Topic Highlight Open Access
Copyright ©2014 Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Aug 28, 2014; 20(32): 11241-11248
Published online Aug 28, 2014. doi: 10.3748/wjg.v20.i32.11241
Advances in pancreatic cancer research: Moving towards early detection
Xiang-Yi He, Yao-Zong Yuan, Department of Gastroenterology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
Author contributions: He XY collected all the references and drafted the article; Yuan YZ chose the topic, organized the structure and revised the article.
Supported by Youth Foundation of Shanghai Municipal Health Bureau (in part), No. 122; Young Teachers Program of Shanghai University, National Natural Science Foundation of China, No. 81302087; and Natural Science Foundation of Shanghai, No. 13ZR1457400
Correspondence to: Yao-Zong Yuan, MD, PhD, Department of Gastroenterology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijin Er Road, Shanghai 200025, China. yyz28@medmail.com.cn
Telephone: +86-21-64150773 Fax: +86-21-64150773
Received: October 27, 2013
Revised: March 2, 2014
Accepted: June 2, 2014
Published online: August 28, 2014

Abstract

Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal forms of cancer. Substantial progress has been made in the understanding of the biology of pancreatic cancer, and advances in patient management have been significant. However, most patients (nearly 80%) who present with locally advanced or metastatic disease have an extremely poor prognosis. Survival is better for those with malignant disease localized to the pancreas, because surgical resection at present offers the only chance of cure. Therefore, the early detection of pancreatic cancer may benefit patients with PDAC. However, its low rate of incidence and the limitations of current screening strategies make early detection difficult. Recent advances in the understanding of the pathogenesis of PDAC suggest that it is possible to detect PDAC in early stages and even identify precursor lesions. The presence of new-onset diabetes mellitus in the early phase of pancreatic cancer may provide clues for its early diagnosis. Advances in the identification of novel circulating biomarkers including serological signatures, autoantibodies, epigenetic markers, circulating tumor cells and microRNAs suggest that they can be used as potential tools for the screening of precursors and early stage PDAC in the future. However, proper screening strategies based on effective screening methodologies need to be tested for clinical application.

Key Words: Pancreatic ductal adenocarcinoma, Early detection, Diabetes mellitus, Pancreatic cancer

Core tip: Because pancreatic cancer is usually detected at an advanced stage and there is a lack of treatment strategies for advanced disease, it remains one of the most lethal solid tumors. Genetic and epigenetic alterations, miRNAs and tumor microenvironment promote the development of pancreatic cancer from precursor lesions to localized disease and further to metastatic disease in several years. An effective screening strategy for pancreatic cancer is therefore needed. New-onset diabetes mellitus associated with pancreatic cancer and recently identified novel circulating biomarkers should be explored as potential screening markers.



INTRODUCTION

Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal forms of cancer. It is the 13th most commonly diagnosed cancer worldwide[1], and the eighth leading cause of cancer death. No early detection tests are available and most patients with localized disease have no recognizable symptoms or signs; as a result, most patients (80%-85%) are not diagnosed until late in the disease, when the cancer has metastasized to other organs[2,3]. Survival is better for those with malignant disease localized to the pancreas, because at present, surgical resection offers the only chance of cure. Because pancreatic cancer responds poorly to radiation and chemotherapy, and so far most of the targeted therapy agents have failed to show a substantial benefit, PDAC in the advanced stage is associated with an extremely poor prognosis[2,3]. Therefore, the early detection of PDAC has gained increasing attention with the aim of improving the outcomes of patients with this disease. Recent studies have improved our understanding of the pathogenesis of PDAC, the relationship between diabetes mellitus (DM) and PDAC, and the role of circulating biomarkers. The present review will discuss the possibility of using these data to detect PDAC in its early stage and propose future research directions.

RECENT ADVANCES IN THE UNDERSTANDING OF THE PATHOGENESIS OF PDAC

Despite the short lifespan of patients diagnosed with PDAC, the disease usually develops over a long period of time. Based on a genetic evolutionary model[4], it is estimated that 10-30 years are required from the initiating mutation until the patient’s death. In this model, there are three critical periods in the genetic evolution of the disease: T1 is related to the formation of precursor lesions such as pancreatic intraepithelial neoplasia (PanIN) and lasts until the infiltrating carcinoma is first formed; T2 describes the period from that time until a metastatic subclone develops within the primary carcinoma; and T3 is the period of metastatic dissemination of that subclone until the patient’s death. A conservative estimate of 11.7, 6.8 and 2.7 years per interval, respectively, has been reported[4].

The development of PDAC involves multiple steps. It may originate from four distinct precursors, i.e., mucinous cystic neoplasm (MCN), intraductal papillary mucinous neoplasm (IPMN), PanIN, and the newly added intraductal tubular papillary neoplasms (ITPNs)[5]. All four harbor varying degrees of dysplasia and stepwise accumulation of genetic alterations, suggesting progression of these lesions from benign toward malignant neoplasms. MCNs have a characteristic ovarian-type stroma. Approximately one-third of MCNs are associated with invasive carcinoma of a ductal phenotype. IPMNs are recently established clinical entities with characteristic features of mucin hypersecretion and duct dilatation. Some IPMNs are associated with invasive carcinoma and IPMNs are recognized as precursors to pancreatic cancer. ITPNs are rare premalignant tumours, with a concomitant invasive component, and were included for the first time in the 2010 World Health Organization classification[5]. PanINs are microscopic proliferative lesions arising from any part of the pancreatic duct system. Low grade PanINs are commonly found in the pancreatic ducts of older individuals, whereas high grade PanINs, previously called carcinoma in situ/severe ductal dysplasia, may eventually give rise to invasive pancreatic cancer. PanINs are divided into four categories based on the degree of dysplasia (1A, 1B, 2, and 3). Appropriate clinical management is critical for patients with MCNs, IPMNs and PanINs. The progression from PanIN to invasive PDAC has been intensively studied and reviewed elsewhere[6]. The established model describes the stepwise progression of PanINs to PDAC through the accumulation of several important genetic aberrations. KRAS mutation may be the first step driving this progression, and it is detected in approximately 99% of PanIN-1 lesions[7]. In addition, to overcome oncogene-induced senescence, loss of function of CDKN2A, and genetic inactivation of TP53, SMAD4, and BRCA2 are also required for the development of PDAC[8]. Further investigation of these precursor lesions is expected to reduce the mortality from pancreatic cancer.

The development of PDAC involves multiple genes. In 2008, detailed, global, genomic analyses found that a large number of genetic alterations (an average of 63) affect a core set of 12 signaling pathways and processes that are each genetically altered in 67%-100% of cases of pancreatic cancer[9]. However, the pathway components that are altered in any individual tumor vary widely. The 12 core pathways[9] are apoptosis, DNA damage control, regulation of G1/S phase transition, Hedgehog signaling, homophilic cell adhesion, integrin signaling, c-Jun N-terminal kinase signaling, KRAS signaling, regulation of invasion, small GTPase-dependent signaling (other than KRAS), TGF-β signaling and Wnt/Notch signaling. In addition to the core pathways described above, more recent data indicate that chromatin regulation[10] and axon guidance[10,11] are additional cellular processes that play a crucial role in pancreatic cancer. The pathways involved in each patient often vary. This finding may help account for the heterogeneous nature of tumors and offer insights into why agents targeting a specific gene in a pathway rarely result in a therapeutic advantage in more than a minor percentage of patients. Therefore, identification of the core players of each pathway and the factors connecting them is important. In addition to whole genomic alterations, numerous investigators have focused on particular PDAC-associated genes through laboratory and patient studies. Our group demonstrated that Periostin[12], X chromosome-linked inhibitor of apoptosis (XIAP)-associated, factor 1 (XAF1, a novel XIAP modulator)[13], angiotensin-converting enzyme 2[14], bone morphogenetic protein-2[15], eEF1A2[16], L1 cell adhesion molecule (L1-CAM)[17], and DJ-1[18] are associated with PDAC proliferation, apoptosis, invasion and/or progression.

MicroRNAs (miRNAs) are a highly conserved family of 18-24-nucleotide RNA molecules that regulate the stability or translational efficiency of complementary target mRNAs. More than 20 miRNAs involved in pancreatic adenocarcinoma biology have been identified and shown to affect tumor growth, metastatic potential, and chemosensitivity[19]. Combinations of miRNAs can be used to differentiate between pancreatic adenocarcinoma and other pancreatic pathologies, as well as to assess prognosis. Manipulations of miRNAs can decrease the rate of growth or reinstall chemosensitivity to certain chemotherapeutic agents. The most extensively studied miRNA, as far as pancreatic cancer is concerned, is miR-21, which has been associated with cell proliferation, metastatic ability, decreased gemcitabine sensitivity, and poor overall survival (OS)[20-23]. Other PDAC associated miRNAs were discussed elsewhere. Recently, several other miRNAs (miR-196a[24,25], 130b[26], 92a[27], 198[28], 221[29,30], 23b[31], 29a[32,33]) were shown to play an important role in PDAC.

Tumors are complex tissues in which mutant cancer cells have conscripted and subverted normal cell types to serve as active collaborators in their neoplastic agenda. Recent studies have shown that PDAC is one of the most stroma-rich cancers. The tumor microenvironment surrounds most of the tumor mass and consists of a dynamic assortment of extracellular matrix components and non-neoplastic cells including fibroblastic, vascular and immune cells. Recent work has revealed that the PDAC stroma supports tumor growth and promotes metastasis, and simultaneously serves as a physical barrier to drug delivery[34]. Pancreatic stellate cells (PSCs) identified in 1998, have the ability to trans-differentiate from a “quiescent” retinoid/lipid storing phenotype in the normal pancreas to an “activated” a-smooth muscle actin producing myofibroblastic phenotype[35]. The activated PSCs produce the extracellular matrix proteins that comprise the pancreatic tumor stroma, to facilitate pancreatic cancer development[35]. Sonic hedgehog signaling has been shown to be restricted to the stromal compartment and enhance the desmoplastic reaction[36]. Findings[36] suggest that increased HIF-1α produced by hypoxic tumors triggers the desmoplastic reaction in pancreatic cancer.

The genetic evolutionary model of PDAC suggests a detection window of several years (T1 + T2) for this disease. The visualization of PDAC precursor lesions using currently available imaging methods is limited; therefore, the detection of precursors is difficult. Improving our understanding of the mechanisms of precursors and PDAC development may help identify tumor biomarkers for this disease.

DM AND PDAC

Because of the low incidence of PDAC in the general population, population-based screening is not recommended. It is more practical to screen individuals at increased risk for PC based on their family history or identifiable genetic predisposition, or patients with diseases known to increase the risk of pancreatic cancer, such as chronic pancreatitis and type II DM. Patients with a family history of pancreatic cancer or mutation carriers (germline mutations in the BRCA2, PALB2, p16, STK11, ATM, PRSS1 genes and Lynch syndrome or Peutz-Jeghers syndrome) should be screened for pancreatic cancer according to the recommendations of the International Cancer of the Pancreas Screening consortium[37]. Here, we will discuss the relationship between DM and PDAC and the possibility of using new-onset DM as a marker for the detection of PDAC.

Increasing evidence suggests that DM is related to PDAC. It is now recognized that although long-standing diabetes is an etiological factor for pancreatic cancer, new-onset diabetes is its manifestation[38-40]. Epidemiological investigations have found that long-term type 2 DM is associated with a 1.5-fold to 2.0-fold increase in the risk of pancreatic cancer[40]. The evidence suggesting that new-onset diabetes is the manifestation of PDAC, or in other words, caused by PDAC, is that: (1) new-onset diabetes is associated with a high prevalence of PDAC; (2) diabetes associated with pancreatic cancer is predominantly new-onset; (3) pancreatic cancer resection ameliorates diabetes; and (4) experimental data. A meta-analysis[41] conducted in 2005 that included 17 case-control and 19 cohort and nested case-control studies published between 1996 and 2005 demonstrated that the combined age-adjusted and sex-adjusted odds ratio (OR) for pancreatic cancer associated with diabetes was 1.8 (95%CI: 1.7-1.9) and was lower still (OR = 1.5) in patients with a ≥ 5 year history of diabetes. In a pooled analysis[42] of 2192 patients with pancreatic cancer and 5113 cancer-free controls in three large case-control studies conducted in the United States, diabetes was associated with a 1.8-fold increase in the risk of pancreatic cancer (95%CI: 1.5-2.1). Risk estimates decreased as the number of years with diabetes increased[42]. A study from our group[43] included 1458 patients with PDAC and 1528 age-, sex- and sociodemographic matched controls and showed that compared with controls, patients with long-standing diabetes (≥ 2-year duration) had a moderately increased risk of PDAC, with an OR (95%CI) of 2.11 (1.51-2.94). Interestingly, a significantly higher risk was observed among cases with new-onset DM (< 2-year duration), with an OR of 4.43 (3.44-5.72) compared to controls without DM. On the other hand, the reported prevalence of DM in pancreatic cancer varies from 23% to 75%, with the majority being new-onset[44]. Our data also showed that 44.7% of PDAC patients harbor DM, and almost 2/3 are new-onset[45]. Data from our group and other groups showed that resolution of DM after pancreatic resection occurs in 41%-57% of PDAC patients with new-onset DM, in contrast to most patients with longstanding DM, who remain diabetic postoperatively[44-46]. Recognition of new-onset diabetes as an early manifestation of pancreatic cancer could improve the detection of asymptomatic, early-stage pancreatic cancer[38].

The impact of DM on the long-term outcomes of patients with PDAC has also been intensively studied recently, although the results are controversial. In patients with stage I-IV pancreatic cancers, DM does not confer a worse prognosis; in fact, diabetics have a statistically significantly superior median survival[47]. A retrospective study of 344 patients who underwent surgical resection of pancreatic cancers showed that perineural invasion was significantly more common in diabetics, with a poor OS[47]. A multi-institutional retrospective study[48] reported a shorter OS and disease-free survival (DFS) in patients with preoperative DM. By stratifying DM into different groups (long-term/new-onset pre-surgical diabetes, resolved/unresolved post-surgical diabetes), we found that the heterogynous DM groups have different impacts on PDAC outcomes: longstanding DM is predictive of poor postoperative DFS and OS, whereas postoperatively resolved new-onset DM is associated with longer DFS and OS[45].

There are several possible mechanisms to explain the effect of diabetes on promoting PDAC progression, including the cellular proliferative effects of hyperglycemia, hyperinsulinemia, abnormalities in insulin/IGF receptor pathways, oxidative stress and inflammatory responses[40]. A prospective nested case-control study that included 449 case patients and 982 control subjects showed that the highest and the lowest quintiles of HbA1c, insulin, and proinsulin were associated with an increased risk for pancreatic cancer. However, in mutually adjusted models, only circulating markers of peripheral insulin resistance (proinsulin), rather than hyperglycemia (HbA1c) or pancreatic β-cell dysfunction (insulin), were independently associated with pancreatic cancer risk[49]. By comparing the proteome of PDAC with and without DM, our previous study indicated that regenerating gene (REG) Iα may be one of the connections between DM and PDAC[50]. The number of REG Iα positive cancer cells was significantly higher in pancreatic cancer patients with diabetes (n = 38) than in subjects without diabetes. Overexpression of the REG Iα protein in pancreatic cancer cell lines resulted in accelerated cell proliferation and consequently tumor growth, both in vitro and in vivo[50]. The IQ motif containing GTPase activating protein 1-exocyst axis is a growth factor- and nutrient-sensor that couples cell growth and division. It may function at the interface of cancer and diabetes[51].

Several preclinical and observational studies have shown that anti-diabetic medications may modify the risk of pancreatic cancer. A case-control study showed that diabetics treated with metformin had a significantly lower risk of pancreatic cancer (OR = 0.38; 95%CI: 0.22-0.69, P = 0.001)[52]. Metformin significantly decreased pancreatic cell growth[53]. These effects could be attributed to disruption of the crosstalk between insulin receptor and GPCR signaling[54], or up-regulation of miR-26a or other factors[53]. However, in a recent meta-analysis that included eleven studies, 1770 cases of pancreatic cancer in 730664 patients with DM were reported, indicating no significant association between metformin, insulin, or TZD use and risk of developing pancreatic cancer, and use of sulfonylureas was associated with a 70% increase in the risk of developing pancreatic cancer[55].

CIRCULATING BIOMARKERS FOR PDAC

Because PDAC develops over a long period of time and the curative response is significantly better in patients with early disease, an early diagnostic marker could positively impact the outcome of patients. Circulating biomarkers are always preferred over others because of their ease of collection and relatively noninvasive nature. The current standard serum marker, sialylated Lewis blood group antigen CA19-9, is widely used, but its use is limited to monitoring responses to therapy and not as a diagnostic marker because of its poor sensitivity (41%-86%) and specificity (33%-100%)[56]. CA19-9 can arise among patients with benign pancreaticobiliary disorders, notably cholestasis, and 5% to 10% of the population does not express Lewis antigens.

In the last two decades, many biomarkers have been tested for PDAC detection, some of which have higher specificity and sensitivity than CA19-9. These are new antibodies such as PAM4 recognizing MUC-1[57], soluble iC3b[58], REG4[59], serum phosphoproteins extracellular signal-regulated kinase (p-ERK1/2)[60], CEACAM1, a proliferation-inducing ligand[61], DJ-1[62], and laminin, gamma 2[63]. Further validation studies including a large number of cases are required for the clinical application of these biomarkers.

DJ-1 is up-regulated in 68.5% of PDAC specimens and correlates with tumor invasion and metastasis. The secretion of DJ-1 by tumor cells implies its potential as a biomarker[64]. Our data showed that the area under the curve (AUC) of serum DJ-1 is higher than that of CA 19-9 in certain patients with PDAC, and serum DJ-1 level also predicts poor patient outcome. Other groups confirmed our results[65] and showed the increase of DJ-1 in pancreatic juice[66].

The sensitivity and specificity of these biomarkers, which may be insufficient when used alone, can be improved by using them in combination or together with CA19-9. In a recent study[67], CA19.9 showed a better AUC in combination with SYCN, REG1B and AGR2 than when used alone. When analyzed in combination, three panels [CA19.9 + REG1B (AUC of 0.88), CA19.9 + SYCN + REG1B (AUC of 0.87) and CA19.9 + AGR2 + REG1B (AUC of 0.87)] showed a significantly better AUC (P < 0.05) than that of CA19.9 alone (AUC of 0.82). The superiority of the combination of biomarkers was also shown by our group and others[64,68].

Several recent reports showed that aberrant miRNA production is an early event in the development of PanIN lesions[69,70]. MiRNAs 21[71,72], 155[71], 16[73], 196a[73], 1290[74], 221[30], 375 (lower in PDAC)[30], and 18a[75] were identified by using miRNA expression profiling or other methods as potential biomarkers of PDAC alone or in combination with CA19-9 and each other[72,73]. A recent meta analysis of three blood based miRNA studies reported that the median specificity and sensitivity were 0.91 and 0.96, respectively[72]. Our group together with other centers[71] in China screened differentially expressed serum miRNAs with Illumina’s sequencing by synthesis technology using pooled serum samples followed by RT-qPCR validation of a large number of samples arranged in multiple stages in 97 PDAC cases and 158 age- and sex-matched cancer-free controls. We established 7 miRNA-based biomarker model (miR-20a, miR-21, miR-24, miR-25, miR-99a, miR-185, and miR-191) for PDAC diagnosis. These biomarkers had high sensitivity and specificity for distinguishing various stages of pancreatic cancer from cancer-free controls and also accurately discriminated pancreatic cancer patients from chronic pancreatitis patients (AUC = 0.993). In 26 stage I pancreatic cancer cases, the positive rate of pancreatic cancer detection by the 7 miRNA-based biomarker set was 96.2%, which was significantly higher than that of CA19-9 (46.2%) or CEA (30.8%) in the same sample set. In 48 stage II pancreatic cancer cases, the positive rate was 91.7%, which was also higher than that of CA19-9 (62.5%) or CEA (31.3%). Although miRNA detection is not currently used as a criterion for PDAC diagnosis, recent investigations indicated that they may be promising biomarkers in the near future.

Circulating tumor cells (CTCs) are tumor cells that have acquired the ability to enter the circulatory system. Studies have reported the presence of CTCs in peripheral blood in 40%-100% of pancreatic cancer patients[76,77], and their potential as biomarkers of PDAC was demonstrated recently[76-83]. CTCs have the potential to provide a surrogate for “real-time biopsy” of tumor biological activity. However, as CTCs are extremely rare, both enrichment and sensitive methods of detection are required for their enumeration[79]. Recently, using a modular system with innovative features, EpCAM positive CTCs were isolated from PDAC patients at high purity (> 86%) and with excellent yields (mean = 53 CTCs per mL)[76]. However, the high cost and involved procedure associated with this system constitute an obstacle to its clinical application.

CONCLUSION

The early detection of pancreatic cancer may benefit patients with PDAC. The slow development and progression of pancreatic cancer are closely associated with the activation of oncogenes, inactivation of tumor suppressor genes, altered expression of miRNAs, and activated tumor microenvironment. Therefore, a better understanding of the pathogenesis of PDAC may help detect PDAC or PDAC precursors at the early stages of the disease. However, a low rate of PDAC incidence and the limitations of current screening strategies make early detection difficult. A cost-effective screening strategy is required. The association of DM with PDAC may provide clues for early diagnosis and assessment of the progression of PDAC. Advances in the identification of novel circulating biomarkers including serological signatures, autoantibodies, epigenetic markers, circulating tumor cells and miRNAs have provided potential tools for the early detection of PDAC. However, there are currently no prospective studies investigating screening methods for PDAC in patients with new-onset DM, and biomarkers useful for this purpose or their combinations remain unidentified. Therefore, further studies are required in this field of research.

Footnotes

P- Reviewer: Chen YP, Hsu jt, Meister T, Kapan S, Kar P S- Editor: Ma YJ L- Editor: Wang TQ E- Editor: Zhang DN

References
1.  Siegel R, Naishadham D, Jemal A. Cancer statistics, 2013. CA Cancer J Clin. 2013;63:11-30.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9215]  [Cited by in F6Publishing: 9719]  [Article Influence: 883.5]  [Reference Citation Analysis (3)]
2.  Stathis A, Moore MJ. Advanced pancreatic carcinoma: current treatment and future challenges. Nat Rev Clin Oncol. 2010;7:163-172.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 555]  [Cited by in F6Publishing: 603]  [Article Influence: 43.1]  [Reference Citation Analysis (0)]
3.  Wolfgang CL, Herman JM, Laheru DA, Klein AP, Erdek MA, Fishman EK, Hruban RH. Recent progress in pancreatic cancer. CA Cancer J Clin. 2013;63:318-348.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 572]  [Cited by in F6Publishing: 636]  [Article Influence: 57.8]  [Reference Citation Analysis (0)]
4.  Yachida S, Jones S, Bozic I, Antal T, Leary R, Fu B, Kamiyama M, Hruban RH, Eshleman JR, Nowak MA. Distant metastasis occurs late during the genetic evolution of pancreatic cancer. Nature. 2010;467:1114-1117.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1872]  [Cited by in F6Publishing: 1837]  [Article Influence: 131.2]  [Reference Citation Analysis (0)]
5.  Cooper CL, O’Toole SA, Kench JG. Classification, morphology and molecular pathology of premalignant lesions of the pancreas. Pathology. 2013;45:286-304.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 47]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
6.  Ottenhof NA, Milne AN, Morsink FH, Drillenburg P, Ten Kate FJ, Maitra A, Offerhaus GJ. Pancreatic intraepithelial neoplasia and pancreatic tumorigenesis: of mice and men. Arch Pathol Lab Med. 2009;133:375-381.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 5]  [Reference Citation Analysis (0)]
7.  Macgregor-Das AM, Iacobuzio-Donahue CA. Molecular pathways in pancreatic carcinogenesis. J Surg Oncol. 2013;107:8-14.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 62]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
8.  Real FX, Cibrián-Uhalte E, Martinelli P. Pancreatic cancer development and progression: remodeling the model. Gastroenterology. 2008;135:724-728.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 24]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
9.  Jones S, Zhang X, Parsons DW, Lin JC, Leary RJ, Angenendt P, Mankoo P, Carter H, Kamiyama H, Jimeno A. Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science. 2008;321:1801-1806.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3082]  [Cited by in F6Publishing: 2881]  [Article Influence: 180.1]  [Reference Citation Analysis (0)]
10.  Biankin AV, Waddell N, Kassahn KS, Gingras MC, Muthuswamy LB, Johns AL, Miller DK, Wilson PJ, Patch AM, Wu J. Pancreatic cancer genomes reveal aberrations in axon guidance pathway genes. Nature. 2012;491:399-405.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1442]  [Cited by in F6Publishing: 1521]  [Article Influence: 126.8]  [Reference Citation Analysis (0)]
11.  Murphy SJ, Hart SN, Lima JF, Kipp BR, Klebig M, Winters JL, Szabo C, Zhang L, Eckloff BW, Petersen GM. Genetic alterations associated with progression from pancreatic intraepithelial neoplasia to invasive pancreatic tumor. Gastroenterology. 2013;145:1098-1109.e1.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 139]  [Cited by in F6Publishing: 138]  [Article Influence: 12.5]  [Reference Citation Analysis (0)]
12.  Ben QW, Jin XL, Liu J, Cai X, Yuan F, Yuan YZ. Periostin, a matrix specific protein, is associated with proliferation and invasion of pancreatic cancer. Oncol Rep. 2011;25:709-716.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 22]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
13.  Huang J, Yao WY, Zhu Q, Tu SP, Yuan F, Wang HF, Zhang YP, Yuan YZ. XAF1 as a prognostic biomarker and therapeutic target in pancreatic cancer. Cancer Sci. 2010;101:559-567.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 42]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
14.  Zhou L, Zhang R, Zhang L, Yao W, Li J, Yuan Y. Angiotensin-converting enzyme 2 acts as a potential molecular target for pancreatic cancer therapy. Cancer Lett. 2011;307:18-25.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 17]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
15.  Liu J, Ben QW, Yao WY, Zhang JJ, Chen DF, He XY, Li L, Yuan YZ. BMP2 induces PANC-1 cell invasion by MMP-2 overexpression through ROS and ERK. Front Biosci (Landmark Ed). 2012;17:2541-2549.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 22]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
16.  Cao H, Zhu Q, Huang J, Li B, Zhang S, Yao W, Zhang Y. Regulation and functional role of eEF1A2 in pancreatic carcinoma. Biochem Biophys Res Commun. 2009;380:11-16.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 34]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
17.  Ben QW, Wang JC, Liu J, Zhu Y, Yuan F, Yao WY, Yuan YZ. Positive expression of L1-CAM is associated with perineural invasion and poor outcome in pancreatic ductal adenocarcinoma. Ann Surg Oncol. 2010;17:2213-2221.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 74]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
18.  He X, Zheng Z, Li J, Ben Q, Liu J, Zhang J, Ji J, Yu B, Chen X, Su L. DJ-1 promotes invasion and metastasis of pancreatic cancer cells by activating SRC/ERK/uPA. Carcinogenesis. 2012;33:555-562.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 75]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
19.  Papaconstantinou IG, Lykoudis PM, Gazouli M, Manta A, Polymeneas G, Voros D. A review on the role of microRNA in biology, diagnosis, and treatment of pancreatic adenocarcinoma. Pancreas. 2012;41:671-677.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 29]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
20.  Kadera BE, Li L, Toste PA, Wu N, Adams C, Dawson DW, Donahue TR. MicroRNA-21 in pancreatic ductal adenocarcinoma tumor-associated fibroblasts promotes metastasis. PLoS One. 2013;8:e71978.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 108]  [Cited by in F6Publishing: 124]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
21.  Giovannetti E, Funel N, Peters GJ, Del Chiaro M, Erozenci LA, Vasile E, Leon LG, Pollina LE, Groen A, Falcone A. 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 337]  [Cited by in F6Publishing: 347]  [Article Influence: 24.8]  [Reference Citation Analysis (0)]
22.  Moriyama T, Ohuchida K, Mizumoto K, Yu J, Sato N, Nabae T, Takahata S, Toma H, Nagai E, Tanaka M. MicroRNA-21 modulates biological functions of pancreatic cancer cells including their proliferation, invasion, and chemoresistance. Mol Cancer Ther. 2009;8:1067-1074.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 233]  [Cited by in F6Publishing: 262]  [Article Influence: 17.5]  [Reference Citation Analysis (0)]
23.  Dillhoff M, Liu J, Frankel W, Croce C, Bloomston M. MicroRNA-21 is overexpressed in pancreatic cancer and a potential predictor of survival. J Gastrointest Surg. 2008;12:2171-2176.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 313]  [Cited by in F6Publishing: 337]  [Article Influence: 21.1]  [Reference Citation Analysis (0)]
24.  Bloomston M, Frankel WL, Petrocca F, Volinia S, Alder H, Hagan JP, Liu CG, Bhatt D, Taccioli C, Croce CM. MicroRNA expression patterns to differentiate pancreatic adenocarcinoma from normal pancreas and chronic pancreatitis. JAMA. 2007;297:1901-1908.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 873]  [Cited by in F6Publishing: 952]  [Article Influence: 56.0]  [Reference Citation Analysis (0)]
25.  Liu M, Du Y, Gao J, Liu J, Kong X, Gong Y, Li Z, Wu H, Chen H. Aberrant expression miR-196a is associated with abnormal apoptosis, invasion, and proliferation of pancreatic cancer cells. Pancreas. 2013;42:1169-1181.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 47]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
26.  Zhao G, Zhang JG, Shi Y, Qin Q, Liu Y, Wang B, Tian K, Deng SC, Li X, Zhu S. MiR-130b is a prognostic marker and inhibits cell proliferation and invasion in pancreatic cancer through targeting STAT3. PLoS One. 2013;8:e73803.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 92]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
27.  Zhang G, Zhou H, Xiao H, Liu Z, Tian H, Zhou T. MicroRNA-92a functions as an oncogene in colorectal cancer by targeting PTEN. Dig Dis Sci. 2014;59:98-107.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 88]  [Cited by in F6Publishing: 91]  [Article Influence: 9.1]  [Reference Citation Analysis (0)]
28.  Marin-Muller C, Li D, Bharadwaj U, Li M, Chen C, Hodges SE, Fisher WE, Mo Q, Hung MC, Yao Q. A tumorigenic factor interactome connected through tumor suppressor microRNA-198 in human pancreatic cancer. Clin Cancer Res. 2013;19:5901-5913.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 61]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
29.  Kawaguchi T, Komatsu S, Ichikawa D, Morimura R, Tsujiura M, Konishi H, Takeshita H, Nagata H, Arita T, Hirajima S. Clinical impact of circulating miR-221 in plasma of patients with pancreatic cancer. Br J Cancer. 2013;108:361-369.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 153]  [Cited by in F6Publishing: 175]  [Article Influence: 15.9]  [Reference Citation Analysis (0)]
30.  Su A, He S, Tian B, Hu W, Zhang Z. MicroRNA-221 mediates the effects of PDGF-BB on migration, proliferation, and the epithelial-mesenchymal transition in pancreatic cancer cells. PLoS One. 2013;8:e71309.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 55]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
31.  Wang P, Zhang J, Zhang L, Zhu Z, Fan J, Chen L, Zhuang L, Luo J, Chen H, Liu L. MicroRNA 23b regulates autophagy associated with radioresistance of pancreatic cancer cells. Gastroenterology. 2013;145:1133-1143.e12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 161]  [Cited by in F6Publishing: 178]  [Article Influence: 16.2]  [Reference Citation Analysis (0)]
32.  Muniyappa MK, Dowling P, Henry M, Meleady P, Doolan P, Gammell P, Clynes M, Barron N. MiRNA-29a regulates the expression of numerous proteins and reduces the invasiveness and proliferation of human carcinoma cell lines. Eur J Cancer. 2009;45:3104-3118.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 88]  [Cited by in F6Publishing: 101]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
33.  Nagano H, Tomimaru Y, Eguchi H, Hama N, Wada H, Kawamoto K, Kobayashi S, Mori M, Doki Y. MicroRNA-29a induces resistance to gemcitabine through the Wnt/β-catenin signaling pathway in pancreatic cancer cells. Int J Oncol. 2013;43:1066-1072.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 66]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
34.  Feig C, Gopinathan A, Neesse A, Chan DS, Cook N, Tuveson DA. The pancreas cancer microenvironment. Clin Cancer Res. 2012;18:4266-4276.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 841]  [Cited by in F6Publishing: 942]  [Article Influence: 85.6]  [Reference Citation Analysis (0)]
35.  Apte MV, Wilson JS, Lugea A, Pandol SJ. A starring role for stellate cells in the pancreatic cancer microenvironment. Gastroenterology. 2013;144:1210-1219.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 303]  [Cited by in F6Publishing: 314]  [Article Influence: 28.5]  [Reference Citation Analysis (0)]
36.  Spivak-Kroizman TR, Hostetter G, Posner R, Aziz M, Hu C, Demeure MJ, Von Hoff D, Hingorani SR, Palculict TB, Izzo J. Hypoxia triggers hedgehog-mediated tumor-stromal interactions in pancreatic cancer. Cancer Res. 2013;73:3235-3247.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 137]  [Cited by in F6Publishing: 144]  [Article Influence: 13.1]  [Reference Citation Analysis (0)]
37.  Canto MI, Harinck F, Hruban RH, Offerhaus GJ, Poley JW, Kamel I, Nio Y, Schulick RS, Bassi C, Kluijt I. International Cancer of the Pancreas Screening (CAPS) Consortium summit on the management of patients with increased risk for familial pancreatic cancer. Gut. 2013;62:339-347.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 546]  [Cited by in F6Publishing: 523]  [Article Influence: 47.5]  [Reference Citation Analysis (0)]
38.  Pannala R, Basu A, Petersen GM, Chari ST. New-onset diabetes: a potential clue to the early diagnosis of pancreatic cancer. Lancet Oncol. 2009;10:88-95.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 341]  [Cited by in F6Publishing: 377]  [Article Influence: 25.1]  [Reference Citation Analysis (0)]
39.  Cui Y, Andersen DK. Diabetes and pancreatic cancer. Endocr Relat Cancer. 2012;19:F9-F26.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 98]  [Cited by in F6Publishing: 100]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
40.  Li D. Diabetes and pancreatic cancer. Mol Carcinog. 2012;51:64-74.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 163]  [Cited by in F6Publishing: 185]  [Article Influence: 15.4]  [Reference Citation Analysis (0)]
41.  Huxley R, Ansary-Moghaddam A, Berrington de González A, Barzi F, Woodward M. Type-II diabetes and pancreatic cancer: a meta-analysis of 36 studies. Br J Cancer. 2005;92:2076-2083.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 758]  [Cited by in F6Publishing: 737]  [Article Influence: 38.8]  [Reference Citation Analysis (0)]
42.  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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 122]  [Cited by in F6Publishing: 110]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
43.  Ben Q, Cai Q, Li Z, Yuan Y, Ning X, Deng S, Wang K. The relationship between new-onset diabetes mellitus and pancreatic cancer risk: a case-control study. Eur J Cancer. 2011;47:248-254.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 67]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
44.  Pannala R, Leirness JB, Bamlet WR, Basu A, Petersen GM, Chari ST. Prevalence and clinical profile of pancreatic cancer-associated diabetes mellitus. Gastroenterology. 2008;134:981-987.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 385]  [Cited by in F6Publishing: 374]  [Article Influence: 23.4]  [Reference Citation Analysis (0)]
45.  He XY, Li JF, Yao WY, Yuan YZ. Resolution of new-onset diabetes after radical pancreatic resection predicts long-term survival in patients with pancreatic ductal cell adenocarcinoma. Ann Surg Oncol. 2013;20:3809-3816.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 20]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
46.  Wu JM, Kuo TC, Yang CY, Chiang PY, Jeng YM, Huang PH, Tien YW. Resolution of diabetes after pancreaticoduodenectomy in patients with and without pancreatic ductal cell adenocarcinoma. Ann Surg Oncol. 2013;20:242-249.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 39]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
47.  Kang SP, Saif MW. Clinical outcome of pancreatic cancer patients with diabetes mellitus: is diabetes a poor prognostic factor? Highlights from the “2010 ASCO Annual Meeting”. Chicago, IL, USA. June 4-8, 2010. JOP. 2010;11:334-335.  [PubMed]  [DOI]  [Cited in This Article: ]
48.  Cannon RM, LeGrand R, Chagpar RB, Ahmad SA, McClaine R, Kim HJ, Rupp C, Cho CS, Brinkman A, Weber S. Multi-institutional analysis of pancreatic adenocarcinoma demonstrating the effect of diabetes status on survival after resection. HPB (Oxford). 2012;14:228-235.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 40]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
49.  Wolpin BM, Bao Y, Qian ZR, Wu C, Kraft P, Ogino S, Stampfer MJ, Sato K, Ma J, Buring JE. Hyperglycemia, insulin resistance, impaired pancreatic β-cell function, and risk of pancreatic cancer. J Natl Cancer Inst. 2013;105:1027-1035.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 118]  [Cited by in F6Publishing: 123]  [Article Influence: 11.2]  [Reference Citation Analysis (0)]
50.  Zhou L, Zhang R, Wang L, Shen S, Okamoto H, Sugawara A, Xia L, Wang X, Noguchi N, Yoshikawa T. Upregulation of REG Ialpha accelerates tumor progression in pancreatic cancer with diabetes. Int J Cancer. 2010;127:1795-1803.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 29]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
51.  Osman MA, Sarkar FH, Rodriguez-Boulan E. A molecular rheostat at the interface of cancer and diabetes. Biochim Biophys Acta. 2013;1836:166-176.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 14]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
52.  Li D, Yeung SC, Hassan MM, Konopleva M, Abbruzzese JL. Antidiabetic therapies affect risk of pancreatic cancer. Gastroenterology. 2009;137:482-488.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 443]  [Cited by in F6Publishing: 480]  [Article Influence: 32.0]  [Reference Citation Analysis (0)]
53.  Li W, Yuan Y, Huang L, Qiao M, Zhang Y. Metformin alters the expression profiles of microRNAs in human pancreatic cancer cells. Diabetes Res Clin Pract. 2012;96:187-195.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 75]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
54.  Kisfalvi K, Eibl G, Sinnett-Smith J, Rozengurt E. Metformin disrupts crosstalk between G protein-coupled receptor and insulin receptor signaling systems and inhibits pancreatic cancer growth. Cancer Res. 2009;69:6539-6545.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 227]  [Cited by in F6Publishing: 251]  [Article Influence: 16.7]  [Reference Citation Analysis (0)]
55.  Singh S, Singh PP, Singh AG, Murad MH, McWilliams RR, Chari ST. Anti-diabetic medications and risk of pancreatic cancer in patients with diabetes mellitus: a systematic review and meta-analysis. Am J Gastroenterol. 2013;108:510-59; quiz 520.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 83]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
56.  Chakraborty S, Baine MJ, Sasson AR, Batra SK. Current status of molecular markers for early detection of sporadic pancreatic cancer. Biochim Biophys Acta. 2011;1815:44-64.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 58]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
57.  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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 96]  [Cited by in F6Publishing: 95]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
58.  Märten A, Büchler MW, Werft W, Wente MN, Kirschfink M, Schmidt J. Soluble iC3b as an early marker for pancreatic adenocarcinoma is superior to CA19.9 and radiology. J Immunother. 2010;33:219-224.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 20]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
59.  Takayama R, Nakagawa H, Sawaki A, Mizuno N, Kawai H, Tajika M, Yatabe Y, Matsuo K, Uehara R, Ono K. Serum tumor antigen REG4 as a diagnostic biomarker in pancreatic ductal adenocarcinoma. J Gastroenterol. 2010;45:52-59.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 37]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
60.  Takano S, Sogawa K, Yoshitomi H, Shida T, Mogushi K, Kimura F, Shimizu H, Yoshidome H, Ohtsuka M, Kato A. Increased circulating cell signalling phosphoproteins in sera are useful for the detection of pancreatic cancer. Br J Cancer. 2010;103:223-231.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 18]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
61.  Wang F, Chen L, Ding W, Wang G, Wu Y, Wang J, Luo L, Cong H, Wang Y, Ju S. Serum APRIL, a potential tumor marker in pancreatic cancer. Clin Chem Lab Med. 2011;49:1715-1719.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 22]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
62.  He XY, Liu BY, Yao WY, Zhao XJ, Zheng Z, Li JF, Yu BQ, Yuan YZ. Serum DJ-1 as a diagnostic marker and prognostic factor for pancreatic cancer. J Dig Dis. 2011;12:131-137.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 42]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
63.  Kosanam H, Prassas I, Chrystoja CC, Soleas I, Chan A, Dimitromanolakis A, Blasutig IM, Rückert F, Gruetzmann R, Pilarsky C. Laminin, gamma 2 (LAMC2): a promising new putative pancreatic cancer biomarker identified by proteomic analysis of pancreatic adenocarcinoma tissues. Mol Cell Proteomics. 2013;12:2820-2832.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 74]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
64.  Pardo M, García A, Thomas B, Piñeiro A, Akoulitchev A, Dwek RA, Zitzmann N. The characterization of the invasion phenotype of uveal melanoma tumour cells shows the presence of MUC18 and HMG-1 metastasis markers and leads to the identification of DJ-1 as a potential serum biomarker. Int J Cancer. 2006;119:1014-1022.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 88]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
65.  Chen Y, Kang M, Lu W, Guo Q, Zhang B, Xie Q, Wu Y. DJ-1, a novel biomarker and a selected target gene for overcoming chemoresistance in pancreatic cancer. J Cancer Res Clin Oncol. 2012;138:1463-1474.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 39]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
66.  Tian M, Cui YZ, Song GH, Zong MJ, Zhou XY, Chen Y, Han JX. 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 155]  [Cited by in F6Publishing: 169]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
67.  Makawita S, Dimitromanolakis A, Soosaipillai A, Soleas I, Chan A, Gallinger S, Haun RS, Blasutig IM, Diamandis EP. Validation of four candidate pancreatic cancer serological biomarkers that improve the performance of CA19.9. BMC Cancer. 2013;13:404.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 41]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
68.  Brand RE, Nolen BM, Zeh HJ, Allen PJ, Eloubeidi MA, Goldberg M, Elton E, Arnoletti JP, Christein JD, Vickers SM. Serum biomarker panels for the detection of pancreatic cancer. Clin Cancer Res. 2011;17:805-816.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 157]  [Cited by in F6Publishing: 175]  [Article Influence: 13.5]  [Reference Citation Analysis (0)]
69.  Xue Y, Abou Tayoun AN, Abo KM, Pipas JM, Gordon SR, Gardner TB, Barth RJ, Suriawinata AA, Tsongalis GJ. MicroRNAs as diagnostic markers for pancreatic ductal adenocarcinoma and its precursor, pancreatic intraepithelial neoplasm. Cancer Genet. 2013;206:217-221.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 58]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
70.  du Rieu MC, Torrisani J, Selves J, Al Saati T, Souque A, Dufresne M, Tsongalis GJ, Suriawinata AA, Carrère N, Buscail L. MicroRNA-21 is induced early in pancreatic ductal adenocarcinoma precursor lesions. Clin Chem. 2010;56:603-612.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 159]  [Cited by in F6Publishing: 165]  [Article Influence: 11.8]  [Reference Citation Analysis (0)]
71.  Wan C, Shen Y, Yang T, Wang T, Chen L, Wen F. Diagnostic value of microRNA for pancreatic cancer: a meta-analysis. Arch Med Sci. 2012;8:749-755.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 34]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
72.  Liu R, Chen X, Du Y, Yao W, Shen L, Wang C, Hu Z, Zhuang R, Ning G, Zhang C. Serum microRNA expression profile as a biomarker in the diagnosis and prognosis of pancreatic cancer. Clin Chem. 2012;58:610-618.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 276]  [Cited by in F6Publishing: 294]  [Article Influence: 22.6]  [Reference Citation Analysis (0)]
73.  Liu J, Gao J, Du Y, Li Z, Ren Y, Gu J, Wang X, Gong Y, Wang W, Kong X. Combination of plasma microRNAs with serum CA19-9 for early detection of pancreatic cancer. Int J Cancer. 2012;131:683-691.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 193]  [Cited by in F6Publishing: 210]  [Article Influence: 16.2]  [Reference Citation Analysis (0)]
74.  Li A, Yu J, Kim H, Wolfgang CL, Canto MI, Hruban RH, Goggins M. MicroRNA array analysis finds elevated serum miR-1290 accurately distinguishes patients with low-stage pancreatic cancer from healthy and disease controls. Clin Cancer Res. 2013;19:3600-3610.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 205]  [Cited by in F6Publishing: 225]  [Article Influence: 20.5]  [Reference Citation Analysis (0)]
75.  Morimura R, Komatsu S, Ichikawa D, Takeshita H, Tsujiura M, Nagata H, Konishi H, Shiozaki A, Ikoma H, Okamoto K. Novel diagnostic value of circulating miR-18a in plasma of patients with pancreatic cancer. Br J Cancer. 2011;105:1733-1740.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 189]  [Cited by in F6Publishing: 202]  [Article Influence: 15.5]  [Reference Citation Analysis (0)]
76.  Kamande JW, Hupert ML, Witek MA, Wang H, Torphy RJ, Dharmasiri U, Njoroge SK, Jackson JM, Aufforth RD, Snavely A. Modular microsystem for the isolation, enumeration, and phenotyping of circulating tumor cells in patients with pancreatic cancer. Anal Chem. 2013;85:9092-9100.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 91]  [Cited by in F6Publishing: 89]  [Article Influence: 8.1]  [Reference Citation Analysis (1)]
77.  Iwanicki-Caron I, Basile P, Toure E, Antonietti M, Lecleire S, Di Fiore A, Oden-Gangloff A, Blanchard F, Lemoine F, Di Fiore F. Usefulness of circulating tumor cell detection in pancreatic adenocarcinoma diagnosis. Am J Gastroenterol. 2013;108:152-155.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 23]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
78.  Sabbaghian MS, Rothberger G, Alongi AP, Gagner JP, Goldberg JD, Rolnitzky L, Chiriboga L, Hajdu CH, Zagzag D, Basch R. Levels of elevated circulating endothelial cell decline after tumor resection in patients with pancreatic ductal adenocarcinoma. Anticancer Res. 2010;30:2911-2917.  [PubMed]  [DOI]  [Cited in This Article: ]
79.  Tjensvoll K, Nordgård O, Smaaland R. Circulating tumor cells in pancreatic cancer patients: methods of detection and clinical implications. Int J Cancer. 2014;134:1-8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 83]  [Cited by in F6Publishing: 97]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
80.  Ren C, Chen H, Han C, Jin G, Wang D, Wang D, Tang D. Detection and molecular analysis of circulating tumor cells for early diagnosis of pancreatic cancer. Med Hypotheses. 2013;80:833-836.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 12]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
81.  Bidard FC, Huguet F, Louvet C, Mineur L, Bouché O, Chibaudel B, Artru P, Desseigne F, Bachet JB, Mathiot C. Circulating tumor cells in locally advanced pancreatic adenocarcinoma: the ancillary CirCe 07 study to the LAP 07 trial. Ann Oncol. 2013;24:2057-2061.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 133]  [Cited by in F6Publishing: 141]  [Article Influence: 12.8]  [Reference Citation Analysis (0)]
82.  Cen P, Ni X, Yang J, Graham DY, Li M. Circulating tumor cells in the diagnosis and management of pancreatic cancer. Biochim Biophys Acta. 2012;1826:350-356.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 33]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
83.  Kurihara T, Itoi T, Sofuni A, Itokawa F, Tsuchiya T, Tsuji S, Ishii K, Ikeuchi N, Tsuchida A, Kasuya K. Detection of circulating tumor cells in patients with pancreatic cancer: a preliminary result. J Hepatobiliary Pancreat Surg. 2008;15:189-195.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 110]  [Cited by in F6Publishing: 118]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]