carbohydrate antigen involved in adhesion and metastatic potential of cancer cells. Acta Biochimica Polonica. 2002;49:303–311. [PubMed] [Google Scholar]
- 142.Vestergaard EM, Hein HO, Meyer H, et al. Reference values and biological variation for tumor marker CA 19-9 in serum for different Lewis and secretor genotypes and evaluation of secretor and Lewis genotyping in a Caucasian population. Clin Chem. 1999;45:54–61. [PubMed] [Google Scholar]
- 143.Hanisch FG, Uhlenbruck G, Dienst C. Structure of tumor-associated carbohydrate antigen Ca 19-9 on human seminal-plasma glycoproteins from healthy donors. Eur J Biochem. 1984;144:467–473. doi: 10.1111/j.1432-1033.1984.tb08489.x. [DOI] [PubMed] [Google Scholar]
- 144.Tempero MA, Uchida E, Takasaki H, Burnett DA, Steplewski Z, Pour PM. Relationship of Carbohydrate Antigen 19-9 and Lewis Antigens in Pancreatic-Cancer. Cancer Research. 1987;47:5501–5503. [PubMed] [Google Scholar]
- 145.Eskelinen M, Haglund U. Developments in serologic detection of human pancreatic adenocarcinoma. Scand J Gastroenterol. 1999;34:833–844. doi: 10.1080/003655299750025273. [DOI] [PubMed] [Google Scholar]
- 146.Sreejayan N, Wittig BM, Von Stillfried N, et al. Human biliary mucin binds to E-selectin: a possible role in modulation of inflammation. Am J Physiology-Gastrointestinal Liver Physiol. 2001;280:G1043–G1048. doi: 10.1152/ajpgi.2001.280.5.G1043. [DOI] [PubMed] [Google Scholar]
- 147.Kannagi R. Carbohydrate antigen sialyl Lewis a--its pathophysiological significance and induction mechanism in cancer progression. Chang Gung Med J. 2007;30:189–209. [PubMed] [Google Scholar]
- 148.Miyazaki K, Ohmori K, Izawa M, et al. Loss of disialyl Lewis(a), the ligand for lymphocyte inhibitory receptor sialic acid-binding immunoglobulin-like lectin-7 (Siglec-7) associated with increased sialyl Lewis(a) expression on human colon cancers. Cancer Res. 2004;64:4498–4505. doi: 10.1158/0008-5472.CAN-03-3614. [DOI] [PubMed] [Google Scholar]
- 149.Ritts RE, Delvillano BC, Go VLW, Herberman RB, Klug TL, Zurawski VR. Initial Clinical-Evaluation of an Immunoradiometric Assay for Ca-19-9 Using the Nci Serum Bank. Int J Cancer. 1984;33:339–345. doi: 10.1002/ijc.2910330310. [DOI] [PubMed] [Google Scholar]
- 150.Satake K, Kanazawa G, Kho I, Chung YS, Umeyama K. A Clinical-Evaluation of Carbohydrate Antigen 19-9 and Carcinoembryonic Antigen in Patients with Pancreatic-Carcinoma. J Surgical Oncol. 1985;29:15–21. doi: 10.1002/jso.2930290106. [DOI] [PubMed] [Google Scholar]
- 151.Safi F, Beger HG, Bittner R, Buchler M, Krautzberger W. Ca-19-9 and Pancreatic Adenocarcinoma. Cancer. 1986;57:779–783. doi: 10.1002/1097-0142(19860215)57:4<779::aid-cncr2820570417>3.0.co;2-c. [DOI] [PubMed] [Google Scholar]
- 152.Duffy MJ, Sturgeon C, Lamerz R, et al. Tumor markers in pancreatic cancer: a European Group on Tumor Markers (EGTM) status report. Ann Oncol. 2010;21:441–447. doi: 10.1093/annonc/mdp332. [DOI] [PubMed] [Google Scholar]
- 153.Tian FH, Appert HE, Myles J, Howard JM. Prognostic Value of Serum Ca-19-9 Levels in Pancreatic Adenocarcinoma. Annals Surgery. 1992;215:350–355. doi: 10.1097/00000658-199204000-00008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Glenn J, Steinberg WM, Kurtzman SH, Steinberg SM, Sindelar WF. Evaluation of the Utility of a Radioimmunoassay for Serum Ca-19-9 Levels in Patients before and after Treatment of Carcinoma of the Pancreas. J Clinical Oncol. 1988;6:462–468. doi: 10.1200/JCO.1988.6.3.462. [DOI] [PubMed] [Google Scholar]
- 155.Forsmark CE, Lambiase L, Vogel SB. Diagnosis of Pancreatic-Cancer and Prediction of Unresectability Using the Tumor-Associated Antigen Ca19-9. Pancreas. 1994;9:731–734. doi: 10.1097/00006676-199411000-00010. [DOI] [PubMed] [Google Scholar]
- 156.Lundin J, Roberts PJ, Kuusela P, Haglund C. The Prognostic Value of Preoperative Serum Levels of Ca-19-9 and Cea in Patients with Pancreatic-Cancer. British J Cancer. 1994;69:515–519. doi: 10.1038/bjc.1994.93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Willett CG, Daly WJ, Warshaw AL. CA 19-9 is an index of response to neoadjunctive chemoradiation therapy in pancreatic cancer. Am J Surgery. 1996;172:350–352. doi: 10.1016/S0002-9610(97)89547-5. [DOI] [PubMed] [Google Scholar]
- 158.Goonetilleke KS, Siriwardena AK. Systematic review of carbohydrate antigen (CA 19-9) as a biochemical marker in the diagnosis of pancreatic cancer. Ejso. 2007;33:266–270. doi: 10.1016/j.ejso.2006.10.004. [DOI] [PubMed] [Google Scholar]
- 159.Gold DV, Modrak DE, Ying Z, Cardillo TM, Sharkey RM, Goldenberg DM. New MUC1 serum immunoassay differentiates pancreatic cancer from pancreatitis. J Clin Oncol. 2006;24:252–258. doi: 10.1200/JCO.2005.02.8282. [DOI] [PubMed] [Google Scholar]
- 160.Tumour markers in gastrointestinal cancers--EGTM recommendations. European Group on Tumour Markers. Anticancer Res. 1999;19:2811–2815. [PubMed] [Google Scholar]
- 161.Szajda SD, Waszkiewicz N, Chojnowska S, Zwierz K. Carbohydrate markers of pancreatic cancer. Biochem Soc Trans. 2011;39:340–343. doi: 10.1042/BST0390340. [DOI] [PubMed] [Google Scholar]
- 162.Duffy MJ. CA 19-9 as a marker for gastrointestinal cancers: a review. Annals of Clinical Biochemistry. 1998;35:364–370. doi: 10.1177/000456329803500304. [DOI] [PubMed] [Google Scholar]
- 163.Lamerz R. Role of tumour markers, cytogenetics. Annals of Oncology. 1999;10:145–149. [PubMed] [Google Scholar]
- 164.Monaghan PJ, Leonard MB, Neithercut WD, Raraty MGT, Sodi R. False positive carbohydrate antigen 19-9 (CA19-9) results due to a low-molecular weight interference in an apparently healthy male. Clinica Chimica Acta. 2009;406:41–44. doi: 10.1016/j.cca.2009.05.012. [DOI] [PubMed] [Google Scholar]
- 165.Kuhlmann KF, van Till JW, Boermeester MA, et al. Evaluation of matrix metalloproteinase 7 in plasma and pancreatic juice as a biomarker for pancreatic cancer. Cancer Epidemiol Biomarkers Prev. 2007;16:886–891. doi: 10.1158/1055-9965.EPI-06-0779. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Chen R, Pan S, Duan X, et al. Elevated level of anterior gradient-2 in pancreatic juice from patients with pre-malignant pancreatic neoplasia. Mol Cancer. 2010;9:149. doi: 10.1186/1476-4598-9-149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Ohuchida K, Mizumoto K, Yu J, et al. S100A6 is increased in a stepwise manner during pancreatic carcinogenesis: clinical value of expression analysis in 98 pancreatic juice samples. Cancer Epidemiol Biomarkers Prev. 2007;16:649–654. doi: 10.1158/1055-9965.EPI-06-0157. [DOI] [PubMed] [Google Scholar]
- 168.Suehara N, Mizumoto K, Niiyama H, Tanaka M. [Specific activation of telomerase in pancreatic cancer tissue and preoperative diagnosis of pancreatic cancer by telomerase in pancreatic juice] Nippon Rinsho. 1998;56:1223–1228. [PubMed] [Google Scholar]
- 169.Li J, Ma Q, Liu H, et al. Relationship between Neural Alteration and Perineural Invasion in Pancreatic Cancer Patients with Hyperglycemia. PLoS One. 2011;6:e17385. doi: 10.1371/journal.pone.0017385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Bunger S, Laubert T, Roblick UJ, Habermann JK. Serum biomarkers for improved diagnostic of pancreatic cancer: a current overview. J Cancer Research and Clinical Oncology. 2011;137:375–389. doi: 10.1007/s00432-010-0965-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Brand RE, Nolen BM, Zeh HJ, et al. Serum Biomarker Panels for the Detection of Pancreatic Cancer. Clin Cancer Res. 2011;17:805–816. doi: 10.1158/1078-0432.CCR-10-0248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Hermann PC, Huber SL, Herrler T, et al. Distinct populations of cancer stem cells determine tumor growth and metastatic activity in human pancreatic cancer. Cell Stem Cell. 2007;1:313–323. doi: 10.1016/j.stem.2007.06.002. [DOI] [PubMed] [Google Scholar]
- 173.Sergeant G, Vankelecom H, Gremeaux L, Topal B. Role of cancer stem cells in pancreatic ductal adenocarcinoma. Nat Rev Clin Oncol. 2009;6:580–586. doi: 10.1038/nrclinonc.2009.127. [DOI] [PubMed] [Google Scholar]
- 174.Bhagwandin VJ, Shay JW. Pancreatic cancer stem cells: fact or fiction? Biochim Biophys Acta. 2009;1792:248–259. doi: 10.1016/j.bbadis.2009.02.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Huang P, Wang CY, Gou SM, Wu HS, Liu T, Xiong JX. Isolation and biological analysis of tumor stem cells from pancreatic adenocarcinoma. World J Gastroenterol. 2008;14:3903–3907. doi: 10.3748/wjg.14.3903. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Toshio K, Itoi T, Sofuni A, et al. Detection of circulating tumor cells in patients with pancreatic carcinoma. Gastroenterology. 2007;132:A351-A. [Google Scholar]
- 177.Kurihara T, Itoi T, Sofuni A, et al. Detection of circulating tumor cells in patients with pancreatic cancer: a preliminary result. J Hepato-Biliary-Pancreatic Surgery. 2008;15:189–195. doi: 10.1007/s00534-007-1250-5. [DOI] [PubMed] [Google Scholar]
- 178.Klos D, Srovnal J, Lovecek M, Hajduch M, Neoral C, Havlik R. Determination of minimal residual disease in pancreatic cancer - clinical benefit. Br J Surgery. 2010;97:S69–S70. [Google Scholar]
- 179.An X, Ding PR, Li YH, et al. Elevated neutrophil to lymphocyte ratio predicts survival in advanced pancreatic cancer. Biomarkers. 2010;15:516–522. doi: 10.3109/1354750X.2010.491557. [DOI] [PubMed] [Google Scholar]
- 180.Chang P, Li Y, Li D. Micronuclei Levels in Peripheral Blood Lymphocytes as a Potential Biomarker for Pancreatic Cancer Risk. Carcinogenesis. 2010;32:210–215. doi: 10.1093/carcin/bgq247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Hoffmann K, Kerner C, Wilfert W, et al. Detection of disseminated pancreatic cells by amplification of cytokeratin-19 with quantitative RT-PCR in blood, bone marrow and peritoneal lavage of pancreatic carcinoma patients. World J Gastroenterol. 2007;13:257–263. doi: 10.3748/wjg.v13.i2.257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Helmstaedter L, Riemann JF. Pancreatic cancer--EUS and early diagnosis. Langenbecks Arch Surg. 2008;393:923–927. doi: 10.1007/s00423-007-0275-1. [DOI] [PubMed] [Google Scholar]
- 183.Levy MJ, Wiersema MJ. Endoscopic ultrasound in the diagnosis and staging of pancreatic cancer. Oncology (Williston Park) 2002;16:29–38. 43; discussion 4, 7–9, 53–6. [PubMed] [Google Scholar]
- 184.Berberat P, Friess H, Kashiwagi M, Beger HG, Buchler MW. Diagnosis and staging of pancreatic cancer by positron emission tomography. World J Surg. 1999;23:882–887. doi: 10.1007/s002689900593. [DOI] [PubMed] [Google Scholar]
- 185.Rocca R, De Angelis C, Daperno M, et al. Endoscopic ultrasound-fine needle aspiration (EUS-FNA) for pancreatic lesions: effectiveness in clinical practice. Dig Liver Dis. 2007;39:768–774. doi: 10.1016/j.dld.2007.04.013. [DOI] [PubMed] [Google Scholar]
- 186.James TA, Gibbs JF. Pancreatic cancer screening: identifying premalignant disease. Future Oncol. 2005;1:191–195. doi: 10.1517/14796694.1.2.191. [DOI] [PubMed] [Google Scholar]
- 187.Iqbal S, Stevens PD. Cholangiopancreatoscopy for targeted biopsies of the bile and pancreatic ducts. Gastrointest Endosc Clin N Am. 2009;19:567–577. doi: 10.1016/j.giec.2009.06.005. [DOI] [PubMed] [Google Scholar]
- 188.Fleming JB, Gonzalez RJ, Petzel MQ, et al. Influence of obesity on cancer-related outcomes after pancreatectomy to treat pancreatic adenocarcinoma. Arch Surg. 2009;144:216–221. doi: 10.1001/archsurg.2008.580. [DOI] [PubMed] [Google Scholar]
- 189.McWilliams RR, Matsumoto ME, Burch PA, et al. Obesity Adversely Affects Survival in Pancreatic Cancer Patients. Cancer. 2010;116:5054–5062. doi: 10.1002/cncr.25465. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Li DH, Morris JS, Liu J, et al. Body Mass Index and Risk, Age of Onset, and Survival in Patients With Pancreatic Cancer. Jama-J the Am Medical Association. 2009;301:2553–2562. doi: 10.1001/jama.2009.886. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Jiao L, Hassan MM, Bondy ML, et al. XRCC2 and XRCC3 gene polymorphism and risk of pancreatic cancer. Am J Gastroenterol. 2008;103:360–367. doi: 10.1111/j.1572-0241.2007.01615.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Iodice S, Gandini S, Maisonneuve P, Lowenfels AB. Tobacco and the risk of pancreatic cancer: a review and meta-analysis. Langenbecks Arch Surg. 2008;393:535–545. doi: 10.1007/s00423-007-0266-2. [DOI] [PubMed] [Google Scholar]
- 193.Li D, Tang H, Hassan MM, Holly EA, Bracci PM, Silverman DT. Diabetes and risk of pancreatic cancer: a pooled analysis of three large case-control studies. Cancer Causes Control. 2011;22:189–197. doi: 10.1007/s10552-010-9686-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Yalniz M, Pour PM. Diabetes mellitus: a risk factor for pancreatic cancer? Langenbecks Arch Surg. 2005;390:66–72. doi: 10.1007/s00423-004-0469-8. [DOI] [PubMed] [Google Scholar]
- 195.Whitcomb DC, Pogue-Geile K. Pancreatitis as a risk for pancreatic cancer. Gastroenterol Clinics North Am. 2002;31:663-+. doi: 10.1016/s0889-8553(02)00004-3. [DOI] [PubMed] [Google Scholar]
- 196.Raimondi S, Lowenfels AB, Morselli-Labate AM, Maisonneuve P, Pezzilli R. Pancreatic cancer in chronic pancreatitis; aetiology, incidence, and early detection. Best Practice Res Clinical Gastroenterol. 2010;24:349–358. doi: 10.1016/j.bpg.2010.02.007. [DOI] [PubMed] [Google Scholar]
- 197.Latchford A, Greenhalf W, Vitone LJ, Neoptolemos JP, Lancaster GA, Phillips RK. Peutz-Jeghers syndrome and screening for pancreatic cancer. Br J Surg. 2006;93:1446–1455. doi: 10.1002/bjs.5609. [DOI] [PubMed] [Google Scholar]
- 198.Klump B, Hsieh CJ, Nehls O, et al. Methylation status of p14ARF and p16INK4a as detected in pancreatic secretions. Br J Cancer. 2003;88:217–222. doi: 10.1038/sj.bjc.6600734. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Wolpin BM, Chan AT, Hartge P, et al. ABO Blood Group and the Risk of Pancreatic Cancer. J National Cancer Institute. 2009;101:424–431. doi: 10.1093/jnci/djp020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Wolpin BM, Kraft P, Gross M, et al. Pancreatic Cancer Risk and ABO Blood Group Alleles: Results from the Pancreatic Cancer Cohort Consortium. Cancer Res. 2010;70:1015–1023. doi: 10.1158/0008-5472.CAN-09-2993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Greer JB, Yazer MH, Raval JS, Barmada MM, Brand RE, Whitcomb DC. Significant association between ABO blood group and pancreatic cancer. World J Gastroenterol. 2010;16:5588–5591. doi: 10.3748/wjg.v16.i44.5588. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Seibert V, Ebert MP, Buschmann T. Advances in clinical cancer proteomics: SELDI-ToF-mass spectrometry and biomarker discovery. Brief Funct Genomic Proteomic. 2005;4:16–26. doi: 10.1093/bfgp/4.1.16. [DOI] [PubMed] [Google Scholar]
- 203.Maurya P, Meleady P, Dowling P, Clynes M. Proteomic approaches for serum biomarker discovery in cancer. Anticancer Res. 2007;27:1247–1255. [PubMed] [Google Scholar]
- 204.Chen R, Pan S, Brentnall TA, Aebersold R. Proteomic profiling of pancreatic cancer for biomarker discovery. Mol Cell Proteomics. 2005;4:523–533. doi: 10.1074/mcp.R500004-MCP200. [DOI] [PubMed] [Google Scholar]
- 205.Mauri P, Scarpa A, Nascimbeni AC, et al. Identification of proteins released by pancreatic cancer cells by multidimensional protein identification technology: a strategy for identification of novel cancer markers. FASEB J. 2005;19:1125–1127. doi: 10.1096/fj.04-3000fje. [DOI] [PubMed] [Google Scholar]
- 206.Rong YF, Jin DY, Hou CR, et al. Proteomics analysis of serum protein profiling in pancreatic cancer patients by DIGE: up-regulation of mannose-binding lectin 2 and myosin light chain kinase 2. Bmc Gastroenterol. 2010;10 doi: 10.1186/1471-230X-10-68. -. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207.Chen R, Brentnall TA, Pan S, et al. Quantitative proteomics analysis reveals that proteins differentially expressed in chronic pancreatitis are also frequently involved in pancreatic cancer. Mol Cellular Proteomics. 2007;6:1331–1342. doi: 10.1074/mcp.M700072-MCP200. [DOI] [PubMed] [Google Scholar]
- 208.Chen R, Yi EC, Donohoe S, et al. Pancreatic cancer proteome: the proteins that underlie invasion, metastasis, and immunologic escape. Gastroenterology. 2005;129:1187–1197. doi: 10.1053/j.gastro.2005.08.001. [DOI] [PubMed] [Google Scholar]
- 209.Ehmann M, Felix K, Hartmann D, et al. Identification of potential markers for the detection of pancreatic cancer through comparative serum protein expression profiling. Pancreas. 2007;34:205–214. doi: 10.1097/01.mpa.0000250128.57026.b2. [DOI] [PubMed] [Google Scholar]
- 210.Ransohoff DF. Bias as a threat to the validity of cancer molecular-marker research. Nat Rev Cancer. 2005;5:142–149. doi: 10.1038/nrc1550. [DOI] [PubMed] [Google Scholar]
- 211.Kruger M, Moser M, Ussar S, et al. SILAC mouse for quantitative proteomics uncovers kindlin-3 as an essential factor for red blood cell function. Cell. 2008;134:353–364. doi: 10.1016/j.cell.2008.05.033. [DOI] [PubMed] [Google Scholar]
- 212.McDonald WH, Yates JR., 3rd Shotgun proteomics and biomarker discovery. Dis Markers. 2002;18:99–105. doi: 10.1155/2002/505397. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213.Chen J, Li D, Killary AM, et al. Polymorphisms of p16, p27, p73, and MDM2 modulate response and survival of pancreatic cancer patients treated with preoperative chemoradiation. Ann Surg Oncol. 2009;16:431–439. doi: 10.1245/s10434-008-0220-8. [DOI] [PubMed] [Google Scholar]
- 214.Tanase CP, Dima S, Mihai M, et al. Caveolin-1 overexpression correlates with tumour progression markers in pancreatic ductal adenocarcinoma. J Molecular Histol. 2009;40:23–29. doi: 10.1007/s10735-008-9209-7. [DOI] [PubMed] [Google Scholar]
- 215.Giovannetti E, Del Tacca M, Mey V, et al. Transcription analysis of human equilibrative nucleoside transporter-1 predicts survival in pancreas cancer patients treated with gemcitabine. Cancer Res. 2006;66:3928–3935. doi: 10.1158/0008-5472.CAN-05-4203. [DOI] [PubMed] [Google Scholar]
- 216.Szafranska AE, Doleshal M, Edmunds HS, et al. Analysis of microRNAs in pancreatic fine-needle aspirates can classify benign and malignant tissues. Clin Chem. 2008;54:1716–1724. doi: 10.1373/clinchem.2008.109603. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217.Chen BZ, Yu SL, Singh S, et al. Identification of microRNAs expressed highly in pancreatic islet-like cell clusters differentiated from human embryonic stem cells. Cell Biol Int. 2011;35:29–37. doi: 10.1042/CBI20090081. [DOI] [PubMed] [Google Scholar]
- 218.Greither T, Grochola LF, Udelnow A, Lautenschlager C, Wurl P, Taubert H. Elevated expression of microRNAs 155, 203, 210 and 222 in pancreatic tumors is associated with poorer survival. Int J Cancer. 2010;126:73–80. doi: 10.1002/ijc.24687. [DOI] [PubMed] [Google Scholar]
- 219.Li YW, VandenBoom TG, Kong DJ, et al. Up-regulation of miR-200 and let-7 by Natural Agents Leads to the Reversal of Epithelial-to-Mesenchymal Transition in Gemcitabine-Resistant Pancreatic Cancer Cells. Cancer Res. 2009;69:6704–6712. doi: 10.1158/0008-5472.CAN-09-1298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220.Stratford JK, Bentrem DJ, Anderson JM, et al. A Six-Gene Signature Predicts Survival of Patients with Localized Pancreatic Ductal Adenocarcinoma. Plos Medicine. 2010;7 doi: 10.1371/journal.pmed.1000307. e1000307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221.Ohuchida K, Mizumoto K, Ishikawa N, et al. The role of S100A6 in pancreatic cancer development and its clinical implication as a diagnostic marker and therapeutic target. Clin Cancer Res. 2005;11:7785–7793. doi: 10.1158/1078-0432.CCR-05-0714. [DOI] [PubMed] [Google Scholar]
- 222.Tian M, Cui YZ, Song GH, et al. Proteomic analysis identifies MMP-9, DJ-1 and A1BG as overexpressed proteins in pancreatic juice from pancreatic ductal adenocarcinoma patients. BMC Cancer. 2008;8:241. doi: 10.1186/1471-2407-8-241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223.Sadakari Y, Ohtsuka T, Ohuchida K, et al. MicroRNA expression analyses in preoperative pancreatic juice samples of pancreatic ductal adenocarcinoma. JOP. 2010;11:587–592. [PubMed] [Google Scholar]
- 224.Habbe N, Koorstra JBM, Mendell JT, et al. MicroRNA miR-155 is a biomarker of early pancreatic neoplasia. Cancer Biol Therapy. 2009;8:340–346. doi: 10.4161/cbt.8.4.7338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225.Ohuchida K, Mizumoto K, Ohhashi S, et al. Twist, a novel oncogene, is upregulated in pancreatic cancer: clinical implication of Twist expression in pancreatic juice. Int J Cancer. 2007;120:1634–1640. doi: 10.1002/ijc.22295. [DOI] [PubMed] [Google Scholar]
- 226.Nakashima A, Murakami Y, Uemura K, et al. Usefulness of Human Telomerase Reverse Transcriptase in Pancreatic Juice as a Biomarker of Pancreatic Malignancy. Pancreas. 2009;38:527–533. doi: 10.1097/MPA.0b013e3181a16d28. [DOI] [PubMed] [Google Scholar]
- 227.Satake K. Diagnosis of pancreatic cancer. Serological markers. Int J Pancreatol. 1991;9:93–98. doi: 10.1007/BF02925584. [DOI] [PubMed] [Google Scholar]
- 228.Cappelli G, Paladini S, D'Agata A. Tumor markers in the diagnosis of pancreatic cancer. Tumori. 1999;85:S19–S21. [PubMed] [Google Scholar]
- 229.Harmenberg U, Wahren B, Wiechel KL. Tumor markers carbohydrate antigens CA 19-9 and CA-50 and carcinoembryonic antigen in pancreatic cancer and benign diseases of the pancreatobiliary tract. Cancer Res. 1988;48:1985–1988. [PubMed] [Google Scholar]
- 230.Liao WC, Wu MS, Wang HP, Tien YW, Lin JT. Serum Heat Shock Protein 27 Is Increased in Chronic Pancreatitis and Pancreatic Carcinoma. Pancreas. 2009;38:422–426. doi: 10.1097/MPA.0b013e318198281d. [DOI] [PubMed] [Google Scholar]
- 231.Karna E, Surazynski A, Orlowski K, et al. Serum and tissue level of insulin-like growth factor-I (IGF-I) and IGF-I binding proteins as an index of pancreatitis and pancreatic cancer. Int J Exp Pathol. 2002;83:239–245. doi: 10.1046/j.1365-2613.2002.00237.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232.Sun ZL, Zhu Y, Wang FQ, et al. Serum proteomic-based analysis of pancreatic carcinoma for the identification of potential cancer biomarkers. Biochim Biophys Acta. 2007;1774:764–771. doi: 10.1016/j.bbapap.2007.04.001. [DOI] [PubMed] [Google Scholar]
- 233.Abbasciano V, Graziano L, Guerra S, et al. Coagulation disorders and tumor markers in the diagnosis of pancreatic cancer. Oncology. 1991;48:377–382. doi: 10.1159/000226963. [DOI] [PubMed] [Google Scholar]
- 234.Firpo MA, Gay DZ, Granger SR, et al. Improved Diagnosis of Pancreatic Adenocarcinoma Using Haptoglobin and Serum Amyloid A in a Panel Screen. World J Surgery. 2009;33:716–722. doi: 10.1007/s00268-008-9853-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235.Bloomston M, Zhou JX, Rosemurgy AS, Frankel W, Muro-Cacho CA, Yeatman TJ. Fibrinogen gamma overexpression in pancreatic cancer identified by large-scale proteomic analysis of serum samples. Cancer Res. 2006;66:2592–2599. doi: 10.1158/0008-5472.CAN-05-3659. [DOI] [PubMed] [Google Scholar]
- 236.Ni XG, Bai XF, Mao YL, et al. The clinical value of serum CEA, CA19-9, and CA242 in the diagnosis and prognosis of pancreatic cancer. Eur J Surg Oncol. 2005;31:164–169. doi: 10.1016/j.ejso.2004.09.007. [DOI] [PubMed] [Google Scholar]
- 237.Dive C, Smith RA, Garner E, et al. Considerations for the use of plasma cytokeratin 18 as a biomarker in pancreatic cancer. British J Cancer. 2010;102:577–582. doi: 10.1038/sj.bjc.6605494. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 238.Zhou J, Hu L, Yu Z, et al. Marker Expression in Circulating Cancer Cells of Pancreatic Cancer Patients. J Surg Res. 2011;171:631–636. doi: 10.1016/j.jss.2010.05.007. [DOI] [PubMed] [Google Scholar]
- 239.Sabbaghian MS, Rothberger G, Alongi AP, et al. Levels of Elevated Circulating Endothelial Cell Decline after Tumor Resection in Patients with Pancreatic Ductal Adenocarcinoma. Anticancer Res. 2010;30:2911–2917. [PubMed] [Google Scholar]
- 240.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;115:1632–1636. [PubMed] [Google Scholar]
- 241.Jiao L, Zhu J, Hassan MM, Evans DB, Abbruzzese JL, Li D. K-ras mutation and p16 and preproenkephalin promoter hypermethylation in plasma DNA of pancreatic cancer patients: in relation to cigarette smoking. Pancreas. 2007;34:55–62. doi: 10.1097/01.mpa.0000246665.68869.d4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242.Salek C, Minarikova P, Benesova L, et al. Mutation status of K-ras, p53 and allelic losses at 9p and 18q are not prognostic markers in patients with pancreatic cancer. Anticancer Res. 2009;29:1803–1810. [PubMed] [Google Scholar]
- 243.Ghaneh P, Kawesha A, Evans JD, Neoptolemos JP. Molecular prognostic markers in pancreatic cancer. J Hepatobiliary Pancreat Surg. 2002;9:1–11. doi: 10.1007/s005340200000. [DOI] [PubMed] [Google Scholar]
- 244.Huang L, Goodrow TL, Zhang SY, Klein-Szanto AJ, Chang H, Ruggeri BA. Deletion and mutation analyses of the P16/MTS-1 tumor suppressor gene in human ductal pancreatic cancer reveals a higher frequency of abnormalities in tumor-derived cell lines than in primary ductal adenocarcinomas. Cancer Res. 1996;56:1137–1141. [PubMed] [Google Scholar]
- 245.Vasen HF, Wasser M, van Mil A, et al. Magnetic Resonance Imaging Surveillance Detects Early-Stage Pancreatic Cancer in Carriers of a p16-Leiden Mutation. Gastroenterology. 2011;140:850–856. doi: 10.1053/j.gastro.2010.11.048. [DOI] [PubMed] [Google Scholar]
- 246.Chen J, Li D, Wei C, et al. Aurora-A and p16 polymorphisms contribute to an earlier age at diagnosis of pancreatic cancer in Caucasians. Clin Cancer Res. 2007;13:3100–3104. doi: 10.1158/1078-0432.CCR-06-2319. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 247.Barbera VM, Martin M, Marinoso L, et al. The 18q21 region in colorectal and pancreatic cancer: independent loss of DCC and DPC4 expression. Biochimica Et Biophysica Acta-Molecular Basis of Disease. 2000;1502:283–296. doi: 10.1016/s0925-4439(00)00054-5. [DOI] [PubMed] [Google Scholar]
- 248.Parsi MA, Li A, Li CP, Goggins M. DNA methylation alterations in endoscopic retrograde cholangiopancreatography brush samples of patients with suspected pancreaticobiliary disease. Clin Gastroenterol Hepatol. 2008;6:1270–1278. doi: 10.1016/j.cgh.2008.07.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 249.Wang J, Chen J, Chang P, et al. MicroRNAs in plasma of pancreatic ductal adenocarcinoma patients as novel blood-based biomarkers of disease. Cancer Prev Res (Phila) 2009;2:807–813. doi: 10.1158/1940-6207.CAPR-09-0094. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 250.Lee EJ, Gusev Y, Jiang J, et al. Expression profiling identifies microRNA signature in pancreatic cancer. Int J Cancer. 2007;120:1046–1054. doi: 10.1002/ijc.22394. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 251.Roldo C, Missiaglia E, Hagan JP, et al. MicroRNA expression abnormalities in pancreatic endocrine and acinar tumors are associated with distinctive pathologic features and clinical behavior. J Clin Oncol. 2006;24:4677–4684. doi: 10.1200/JCO.2005.05.5194. [DOI] [PubMed] [Google Scholar]
- 252.Bhatti I, Lee A, James V, et al. Knockdown of microRNA-21 Inhibits Proliferation and Increases Cell Death by Targeting Programmed Cell Death 4 (PDCD4) in Pancreatic Ductal Adenocarcinoma. J Gastrointest Surg. 2010;15:199–208. doi: 10.1007/s11605-010-1381-x. [DOI] [PubMed] [Google Scholar]
- 253.Haglund C, Lundin J, Kuusela P, Roberts PJ. CA 242, a new tumour marker for pancreatic cancer: a comparison with CA 19-9, CA 50 and CEA. Br J Cancer. 1994;70:487–492. doi: 10.1038/bjc.1994.332. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 254.Navaglia F, Fogar P, Basso D, et al. Pancreatic cancer biomarkers discovery by surface-enhanced laser desorption and ionization time-of-flight mass spectrometry. Clin Chem Lab Med. 2009;47:713–723. doi: 10.1515/CCLM.2009.158. [DOI] [PubMed] [Google Scholar]
- 255.Safi F, Roscher R, Beger HG. Tumor markers in pancreatic cancer. Sensitivity and specificity of CA 19-9. Hepatogastroenterology. 1989;36:419–423. [PubMed] [Google Scholar]
- 256.Masson P, Palsson B, Andren-Sandberg A. Cancer-associated tumour markers CA 19-9 and CA-50 in patients with pancreatic cancer with special reference to the Lewis blood cell status. Br J Cancer. 1990;62:118–121. doi: 10.1038/bjc.1990.241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 257.Ramachandran V, Arumugam T, Wang HM, Logsdon CD. Anterior gradient 2 is expressed and secreted during the development of pancreatic cancer and promotes cancer cell survival. Cancer Res. 2008;68:7811–7818. doi: 10.1158/0008-5472.CAN-08-1320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 258.Allum WH, Stokes HJ, Macdonald F, Fielding JW. Demonstration of carcinoembryonic antigen (CEA) expression in normal, chronically inflamed, and malignant pancreatic tissue by immunohistochemistry. J Clin Pathol. 1986;39:610–614. doi: 10.1136/jcp.39.6.610. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 259.Haas M, Laubender RP, Stieber P, et al. Prognostic relevance of CA 19-9 CEA, CRP, and LDH kinetics in patients treated with palliative second-line therapy for advanced pancreatic cancer. Tumor Biology. 2010;31:351–357. doi: 10.1007/s13277-010-0044-6. [DOI] [PubMed] [Google Scholar]
- 260.Duraker N, Hot S, Polat Y, Hobek A, Gencler N, Urhan N. CEA, CA 19-9, and CA 125 in the differential diagnosis of benign and malignant pancreatic diseases with or without jaundice. J Surgical Oncol. 2007;95:142–147. doi: 10.1002/jso.20604. [DOI] [PubMed] [Google Scholar]
- 261.Macdonald F, Downing R, Allum WH. Expression of Ca125 in Pancreatic-Carcinoma and Chronic-Pancreatitis. Br J Cancer. 1988;58:505–506. doi: 10.1038/bjc.1988.251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 262.Hasholzner U, Stieber P, Reiter W, Zimmermann A, Hofmann K, Schalhorn A. CA 242 in comparison with established tumour markers in colorectal, pancreatic and lung cancer. Anticancer Res. 1999;19:2477–2480. [PubMed] [Google Scholar]
- 263.Pasanen PA, Eskelinen M, Partanen K, Pikkarainen P, Penttila I, Alhava E. Receiver operating characteristic (ROC) curve analysis of the tumour markers CEA, CA 50 and CA 242 in pancreatic cancer, results from a prospective study. Br J Cancer. 1993;67:852–855. doi: 10.1038/bjc.1993.156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 264.Lundin J, Roberts PJ, Kuusela P, Haglund C. Prognostic significance of serum CA 242 in pancreatic cancer. A comparison with CA 19-9. Anticancer Res. 1995;15:2181–2186. [PubMed] [Google Scholar]
- 265.Kim J-E, Lee KT, Lee JK, Paik SW, Rhee JC, Choi KW. Clinical usefulness of carbohydrate antigen 19-9 as a screening test for pancreatic cancer in an asymptomatic population. J Gastroenterology and Hepatology. 2004;19:182–186. doi: 10.1111/j.1440-1746.2004.03219.x. [DOI] [PubMed] [Google Scholar]
- 266.Lee MX, Saif MW. Screening for early pancreatic ductal adenocarcinoma: an urgent call! JOP. 2009;10:104–108. [PubMed] [Google Scholar]
- 267.Cwik G, Wallner G, Skoczylas T, Ciechanski A, Zinkiewicz K. Cancer antigens 19-9 and 125 in the differential diagnosis of pancreatic mass lesions. Arch Surg. 2006;141:968–973. doi: 10.1001/archsurg.141.10.968. discussion 74. [DOI] [PubMed] [Google Scholar]
- 268.Banfi G, Zerbi A, Pastori S, Parolini D, Dicarlo V, Bonini P. Behavior of Tumor-Markers Ca19.9, Ca195, Cam43, Ca242, and Tps in the Diagnosis and Follow-up of Pancreatic-Cancer. Clinical Chem. 1993;39:420–423. [PubMed] [Google Scholar]
- 269.Jiang XT, Tao HQ, Zou SC. Detection of serum tumor markers in the diagnosis and treatment of patients with pancreatic cancer. Hepatobiliary Pancreat Dis Int. 2004;3:464–468. [PubMed] [Google Scholar]

