Topic Highlight Open Access
Copyright ©The Author(s) 2016. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastrointest Oncol. Jan 15, 2016; 8(1): 18-29
Published online Jan 15, 2016. doi: 10.4251/wjgo.v8.i1.18
MicroRNA in pancreatic ductal adenocarcinoma and its precursor lesions
Yasmin G Hernandez, Department of Internal Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
Aimee L Lucas, Henry Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
Author contributions: Hernandez YG and Lucas AL wrote the paper.
Conflict-of-interest statement: Authors declare no conflict of interest for this article.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Aimee L Lucas, MD, MS, Assistant Professor of Medicine, Henry Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, Box #1069, New York, NY 10029, United States. aimee.lucas@mssm.edu
Telephone: +1-212-2410101 Fax: +1-646-5379616
Received: April 29, 2015
Peer-review started: May 7, 2015
First decision: September 8, 2015
Revised: November 4, 2015
Accepted: December 1, 2015
Article in press: December 2, 2015
Published online: January 15, 2016

Abstract

Pancreatic ductal adenocarcinoma (PDAC) is the 4th deadliest cancer in the United States, due to its aggressive nature, late detection, and resistance to chemotherapy. The majority of PDAC develops from 3 precursor lesions, pancreatic intraepithelial lesions (PanIN), intraductual papillary mucinous neoplasm (IPMN), and mucinous cystic neoplasm. Early detection and surgical resection can increase PDAC 5-year survival rate from 6% for Stage IV to 50% for Stage I. To date, there are no reliable biomarkers that can detect PDAC. MicroRNAs (miRNA) are small noncoding RNAs (18-25 nucleotides) that regulate gene expression by affecting translation of messenger RNA (mRNA). A large body of evidence suggests that miRNAs are dysregulated in various types of cancers. MiRNA has been profiled as a potential biomarker in pancreatic tumor tissue, blood, cyst fluid, stool, and saliva. Four miRNA biomarkers (miR-21, miR-155, miR-196, and miR-210) have been consistently dysregulated in PDAC. MiR-21, miR-155, and miR-196 have also been dysregulated in IPMN and PanIN lesions suggesting their use as early biomarkers of this disease. In this review, we explore current knowledge of miRNA sampling, miRNA dysregulation in PDAC and its precursor lesions, and advances that have been made in using miRNA as a biomarker for PDAC and its precursor lesions.

Key Words: Pancreatic cancer, MicroRNA, Biomarkers, Pancreatic intraepithelial lesions, Intraductal papillary mucinous neoplasm

Core tip: Reliable biomarkers are needed to detect pancreatic ductal adenocarcinoma (PDAC) early in order to decrease mortality. In this review, we discuss what the current knowledge is on microRNA (miRNA) in PDAC and its precursor lesions. MiR-21, miR-155, miR-196, miR-210 are dysregulated in tissue, serum, cyst fluid, and stool of PDAC patients. MiR-21, miR-155, and miR-196 are dysregulated in intraductual papillary mucinous neoplasm and pancreatic intraepithelial lesions demonstrating that these miRNAs may serve as potential biomarkers for early stage lesions and cancer.



INTRODUCTION

Pancreatic ductal adenocarcinoma (PDAC) is the 4th deadliest cancer in the United States, due to its aggressive nature, late detection, and resistance to chemotherapy[1,2]. The majority of PDAC develops from 3 precursor lesions, pancreatic intraepithelial lesions (PanIN), intraductual papillary mucinous neoplasm (IPMN), and mucinous cystic neoplasm (MCN)[3]. The cystic precursor lesions of the pancreas are detectable by certain imaging modalities such as Endoscopic ultrasound (EUS)[4-6], Magnetic Resonance Imaging of the Abdomen with Cholangiopancreatography[7], and computerized tomography scan[8,9]. To date, there is no modality that clearly detects PanIN lesions, although studies have suggested a correlation between multifocal PanIN and lobular atrophy of the pancreas on EUS[10].

Early detection and surgical resection can increase PDAC 5-year survival rate from 6% for Stage IV to 50% for Stage I[11,12]. Detection and surgical removal of precursor lesions has the potential to be curative. Because of this, there has been much research focused on identification of individuals at high-risk of PDAC, detection of early stage lesions, and on the discovery of reliable biomarkers of this deadly disease. Carbohydrate antigen (CA) 19-9 is a poor biomarker of PDAC, as it is elevated in 30%-40% of benign diseases of the pancreas[13,14] with a sensitivity of 79% (70%-90%) and specificity of 82% (68%-91%)[15] for PDAC.

MicroRNA (miRNA) expression has been studied in tumor detection, cancer development and progression, and prognosis[16]. MiRNAs are small noncoding RNAs (18-25 nucleotides) that regulate gene expression by affecting translation of messenger RNA (mRNA)[16-18]. MiRNA function to stabilize mRNA transcripts via post-transcriptional gene silencing via inhibition of the translation process or cleavage of their target mRNAs[16,19]. Over the last decade, the role of miRNA in cancer development and detection has evolved. MiRNA is very stable in tissue, plasma, stool, and other fluids and can be quantified in very small sample sizes, making it an excellent potential biomarker for the detection of PDAC. Current priorities include: (1) identification of miRNAs that are reliably dysregulated in PDAC; (2) determining which sample source(s) are easily accessible and have the highest yield for detecting these biomarkers; and (3) development of novel ways in which to use this information to detect early onset PDAC and precursor lesions. Array-based analysis is used to evaluate the expression levels of thousands of miRNAs in various tissue types. Subsequent trials have validated these findings by performing quantitative real time PCR (QRT-PCR) and performed receiver operating characteristics (ROC) analysis to determine the sensitivity and specificity of these miRNAs as potential biomarkers for early or advanced disease. Some miRNAs, such as miR-21, miR-155, miR-196a, and miR-210 have stood out as potential biomarkers of this highly fatal disease[20-24]. Our review is aimed at exploring current knowledge of miRNA sampling, miRNA dysregulation in PDAC and its precursor lesions, and advances that have been made in using miRNA as a biomarker for PDAC and its precursor lesions.

ROLE OF MIRNA IN PDAC DETECTION: SAMPLES FROM TISSUE, SERUM, PANCREATIC JUICE, STOOL AND SALIVA

Attention has been paid to circulating serological signatures, autoantibodies, epigenetic markers, circulating tumor cells (TCs), and miRNAs in order to detect PDAC at an earlier stage of disease[25,26]. The use of miRNA for diagnosis and screening is still an evolving field; in the right patient population, an ideal miRNA test would be highly sensitive and specific, minimally-invasive and cost-effective. MiRNA expression in PDAC was first examined in PDAC tissue cells[27]. Now miRNA has been found in serum, blood, whole plasma, stool, saliva, and cyst fluid (Table 1). Current knowledge is described below.

Table 1 MiRNA in pancreatic ductal adenocarcinoma.
MiRNAWhole pancreasSerum and plasmaSalivaStoolPancreatic juice
miR-10b↑[54]
miR-16↑[52]
miR-18a↑[29,56,57]
miR-20a↑[55,134]↓[62]
miR-21↑[22,24,27,32,34,38,55,71]↑[55,71]↑[63]↔[71]↑[61,62,71]
miR-24↑[55]
miR-27a-3p↑[134]↑[62]
miR-29c↓[27]
miR-30a-3p↓[27]
miR-30c↓[37]
miR-31↑[27]
miR-34a↑[37]
miR-96↓[27,135]↓[62]
miR-99a↑[52]
miR-101↑[32]
miR-106b↑[54]
miR-130b↓[27]
miR-135b↑[31]
miR-139-3p↓[37,58]
miR-141↓[27]
miR-143↑[27,71]↓[71]
miR-145↑[27]
miR-146a↑[27]↑[62]
miR-148a↓[27,29]
miR-148b↓[27-29]
miR-150↑[27]
miR-155↑[22-24,27,32,66,71]↑[22,54]↑[63]↓[71]↑[61,71]
miR-181a,b,d↑[24]↑[72]
miR-185↑[52,55,134]
miR-191↑[55]
miR-192↑[37,58]
miR-194↑[37]
miR-196a↑[22,23,27,49,52,71]↑[21,22,52,71]↑[72]↓[71]↑[62,71]
miR-196b↑[27]↑[60]
miR-200a↑[136]↑[62]
miR-200b↑[136]
miR-205
miR-210↑[27,71,137,138]↑[137]↑[72], ↔ [71]↑[60,71]
miR-212↑[38]↑[37]
miR-216↓[27,38]↓[63]↓[71]
miR-217↓[27]↓[62]
miR-222↑[24,27,38]
miR-223↑[27]
miR-375↓[27,71]↔[71]
miR-492↓[59]↑[60]
miR-494↓[27,74]
miR-508-5p↓[37]
miR-513a-5p↓[37]
miR-602↑[37]
miR-630↓[37]
miR-663a↓[59]
miR-801↑[37]
miR-887↓[37]
miR-923↓[37]
miR-940↑[74]↑[74]
miR-1290↑[30]
miR-1427↑[60]
miR-3679-5p↓[74]↓[72,74]
MiRNA in whole pancreas tissue or PDAC biopsies

Szafranska et al[27] performed the first analysis comparing miRNA expression in normal pancreas tissue, chronic pancreatitis (CP) tissue and PDAC tissue. On imaging, it can be challenging to distinguish CP from PDAC given the thick stroma and inflammation that may be found in both of these conditions. Furthermore, it is unclear if the aberrant expression of particular miRNAs is secondary to the desmoplastic reaction in CP and PDAC, and not related to tumorigenesis itself. They and others have found that miRNA-216 and miRNA-217 are significantly down-regulated in PDAC and miRNA-143, miR-145, miR-146a, miR-148a, miR-150, miR-155, miR-196a, miR-196b, miR-210, miR-222, miR-223, miR-31 are up-regulated in PDAC[24,27-29]. However, this study also demonstrated that dysregulation of miRNA-196a, miR-196b, miR-203, miR-210, miR-222, miR-217, and miR-375 were found only in PDAC, whereas miRNA-29c, miR-96, miR-143, miR-145, miR-148b, and miR-150 were abnormally expressed in both CP and PDAC. This may suggest that the latter are responsible for causing the desmoplastic reaction as opposed to tumorigenesis. MiR-1290 is elevated in early stage PDAC compared to normal controls[30]. Additionally, miR-135b has been shown to be an effective biomarker for distinguishing PDAC from CP with high sensitivity and specificity[31].

MiR-21, MiR-155, and miR-196 have been demonstrated by multiple groups to differentiate PDAC from non-cancerous lesions of the pancreas[20-24,27,32]. Special attention has been placed on the role of miR-21 in PDAC, as it has been implicated in tumorigenesis, TC invasion, the desmoplastic reaction, and metastasis of TC[33-36]. Further studies did not demonstrate that miR-21 expression in stromal cells correlated with tumor stage.

MiR-192 has also been found to be present in pancreatic TC, but is seldom seen in stromal cells and not found in adjacent normal pancreas tissue[37]. In this same study, miR-194 expression was detected in PDAC tissue, but not found in the surrounding normal pancreatic tissue. Unfortunately, despite these findings, no significant difference was found between the serum levels of miR-194 in patients with PDAC and healthy controls.

One proposed mechanism for PDAC development includes signaling between the molecular markers of the desmoplastic reaction and TCs[38-41]. Liffers et al[29] demonstrated that miR-148a is down-regulated in microdissected PDAC tissue and when over-expressed prevents tumor growth. This suggests that miR-148a may have a crucial role in the molecular signaling by which tumorigenesis occurs. While it is important to find biomarkers that are deregulated in PDAC, it is also important to understand which miRNAs are involved in these aberrant signaling pathways.

MiRNA in serum and plasma of PDAC patients

MiRNAs are known to have organ-specific expression in many human cancers[42,43]. Less than a decade ago, studies found that miRNA could reliably be detected in the serum in both animal models and humans[44,45], and since that time, there has been much research dedicated to identifying which miRNAs have differential expression and the implications of these findings in the detection, staging, treatment, and prognosis of cancers[46-50].

Attempts to use miRNA biomarkers in conjunction with CA19-9 have yielded mixed results. A study examining 847 different miRNAs in patients with PDAC found increased expression of miR-375 in PDAC as opposed to controls. MiR-375 did not improve detection nor predict prognosis in patients with PDAC when compared to CA19-9 alone[51]. Liu et al[52] found that using serum miR-16 and miR-196a in combination with CA19-9 increased detection of PDAC and Stage I lesions when compared to either modality alone, which suggests that miR-16 and miR-196a may be deregulated early in PDAC. These biomarkers were also up-regulated in studies performed on pancreas tissue, demonstrating that miR-16 and miR-196a can be used as peripheral biomarkers of PDAC. Gao, et al[53] also demonstrated that miR-16, when combined with CA19-9, served as a potential biomarker for detection of PDAC when compared to patients with CP.

One limitation of CA19-9 as a biomarker is that it is elevated in a large portion of patients with benign pancreatic diseases. Because of this limitation, studies have evaluated the miRNA expression of patients with PDAC compared to those with benign diseases such as CP or choledocolithiasis. They found that miR-10b, miR-155, and miR-106b were consistently elevated in the serum of patients with PDAC but not in those with benign pancreatic disease[54]. Liu et al[55] have demonstrated that up-regulation of miR-20a, miR-21, miR-24, miR-25, miR-99a, miR-185, and miR-191 can be used to distinguish PDAC from healthy controls and CP. Additionally, miR-135b has been shown to be an effective biomarker for distinguishing PDAC from CP with high sensitivity and specificity[31]. MiR-18a levels have also been shown to have increased expression in patients with PDAC and interestingly decrease after surgical resection suggesting that miR-18a levels may be a good marker to not only detect disease but also to monitor disease recurrence[29,56,57]. Zhang et al[37] also demonstrated that miR-194, miR-192, miR-602, miR-801, miR-212, miR-34a are up-regulated in PDAC, while miR-923, miR-139-3p, miR-513a-5p, miR-630, miR-30c-1, miR-887, miR-508-5p, and miR-139a-5p were down-regulated in PDAC specimens[37,58]. From these data, they demonstrated that miR-192 is neither present in the stromal cells of the pancreas nor the serum, but it is up-regulated in PDAC TCs and is involved in cell proliferation of PANC-1 TC lines in vitro[58]. Lin et al[59] performed microarray on 1711 serum miRNAs and found that 23 were down-regulated and 22 were up-regulated in the serum of PDAC patients when compared to normal controls. Of these, miR-492 and miR-663a were found to have decreased expression that was statistically significant in PDAC; however, only miR-663a was found to have a positive correlation with stage of disease[59]. Further studies are needed to determine which miRNAs will be clinically relevant.

Pancreatic juice miRNA

Pancreatic juice sampling requires an invasive endoscopic procedure, but studying the miRNA concentrations of patients with PDAC, benign pancreatic lesions, and healthy controls can shed light on potential biomarkers for detecting disease as they are found in high concentration in cyst fluid. As EUS and endoscopic retrograde cholangiopancreatogram (ERCP) are two methods by which pancreatic masses are frequently detected and sampled, these specimens could be sent for miRNA analysis in order to determine the malignant potential of these lesions. Wang et al[60] performed microarray of 49 miRNAs on secretin-stimulated pancreatic juice of a group of patients with PDAC, CP, and normal controls. They demonstrated that miR-205, miR-210, miR-492, and miR-1427 are all significantly elevated in PDAC when compared to controls; however, this statistical significance does not exist when compared to patients with CP[60]. Additionally, by using ROC curves, they determined that combining these 4 miRNAs with serum CA19-9 the sensitivity and specificity of PDAC detection is 91% and 100% respectively, though this analysis was limited by a sample size of 6. Other groups have evaluated the pancreatic juice of patients with PDAC pre-operatively via ERCP and from post-operative specimens[61,62]. Sadakari et al[61] analyzed the expression of miR-155 and miR-21 in pancreatic juice sampled via ERCP and found that these miRNAs were significantly elevated when compared to patients with CP and healthy controls, though the levels did not correlate with pancreatic juice cytology[61]. Again these findings are consistent with those from pancreatic tissue and serum. Hong et al[62] evaluated 158 miRNAs in post-operative fine needle aspiration specimens and found by qRT-PCR that miR-21, miR-27a, miR-146a, and miR-186a were significantly over-expressed in PDAC tissue and miR-217, miR-20a, and miR-96 were significantly down-regulated in PDAC tissue when compared to normal controls[62]. These two studies have demonstrated the feasibility of detecting miRNA from pancreatic juice, thus indicating the potential for using pancreatic juice biomarkers to detect early lesions given the higher concentration of miRNA in this fluid sample.

MiRNA in stool specimens

Frozen stool specimens may serve as potential non-invasive biomarker samples for PDAC. Over-expressed miRNAs from gastrointestinal cancers are shed from the exfoliative cells of the gastrointestinal tract. Intraluminal release of pancreatic juice also allows for detection of miRNAs in the stool[63-69]. Previous studies have largely focused on genetic markers of tumorigenesis and not miRNA[70]. Yang et al[63] performed a feasibility study of using stool miRNAs as a potential screening tool for detection of PDAC. They evaluated expression of 5 miRNAs that had been previously shown to be over-expressed in PDAC and found that miRNA-21 and miR-155 were over-expressed and miR-216 was under-expressed in all PDAC stool specimens when compared to normal controls and CP patients. These findings are consistent with what has been found in whole pancreas, pancreatic cyst fluid, and serum specimens. Additionally, with ROC analysis they demonstrated that combining miR-21 and miR-155 in stool samples there was a sensitivity of 93.33% and specificity of 66.67%. When they combined all 3 miRNAs (miR-21, miR-155, and miR-216), the sensitivity and specificity were 83.33% each. Link et al[71] selected 7 miRNAs (miR-21, miR-210, miR-143, miR-155, miR-196a, miR-216a, miR-375) and determined that like Yang’s group miR-216a was found in lower concentrations in the stool of patients with PDAC. However, unlike Yang’s group, they found that miR-155 was down-regulated in this population and miR-21 was unchanged in the stool of controls compared to CP or PDAC[71].

Ren et al[72] also evaluated the expression of miR-21, miR-155, miR-181a, miR-196a, and miR-210 and found that miR-181b, miR-196a, and miR-210 were significantly over-expressed in PDAC patients when compared to controls, but only miR-181b and miR-210 were elevated in CP patients, though these elevations were not significant. Ren et al[72] established a positive correlation between miR-196a levels and tumor size, which had not been previously described in studies of the serum or stool. Overall, while studies of fecal miRNA have demonstrated feasibility, conflicting data have emerged on which miRNAs are differentially expressed in the stool of PDAC, benign pancreatic disease, and normal controls.

Salivary miRNA

The field of salivanomics has been developing since blood molecules have been found in saliva[73]. As with stool miRNA, salivary miRNA may serve as a non-invasive biomarker for PDAC. Xie et al[74] is the only group to have evaluated salivary miRNA in PDAC. They conducted a microarray of 2006 miRNAs and noted that 10, including miR-4433-5p, miR-4665-3p, miR-940, miR-1273g-3p, miR-3676-5p, miR-3679-5p, miR-3940-5p, miR-4327, miR-4442, and miR-5100, were up-regulated or down-regulated in salivary samples. Of these, only miR-940 was significantly up-regulated and miR-3679-5p was significantly down-regulated in the PDAC specimens during the validation phase of the study. Until now, neither has been implicated in PDAC in the serum, stool, whole pancreas, or pancreatic juice. More studies are needed in this area.

ROLE OF MIRNA IN DETECTION OF PRECURSOR LESIONS

The absence of symptoms in early disease makes PDAC a cancer that is detected at very late stages when mortality approaches 100%. Much research has been dedicated to detecting miRNA in patients with PDAC as a novel biomarker for the presence of disease. Given the aggressive nature of PDAC, detection of precursor lesions with malignant potential would be critical to increasing the survival of these patients. PanIN lesions are microscopic PDAC precursor lesions that are graded 1-3 and are categorized based on the level of architectural and cytological atypia that is present[75,76]. Grade 1a is early intraepithelial proliferative lesions that have flat architecture, while grade 1b lesions have papillary architecture. PanIN-2 lesions have moderate abnormalities and PanIN-3 lesions have severe abnormalities, though none of these lesions invade the basement membrane[75,76]. IPMN lesions are mucin-producing cystic tumors, which arise from the epithelium of the pancreatic ducts and have the potential for malignant transformation[77,78]. They are categorized by main duct type (MD) or branch duct type (BD) and histologically are classified as having low-, intermediate-, and high-grade dysplasia[3]. Their malignant potential differs based on their location within the pancreatic ducts, and MD-IPMN carry a 44%-48% risk compared to BD-IPMNs, which only carry a 11%-17% risk of malignant transformation[79-82]. MCN are also mucin-producing epithelial neoplasms with ovarian-type stroma occur primarily in middle-aged females and are located in the body and tail of the pancreas and carry a 12% chance of tumor progression[83-86]. Cystic fluid is analyzed for CEA and amylase as other tumor markers have not demonstrated reliability in detecting malignant lesions.

As cystic neoplasms of the pancreas carry the risk of malignant transformation, determining a way to accurately predict which will progress to invasive carcinomas may guide surgical management and treatment decisions. MiRNA has been examined in PanIN lesions and IPMN as a potential candidate for early detection and the likelihood of progression to cancer. Understanding which miRNAs become deregulated early in the disease process may lead to advances for treatment decisions (Table 2).

Table 2 MiRNA in precursor lesions.
MiRNAIPMNPanIN-1PanIN-2PanIN-3
miR-10b↑[101]↑[101]
miR-21↑[23,32,87,88]↑[100,101]↑[98,99]↑[98,99]
miR-92a↑[93]
miR-99a↓[91,93]
miR-99b↓[91]
miR-100↓[91,93]
miR-101↓[32]
miR-125b↑[93]
miR-126↓ [91]
miR-130a↓ [91]
miR-145↑[93]
miR-148↓[101]↓[101]
miR-155↑[23,32]↑[101]↑[98]↑[98]
miR-182↑[101]
miR-196a↑[90]↑[97]↑[97]
miR-196b↑[97]↑[97]
miR-200a↑[101]
miR-200b↑[101]
miR-212↓[93]
miR-217↓[101]↓[101]
miR-221↑[87]
miR-296-5p↑[101]
miR-342-3p↓[91]
miR-483-3p↑[88,93]
MiRNA in IPMN lesions

Given the increased use of abdominal imaging, more pancreatic cystic lesions are being detected. There are guidelines in place to help guide management based on cystic characteristics that are consistent with malignancy[77]. The first study looking at miRNA expression levels in precursor lesions of the pancreas was performed by Habbe et al[23] who determined that miR-155 and miR-21 were over-expressed in the IPMN neoplastic epithelium, specifically those with carcinoma-in-situ[23,87]. MiR-155 was also significantly elevated in the pancreatic juice of these patients. While the levels of up-regulation of miRNA-21 and miR-155 correlated with the degree of cellular atypia found in the IPMN lesions, the study lacked long-term outcome data, highlighting the need for large and more longitudinal studies. Caponi et al[32] established a relationship between expression of miR-21 in invasive IPMNs and clinical outcome and observed that higher levels of miR-21 were correlated with worse overall and disease-free survival. Furthermore, they demonstrated that miR-155 and miR-21 had higher expression levels in invasive IPMN lesions when compared to non-invasive lesions, suggesting that these miRNAs may serve as early markers of malignant transformation[32,88]. MiR-101 has been shown to be down-regulated in invasive IPMNs when compared to non-invasive IPMNs and normal tissue. This deregulation of MiR-101 has not been described in PDAC samples, which may indicate that miR-101 plays a role in tumor invasion[32,89]. As with studies of miRNA in the serum and tissue of PDAC patients, miR-196a was found to be up-regulated in the pancreatic juice of intestinal-type IPMN[90]. In a recent study miR-100, miR-99b, miR-99a, miR-342-3p, miR-126, miR-130a were found to be down-regulated were up-regulated in high-risk vs low-risk IPMN lesions[91]. Furthermore, low miR-99b in IPMN fluid was associated with MD involvement, which is associated with a greater risk for transformation into a malignant neoplasm. Abue et al[88] found that miR-483-3p was up-regulated in PDAC cells and plasma when compared to IPMN lesions and may also serve as a useful biomarker in differentiating IPMN lesions with malignant potential from normal tissue and PDAC. The down-regulated miRNAs correlated with high-risk IPMNs and may be involved in cyst invasion and progression. Lee et al[92] found that miRNA expression varied amongst pancreatic cystic neoplasm. Specifically, miR-31-5p, miR-4830-5p, miR-99a-5p, and miR-375 were characteristic of serous cyst adenomas (SCA), whereas miR-10-5p, miR-202-3p, miR-210, and miR-375 differentiated MCN from SCA, IPMN, and PDAC[92]. It is unclear why this overlap in miR-375 occurs[92]. Henry et al[93] found that miR-92a, miR-99a, miR-100, miR-125b, miR-145, miR-212 and miR-483 were differentially expressed between benign and pre-malignant and malignant lesions of the pancreas and they suggested that a high amount of RNA present in the cystic fluid may suggest the presence of malignant transformation[93]. As previously described miR-21, miR-155, miR-196a have been implicated in both PDAC and IPMN and given these widely replicated results, studies aimed at detecting these biomarkers in serum, saliva, and stool could help to determine, in a non-invasive way, if they are increased in pre-malignant lesions.

MiRNA in panin lesions

Currently, PanIN lesions are found in the neighboring pancreatic tissue of patients with PDAC; however, there is no consistent way to detect the presence of these lesions[10,94-96]. Identifying a biomarker to detect PanIN lesions may be critical in early detection of PDAC. Slater et al[97] demonstrated that miRNA-196a and miR-196b were elevated in PDAC and PanIN-2/3 lesions in both animal models of PDAC and humans with PDAC. Ryu et al[98] demonstrated that miR-155 is up-regulated in PanIN-2 and PanIN-3 lesions when compared to neighboring healthy pancreatic tissue, but not in PanIN-1 lesions. Furthermore, miR-21 has been shown to be over-expressed in PanIN-2[98,99] and PanIN-3[98,99], but not PanIN-1 lesions suggesting that this is a marker for later disease. These are significant findings as miR-155 and miR-21 have been shown to be up-regulated in IPMN lesions and PDAC suggesting that they are early markers for cells with malignant potential. Yu et al[100] found that miR-196b was up-regulated in PanIN-3 lesions, which correlates with previous studies that have found that miR-196b is up-regulated in PDAC lesions[100,101]. Importantly, miR-21, miR-155, miR200a, miR-200b, miR-182, and miR-296-5p were deregulated as early as PanIN-1 lesions and remained deregulated until progression to PDAC, with the exception of miR-200c that normalized in PanIN-3 lesions. A recent publication describing miRNA expression in PanIN lesions found that miRNA-148a and miR-217 were down-regulated while miR-10b was up-regulated in PanIN-2 and PanIN-3 lesions[101]. While miR-21 has been shown repeatedly to be over-expressed in PDAC, there are conflicting studies on its deregulation in early PanIN lesions suggesting that it may represent a later and more aggressive dysregulation in the progression to PDAC. A non-invasive method to detect advanced PanIN lesions would represent a significant advance in the field.

DISCUSSION

While PDAC is the fourth most common cause of cancer-related deaths, there is still no reliable way to detect early disease and patients present with late-stage disease with a nearly 100% mortality. Research in the field of biomarkers shows a great deal of promise as current research aims to understand the molecular mechanisms and stromal microenvironment of this deadly tumor. MiRNA are small nucleotides that control the genetic expression in all cells and importantly in an organ-specific manner. Abberant miRNA expression has been identified in various cancers[102-106], and factors such as transcriptional deregulation, epigenetic alterations, mutations, DNA copy number abnormalities, and defects in the miRNA biogenesis pathway may account for these differences in expression[107,108]. C-myc and p53 are two transcriptional factors that have been associated transcriptional deregulation of miRNA[109-111], Epigenetic regulation of miRNAs by DNA methylation and histone tail modification play a role in miRNA expression through chromatin remodeling[105,112-114]. Both germ-line and somatic mutations are responsible for miRNA expression levels in various types of cancers[115-117]. It has been described by Calin et al[118], that miRNAs are located a fragile sites on the chromosome, minimal regions of loss of heterozygosity, minimal regions of amplifications, and common breakpoints, thus increasing the risk for DNA copy abnormalities. DNA copy abnormalities have been found in melanoma, breast cancer, ovarian cancer, leukemia, colorectal cancer[119-122]. Lastly, defects in miRNA biogenesis pathway may contribute to varying expression levels and cancer phenotype as miRNA undergoes complex processing intracellulary prior to reaching its mature form[123-127]. In addition to the aforementioned mechanisms, dietary components, such as folate, retinoids, curcumin, and Vitamin D have been implicated in the modulation of miRNA expression[128-130]. Some miRNAs have been shown to increase muscle loss in cancer cachexia and specifically, increased miR-21 levels have been shown to increase muscle breakdown in pancreatic cancer[131,132]. Deeper understanding of the regulatory mechanisms of miRNA expression will hopefully give new insight to the factors responsible for miRNA deregulation and lead to miRNA-based diagnostic testing and miRNA-directed therapy for PDAC.

Some limitations that exist with the current miRNA research at this time include standardization of extraction, reproducibility of testing, diagnostic yield in the various sample methods, and small sample sizes. Additionally, despite finding biomarkers for this disease, there is limited evidence that miRNA will impact PDAC-related mortality. Dysregulation of miRNA affects the cell cycle, proliferation, apoptosis, epigenetics, oncogenesis, tumor differentiation, tumor invasion, tumor metastasis and migration, prognosis, and chemoresistance in numerous cancers[133]. Increased efforts to understand the biological function of miRNA expression and its effects on cancer development are needed.

Despite these limitations, great advances have been made in this field and now miRNA expression is being analyzed not just in pancreatic tissue and cystic fluid, but also in stool, saliva, and serum; which would lead to non-invasive ways by which to analyze the expression levels of miRNA in patients at high risk. There have been great efforts to identify which of the greater than 2000 miRNAs are deregulated in PDAC and its precursor lesions and miRNA-21, miR-155, and miR-196b seem to be dysregulated in both early lesions and advanced cancer and show promise as potential screening tools in the future.

Footnotes

P- Reviewer: Slotwinski R S- Editor: Ji FF L- Editor: A E- Editor: Jiao XK

References
1.  Li D, Xie K, Wolff R, Abbruzzese JL. Pancreatic cancer. Lancet. 2004;363:1049-1057.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1481]  [Cited by in F6Publishing: 1516]  [Article Influence: 75.8]  [Reference Citation Analysis (0)]
2.  Jemal A, Siegel R, Ward E, Hao Y, Xu J, Murray T, Thun MJ. Cancer statistics, 2008. CA Cancer J Clin. 2008;58:71-96.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8283]  [Cited by in F6Publishing: 8168]  [Article Influence: 510.5]  [Reference Citation Analysis (0)]
3.  Klöppel G, Basturk O, Schlitter AM, Konukiewitz B, Esposito I. Intraductal neoplasms of the pancreas. Semin Diagn Pathol. 2014;31:452-466.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 72]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
4.  Rösch T, Lorenz R, Braig C, Feuerbach S, Siewert JR, Schusdziarra V, Classen M. Endoscopic ultrasound in pancreatic tumor diagnosis. Gastrointest Endosc. 1991;37:347-352.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 315]  [Cited by in F6Publishing: 315]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
5.  Müller MF, Meyenberger C, Bertschinger P, Schaer R, Marincek B. Pancreatic tumors: evaluation with endoscopic US, CT, and MR imaging. Radiology. 1994;190:745-751.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 357]  [Cited by in F6Publishing: 293]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
6.  Chang KJ. Endoscopic ultrasound-guided fine needle aspiration in the diagnosis and staging of pancreatic tumors. Gastrointest Endosc Clin N Am. 1995;5:723-734.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Park MJ, Kim YK, Choi SY, Rhim H, Lee WJ, Choi D. Preoperative detection of small pancreatic carcinoma: value of adding diffusion-weighted imaging to conventional MR imaging for improving confidence level. Radiology. 2014;273:433-443.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 51]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
8.  Canto MI, Goggins M, Hruban RH, Petersen GM, Giardiello FM, Yeo C, Fishman EK, Brune K, Axilbund J, Griffin C. Screening for early pancreatic neoplasia in high-risk individuals: a prospective controlled study. Clin Gastroenterol Hepatol. 2006;4:766-781; quiz 665.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 8]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
9.  Canto MI, Goggins M, Yeo CJ, Griffin C, Axilbund JE, Brune K, Ali SZ, Jagannath S, Petersen GM, Fishman EK. Screening for pancreatic neoplasia in high-risk individuals: an EUS-based approach. Clin Gastroenterol Hepatol. 2004;2:606-621.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 325]  [Cited by in F6Publishing: 303]  [Article Influence: 15.2]  [Reference Citation Analysis (0)]
10.  Brune K, Abe T, Canto M, O’Malley L, Klein AP, Maitra A, Volkan Adsay N, Fishman EK, Cameron JL, Yeo CJ. Multifocal neoplastic precursor lesions associated with lobular atrophy of the pancreas in patients having a strong family history of pancreatic cancer. Am J Surg Pathol. 2006;30:1067-1076.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Egawa S, Takeda K, Fukuyama S, Motoi F, Sunamura M, Matsuno S. Clinicopathological aspects of small pancreatic cancer. Pancreas. 2004;28:235-240.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 116]  [Cited by in F6Publishing: 111]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
12.  Ishikawa O, Ohigashi H, Imaoka S, Nakaizumi A, Uehara H, Kitamura T, Kuroda C. Minute carcinoma of the pancreas measuring 1 cm or less in diameter--collective review of Japanese case reports. Hepatogastroenterology. 1999;46:8-15.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Satake K, Kanazawa G, Kho I, Chung Y, Umeyama K. Evaluation of serum pancreatic enzymes, carbohydrate antigen 19-9, and carcinoembryonic antigen in various pancreatic diseases. Am J Gastroenterol. 1985;80:630-636.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Rosty C, Goggins M. Early detection of pancreatic carcinoma. Hematol Oncol Clin North Am. 2002;16:37-52.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 109]  [Cited by in F6Publishing: 118]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
15.  Goonetilleke KS, Siriwardena AK. Systematic review of carbohydrate antigen (CA 19-9) as a biochemical marker in the diagnosis of pancreatic cancer. Eur J Surg Oncol. 2007;33:266-270.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 539]  [Cited by in F6Publishing: 562]  [Article Influence: 31.2]  [Reference Citation Analysis (1)]
16.  Sethi S, Kong D, Land S, Dyson G, Sakr WA, Sarkar FH. Comprehensive molecular oncogenomic profiling and miRNA analysis of prostate cancer. Am J Transl Res. 2013;5:200-211.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Sethi A, Sholl LM. Emerging Evidence for MicroRNAs as Regulators of Cancer Stem Cells. Cancers (Basel). 2011;3:3957-3971.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 8]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
18.  Keller A, Leidinger P, Borries A, Wendschlag A, Wucherpfennig F, Scheffler M, Huwer H, Lenhof HP, Meese E. miRNAs in lung cancer - studying complex fingerprints in patient’s blood cells by microarray experiments. BMC Cancer. 2009;9:353.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 116]  [Cited by in F6Publishing: 124]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
19.  Hassan O, Ahmad A, Sethi S, Sarkar FH. Recent updates on the role of microRNAs in prostate cancer. J Hematol Oncol. 2012;5:9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 57]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
20.  Tang S, Bonaroti J, Unlu S, Liang X, Tang D, Zeh HJ, Lotze MT. Sweating the small stuff: microRNAs and genetic changes define pancreatic cancer. Pancreas. 2013;42:740-759.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 21]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
21.  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)]
22.  Wang J, Chen J, Chang P, LeBlanc A, Li D, Abbruzzesse JL, Frazier ML, Killary AM, Sen S. MicroRNAs in plasma of pancreatic ductal adenocarcinoma patients as novel blood-based biomarkers of disease. Cancer Prev Res (Phila). 2009;2:807-813.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 392]  [Cited by in F6Publishing: 418]  [Article Influence: 27.9]  [Reference Citation Analysis (0)]
23.  Habbe N, Koorstra JB, Mendell JT, Offerhaus GJ, Ryu JK, Feldmann G, Mullendore ME, Goggins MG, Hong SM, Maitra A. MicroRNA miR-155 is a biomarker of early pancreatic neoplasia. Cancer Biol Ther. 2009;8:340-346.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 221]  [Cited by in F6Publishing: 249]  [Article Influence: 16.6]  [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.  Yi JM, Guzzetta AA, Bailey VJ, Downing SR, Van Neste L, Chiappinelli KB, Keeley BP, Stark A, Herrera A, Wolfgang C. Novel methylation biomarker panel for the early detection of pancreatic cancer. Clin Cancer Res. 2013;19:6544-6555.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 96]  [Cited by in F6Publishing: 108]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
26.  He XY, Yuan YZ. Advances in pancreatic cancer research: moving towards early detection. World J Gastroenterol. 2014;20:11241-11248.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 52]  [Cited by in F6Publishing: 54]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
27.  Szafranska AE, Davison TS, John J, Cannon T, Sipos B, Maghnouj A, Labourier E, Hahn SA. MicroRNA expression alterations are linked to tumorigenesis and non-neoplastic processes in pancreatic ductal adenocarcinoma. Oncogene. 2007;26:4442-4452.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 483]  [Cited by in F6Publishing: 520]  [Article Influence: 30.6]  [Reference Citation Analysis (0)]
28.  Hanoun N, Delpu Y, Suriawinata AA, Bournet B, Bureau C, Selves J, Tsongalis GJ, Dufresne M, Buscail L, Cordelier P, Torrisani J. The silencing of microRNA 148a production by DNA hypermethylation is an early event in pancreatic carcinogenesis. Clin Chem. 2010;56:1107-1118.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 112]  [Cited by in F6Publishing: 124]  [Article Influence: 8.9]  [Reference Citation Analysis (0)]
29.  Liffers ST, Munding JB, Vogt M, Kuhlmann JD, Verdoodt B, Nambiar S, Maghnouj A, Mirmohammadsadegh A, Hahn SA, Tannapfel A. MicroRNA-148a is down-regulated in human pancreatic ductal adenocarcinomas and regulates cell survival by targeting CDC25B. Lab Invest. 2011;91:1472-1479.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 96]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
30.  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)]
31.  Munding JB, Adai AT, Maghnouj A, Urbanik A, Zöllner H, Liffers ST, Chromik AM, Uhl W, Szafranska-Schwarzbach AE, Tannapfel A. Global microRNA expression profiling of microdissected tissues identifies miR-135b as a novel biomarker for pancreatic ductal adenocarcinoma. Int J Cancer. 2012;131:E86-E95.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 86]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
32.  Caponi S, Funel N, Frampton AE, Mosca F, Santarpia L, Van der Velde AG, Jiao LR, De Lio N, Falcone A, Kazemier G. The good, the bad and the ugly: a tale of miR-101, miR-21 and miR-155 in pancreatic intraductal papillary mucinous neoplasms. Ann Oncol. 2013;24:734-741.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 73]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
33.  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)]
34.  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)]
35.  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)]
36.  Hwang JH, Voortman J, Giovannetti E, Steinberg SM, Leon LG, Kim YT, Funel N, Park JK, Kim MA, Kang GH. Identification of microRNA-21 as a biomarker for chemoresistance and clinical outcome following adjuvant therapy in resectable pancreatic cancer. PLoS One. 2010;5:e10630.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 219]  [Cited by in F6Publishing: 228]  [Article Influence: 16.3]  [Reference Citation Analysis (0)]
37.  Zhang J, Zhao CY, Zhang SH, Yu DH, Chen Y, Liu QH, Shi M, Ni CR, Zhu MH. Upregulation of miR-194 contributes to tumor growth and progression in pancreatic ductal adenocarcinoma. Oncol Rep. 2014;31:1157-1164.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Lee EJ, Gusev Y, Jiang J, Nuovo GJ, Lerner MR, Frankel WL, Morgan DL, Postier RG, Brackett DJ, Schmittgen TD. Expression profiling identifies microRNA signature in pancreatic cancer. Int J Cancer. 2007;120:1046-1054.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 632]  [Cited by in F6Publishing: 679]  [Article Influence: 39.9]  [Reference Citation Analysis (0)]
39.  Lou E, Subramanian S, Steer CJ. Pancreatic cancer: modulation of KRAS, MicroRNAs, and intercellular communication in the setting of tumor heterogeneity. Pancreas. 2013;42:1218-1226.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 16]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
40.  Rowley DR. Reprogramming the tumor stroma: a new paradigm. Cancer Cell. 2014;26:451-452.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 12]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
41.  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: 333]  [Article Influence: 30.3]  [Reference Citation Analysis (0)]
42.  Liu CG, Calin GA, Meloon B, Gamliel N, Sevignani C, Ferracin M, Dumitru CD, Shimizu M, Zupo S, Dono M. An oligonucleotide microchip for genome-wide microRNA profiling in human and mouse tissues. Proc Natl Acad Sci USA. 2004;101:9740-9744.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 741]  [Cited by in F6Publishing: 703]  [Article Influence: 35.2]  [Reference Citation Analysis (0)]
43.  He L, Thomson JM, Hemann MT, Hernando-Monge E, Mu D, Goodson S, Powers S, Cordon-Cardo C, Lowe SW, Hannon GJ. A microRNA polycistron as a potential human oncogene. Nature. 2005;435:828-833.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2729]  [Cited by in F6Publishing: 2783]  [Article Influence: 146.5]  [Reference Citation Analysis (0)]
44.  Mitchell PS, Parkin RK, Kroh EM, Fritz BR, Wyman SK, Pogosova-Agadjanyan EL, Peterson A, Noteboom J, O’Briant KC, Allen A. Circulating microRNAs as stable blood-based markers for cancer detection. Proc Natl Acad Sci USA. 2008;105:10513-10518.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5636]  [Cited by in F6Publishing: 6065]  [Article Influence: 379.1]  [Reference Citation Analysis (0)]
45.  Chen X, Ba Y, Ma L, Cai X, Yin Y, Wang K, Guo J, Zhang Y, Chen J, Guo X. Characterization of microRNAs in serum: a novel class of biomarkers for diagnosis of cancer and other diseases. Cell Res. 2008;18:997-1006.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3218]  [Cited by in F6Publishing: 3388]  [Article Influence: 211.8]  [Reference Citation Analysis (0)]
46.  Ali S, Saleh H, Sethi S, Sarkar FH, Philip PA. MicroRNA profiling of diagnostic needle aspirates from patients with pancreatic cancer. Br J Cancer. 2012;107:1354-1360.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 88]  [Cited by in F6Publishing: 101]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
47.  Qazi AM, Gruzdyn O, Semaan A, Seward S, Chamala S, Dhulipala V, Sethi S, Ali-Fehmi R, Philip PA, Bouwman DL. Restoration of E-cadherin expression in pancreatic ductal adenocarcinoma treated with microRNA-101. Surgery. 2012;152:704-711; discussion 711-713.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 25]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
48.  Brunet Vega A, Pericay C, Moya I, Ferrer A, Dotor E, Pisa A, Casalots À, Serra-Aracil X, Oliva JC, Ruiz A. microRNA expression profile in stage III colorectal cancer: circulating miR-18a and miR-29a as promising biomarkers. Oncol Rep. 2013;30:320-326.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 114]  [Cited by in F6Publishing: 125]  [Article Influence: 11.4]  [Reference Citation Analysis (0)]
49.  Kong X, Du Y, Wang G, Gao J, Gong Y, Li L, Zhang Z, Zhu J, Jing Q, Qin Y. 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 108]  [Cited by in F6Publishing: 122]  [Article Influence: 9.4]  [Reference Citation Analysis (0)]
50.  Ulivi P, Foschi G, Mengozzi M, Scarpi E, Silvestrini R, Amadori D, Zoli W. Peripheral blood miR-328 expression as a potential biomarker for the early diagnosis of NSCLC. Int J Mol Sci. 2013;14:10332-10342.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 57]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
51.  Carlsen AL, Joergensen MT, Knudsen S, de Muckadell OB, Heegaard NH. Cell-free plasma microRNA in pancreatic ductal adenocarcinoma and disease controls. Pancreas. 2013;42:1107-1113.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 26]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
52.  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)]
53.  Gao L, He SB, Li DC. Effects of miR-16 plus CA19-9 detections on pancreatic cancer diagnostic performance. Clin Lab. 2014;60:73-77.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  Cote GA, Gore AJ, McElyea SD, Heathers LE, Xu H, Sherman S, Korc M. A pilot study to develop a diagnostic test for pancreatic ductal adenocarcinoma based on differential expression of select miRNA in plasma and bile. Am J Gastroenterol. 2014;109:1942-1952.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 86]  [Article Influence: 8.6]  [Reference Citation Analysis (0)]
55.  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)]
56.  Komatsu S, Ichikawa D, Takeshita H, Morimura R, Hirajima S, Tsujiura M, Kawaguchi T, Miyamae M, Nagata H, Konishi H. Circulating miR-18a: a sensitive cancer screening biomarker in human cancer. In Vivo. 2014;28:293-297.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  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)]
58.  Zhao C, Zhang J, Zhang S, Yu D, Chen Y, Liu Q, Shi M, Ni C, Zhu M. Diagnostic and biological significance of microRNA-192 in pancreatic ductal adenocarcinoma. Oncol Rep. 2013;30:276-284.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 76]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
59.  Lin MS, Chen WC, Huang JX, Gao HJ, Sheng HH. Aberrant expression of microRNAs in serum may identify individuals with pancreatic cancer. Int J Clin Exp Med. 2014;7:5226-5234.  [PubMed]  [DOI]  [Cited in This Article: ]
60.  Wang J, Raimondo M, Guha S, Chen J, Diao L, Dong X, Wallace MB, Killary AM, Frazier ML, Woodward TA. Circulating microRNAs in Pancreatic Juice as Candidate Biomarkers of Pancreatic Cancer. J Cancer. 2014;5:696-705.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 92]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
61.  Sadakari Y, Ohtsuka T, Ohuchida K, Tsutsumi K, Takahata S, Nakamura M, Mizumoto K, Tanaka M. MicroRNA expression analyses in preoperative pancreatic juice samples of pancreatic ductal adenocarcinoma. JOP. 2010;11:587-592.  [PubMed]  [DOI]  [Cited in This Article: ]
62.  Hong TH, Park IY. MicroRNA expression profiling of diagnostic needle aspirates from surgical pancreatic cancer specimens. Ann Surg Treat Res. 2014;87:290-297.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 41]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
63.  Yang JY, Sun YW, Liu DJ, Zhang JF, Li J, Hua R. MicroRNAs in stool samples as potential screening biomarkers for pancreatic ductal adenocarcinoma cancer. Am J Cancer Res. 2014;4:663-673.  [PubMed]  [DOI]  [Cited in This Article: ]
64.  Leung WK, To KF, Man EP, Chan MW, Hui AJ, Ng SS, Lau JY, Sung JJ. Detection of hypermethylated DNA or cyclooxygenase-2 messenger RNA in fecal samples of patients with colorectal cancer or polyps. Am J Gastroenterol. 2007;102:1070-1076.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 84]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
65.  Nagasaka T, Tanaka N, Cullings HM, Sun DS, Sasamoto H, Uchida T, Koi M, Nishida N, Naomoto Y, Boland CR. Analysis of fecal DNA methylation to detect gastrointestinal neoplasia. J Natl Cancer Inst. 2009;101:1244-1258.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 94]  [Cited by in F6Publishing: 107]  [Article Influence: 7.1]  [Reference Citation Analysis (0)]
66.  Link A, Balaguer F, Shen Y, Nagasaka T, Lozano JJ, Boland CR, Goel A. Fecal MicroRNAs as novel biomarkers for colon cancer screening. Cancer Epidemiol Biomarkers Prev. 2010;19:1766-1774.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 216]  [Cited by in F6Publishing: 242]  [Article Influence: 17.3]  [Reference Citation Analysis (0)]
67.  Deschner EE. Early proliferative changes in gastrointestinal neoplasia. Am J Gastroenterol. 1982;77:207-211.  [PubMed]  [DOI]  [Cited in This Article: ]
68.  Kalimutho M, Del Vecchio Blanco G, Di Cecilia S, Sileri P, Cretella M, Pallone F, Federici G, Bernardini S. Differential expression of miR-144* as a novel fecal-based diagnostic marker for colorectal cancer. J Gastroenterol. 2011;46:1391-1402.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 92]  [Cited by in F6Publishing: 100]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
69.  Kanaoka S, Yoshida K, Miura N, Sugimura H, Kajimura M. Potential usefulness of detecting cyclooxygenase 2 messenger RNA in feces for colorectal cancer screening. Gastroenterology. 2004;127:422-427.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 58]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
70.  Haug U, Wente MN, Seiler CM, Jesenofsky R, Brenner H. Stool testing for the early detection of pancreatic cancer: rationale and current evidence. Expert Rev Mol Diagn. 2008;8:753-759.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 24]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
71.  Link A, Becker V, Goel A, Wex T, Malfertheiner P. Feasibility of fecal microRNAs as novel biomarkers for pancreatic cancer. PLoS One. 2012;7:e42933.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 60]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
72.  Ren Y, Gao J, Liu JQ, Wang XW, Gu JJ, Huang HJ, Gong YF, Li ZS. Differential signature of fecal microRNAs in patients with pancreatic cancer. Mol Med Rep. 2012;6:201-209.  [PubMed]  [DOI]  [Cited in This Article: ]
73.  Patel RS, Jakymiw A, Yao B, Pauley BA, Carcamo WC, Katz J, Cheng JQ, Chan EK. High resolution of microRNA signatures in human whole saliva. Arch Oral Biol. 2011;56:1506-1513.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 82]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
74.  Xie Z, Yin X, Gong B, Nie W, Wu B, Zhang X, Huang J, Zhang P, Zhou Z, Li Z. Salivary microRNAs show potential as a noninvasive biomarker for detecting resectable pancreatic cancer. Cancer Prev Res (Phila). 2015;8:165-173.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 92]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
75.  Hruban RH, Adsay NV, Albores-Saavedra J, Compton C, Garrett ES, Goodman SN, Kern SE, Klimstra DS, Klöppel G, Longnecker DS. Pancreatic intraepithelial neoplasia: a new nomenclature and classification system for pancreatic duct lesions. Am J Surg Pathol. 2001;25:579-586.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 855]  [Cited by in F6Publishing: 759]  [Article Influence: 33.0]  [Reference Citation Analysis (0)]
76.  Hruban RH, Takaori K, Klimstra DS, Adsay NV, Albores-Saavedra J, Biankin AV, Biankin SA, Compton C, Fukushima N, Furukawa T. An illustrated consensus on the classification of pancreatic intraepithelial neoplasia and intraductal papillary mucinous neoplasms. Am J Surg Pathol. 2004;28:977-987.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 788]  [Cited by in F6Publishing: 705]  [Article Influence: 35.3]  [Reference Citation Analysis (0)]
77.  Tanaka M, Fernández-del Castillo C, Adsay V, Chari S, Falconi M, Jang JY, Kimura W, Levy P, Pitman MB, Schmidt CM. International consensus guidelines 2012 for the management of IPMN and MCN of the pancreas. Pancreatology. 2012;12:183-197.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1714]  [Cited by in F6Publishing: 1546]  [Article Influence: 128.8]  [Reference Citation Analysis (0)]
78.  Adsay NV, Bagci P, Tajiri T, Oliva I, Ohike N, Balci S, Gonzalez RS, Basturk O, Jang KT, Roa JC. Pathologic staging of pancreatic, ampullary, biliary, and gallbladder cancers: pitfalls and practical limitations of the current AJCC/UICC TNM staging system and opportunities for improvement. Semin Diagn Pathol. 2012;29:127-141.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 93]  [Cited by in F6Publishing: 89]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
79.  Loftus EV, Olivares-Pakzad BA, Batts KP, Adkins MC, Stephens DH, Sarr MG, DiMagno EP. Intraductal papillary-mucinous tumors of the pancreas: clinicopathologic features, outcome, and nomenclature. Members of the Pancreas Clinic, and Pancreatic Surgeons of Mayo Clinic. Gastroenterology. 1996;110:1909-1918.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 269]  [Cited by in F6Publishing: 244]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
80.  Furukawa T, Hatori T, Fujita I, Yamamoto M, Kobayashi M, Ohike N, Morohoshi T, Egawa S, Unno M, Takao S. Prognostic relevance of morphological types of intraductal papillary mucinous neoplasms of the pancreas. Gut. 2011;60:509-516.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 239]  [Cited by in F6Publishing: 201]  [Article Influence: 15.5]  [Reference Citation Analysis (0)]
81.  Valsangkar NP, Morales-Oyarvide V, Thayer SP, Ferrone CR, Wargo JA, Warshaw AL, Fernández-del Castillo C. 851 resected cystic tumors of the pancreas: a 33-year experience at the Massachusetts General Hospital. Surgery. 2012;152:S4-12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 257]  [Cited by in F6Publishing: 284]  [Article Influence: 23.7]  [Reference Citation Analysis (0)]
82.  Rodriguez JR, Salvia R, Crippa S, Warshaw AL, Bassi C, Falconi M, Thayer SP, Lauwers GY, Capelli P, Mino-Kenudson M. Branch-duct intraductal papillary mucinous neoplasms: observations in 145 patients who underwent resection. Gastroenterology. 2007;133:72-79; quiz 309-310.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 352]  [Cited by in F6Publishing: 311]  [Article Influence: 18.3]  [Reference Citation Analysis (0)]
83.  Goh BK, Tan YM, Chung YF, Chow PK, Cheow PC, Wong WK, Ooi LL. A review of mucinous cystic neoplasms of the pancreas defined by ovarian-type stroma: clinicopathological features of 344 patients. World J Surg. 2006;30:2236-2245.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 150]  [Cited by in F6Publishing: 122]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
84.  Crippa S, Fernández-Del Castillo C, Salvia R, Finkelstein D, Bassi C, Domínguez I, Muzikansky A, Thayer SP, Falconi M, Mino-Kenudson M. Mucin-producing neoplasms of the pancreas: an analysis of distinguishing clinical and epidemiologic characteristics. Clin Gastroenterol Hepatol. 2010;8:213-219.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 236]  [Cited by in F6Publishing: 255]  [Article Influence: 18.2]  [Reference Citation Analysis (2)]
85.  Zamboni G, Scarpa A, Bogina G, Iacono C, Bassi C, Talamini G, Sessa F, Capella C, Solcia E, Rickaert F. Mucinous cystic tumors of the pancreas: clinicopathological features, prognosis, and relationship to other mucinous cystic tumors. Am J Surg Pathol. 1999;23:410-422.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 493]  [Cited by in F6Publishing: 386]  [Article Influence: 15.4]  [Reference Citation Analysis (0)]
86.  Crippa S, Fernández-del Castillo C. Management of intraductal papillary mucinous neoplasms. Curr Gastroenterol Rep. 2008;10:136-143.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 14]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
87.  Ryu JK, Matthaei H, Dal Molin M, Hong SM, Canto MI, Schulick RD, Wolfgang C, Goggins MG, Hruban RH, Cope L. Elevated microRNA miR-21 levels in pancreatic cyst fluid are predictive of mucinous precursor lesions of ductal adenocarcinoma. Pancreatology. 2011;11:343-350.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 85]  [Cited by in F6Publishing: 133]  [Article Influence: 10.2]  [Reference Citation Analysis (0)]
88.  Abue M, Yokoyama M, Shibuya R, Tamai K, Yamaguchi K, Sato I, Tanaka N, Hamada S, Shimosegawa T, Sugamura K. Circulating miR-483-3p and miR-21 is highly expressed in plasma of pancreatic cancer. Int J Oncol. 2015;46:539-547.  [PubMed]  [DOI]  [Cited in This Article: ]
89.  Nakahara O, Takamori H, Iwatsuki M, Baba Y, Sakamoto Y, Tanaka H, Chikamoto A, Horino K, Beppu T, Kanemitsu K. Carcinogenesis of intraductal papillary mucinous neoplasm of the pancreas: loss of microRNA-101 promotes overexpression of histone methyltransferase EZH2. Ann Surg Oncol. 2012;19 Suppl 3:S565-S571.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 27]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
90.  Aso T, Ohtsuka T, Matsunaga T, Kimura H, Watanabe Y, Tamura K, Ideno N, Osoegawa T, Takahata S, Shindo K. “High-risk stigmata” of the 2012 international consensus guidelines correlate with the malignant grade of branch duct intraductal papillary mucinous neoplasms of the pancreas. Pancreas. 2014;43:1239-1243.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 48]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
91.  Permuth-Wey J, Chen YA, Fisher K, McCarthy S, Qu X, Lloyd MC, Kasprzak A, Fournier M, Williams VL, Ghia KM. A genome-wide investigation of microRNA expression identifies biologically-meaningful microRNAs that distinguish between high-risk and low-risk intraductal papillary mucinous neoplasms of the pancreas. PLoS One. 2015;10:e0116869.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 36]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
92.  Lee LS, Szafranska-Schwarzbach AE, Wylie D, Doyle LA, Bellizzi AM, Kadiyala V, Suleiman S, Banks PA, Andruss BF, Conwell DL. Investigating MicroRNA Expression Profiles in Pancreatic Cystic Neoplasms. Clin Transl Gastroenterol. 2014;5:e47.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 31]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
93.  Henry JC, Bassi C, Giovinazzo F, Bloomston M. MicroRNA from pancreatic duct aspirate differentiates cystic lesions of the pancreas. Ann Surg Oncol. 2013;20 Suppl 3:S661-S666.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 14]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
94.  Brosens LA, Hackeng WM, Offerhaus GJ, Hruban RH, Wood LD. Pancreatic adenocarcinoma pathology: changing “landscape”. J Gastrointest Oncol. 2015;6:358-374.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 31]  [Reference Citation Analysis (0)]
95.  Andea A, Sarkar F, Adsay VN. Clinicopathological correlates of pancreatic intraepithelial neoplasia: a comparative analysis of 82 cases with and 152 cases without pancreatic ductal adenocarcinoma. Mod Pathol. 2003;16:996-1006.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 204]  [Cited by in F6Publishing: 187]  [Article Influence: 9.4]  [Reference Citation Analysis (0)]
96.  Shi C, Klein AP, Goggins M, Maitra A, Canto M, Ali S, Schulick R, Palmisano E, Hruban RH. Increased Prevalence of Precursor Lesions in Familial Pancreatic Cancer Patients. Clin Cancer Res. 2009;15:7737-7743.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 162]  [Cited by in F6Publishing: 160]  [Article Influence: 10.7]  [Reference Citation Analysis (0)]
97.  Slater EP, Strauch K, Rospleszcz S, Ramaswamy A, Esposito I, Klöppel G, Matthäi E, Heeger K, Fendrich V, Langer P. MicroRNA-196a and -196b as Potential Biomarkers for the Early Detection of Familial Pancreatic Cancer. Transl Oncol. 2014;7:464-471.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 58]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
98.  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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in F6Publishing: 105]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
99.  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)]
100.  Yu J, Li A, Hong SM, Hruban RH, Goggins M. MicroRNA alterations of pancreatic intraepithelial neoplasias. Clin Cancer Res. 2012;18:981-992.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 169]  [Cited by in F6Publishing: 173]  [Article Influence: 14.4]  [Reference Citation Analysis (0)]
101.  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)]
102.  Volinia S, Calin GA, Liu CG, Ambs S, Cimmino A, Petrocca F, Visone R, Iorio M, Roldo C, Ferracin M. A microRNA expression signature of human solid tumors defines cancer gene targets. Proc Natl Acad Sci USA. 2006;103:2257-2261.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4162]  [Cited by in F6Publishing: 4429]  [Article Influence: 246.1]  [Reference Citation Analysis (0)]
103.  Cummins JM, Velculescu VE. Implications of micro-RNA profiling for cancer diagnosis. Oncogene. 2006;25:6220-6227.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 196]  [Cited by in F6Publishing: 213]  [Article Influence: 11.8]  [Reference Citation Analysis (0)]
104.  Yanaihara N, Caplen N, Bowman E, Seike M, Kumamoto K, Yi M, Stephens RM, Okamoto A, Yokota J, Tanaka T. Unique microRNA molecular profiles in lung cancer diagnosis and prognosis. Cancer Cell. 2006;9:189-198.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2314]  [Cited by in F6Publishing: 2327]  [Article Influence: 129.3]  [Reference Citation Analysis (0)]
105.  Zhang L, Volinia S, Bonome T, Calin GA, Greshock J, Yang N, Liu CG, Giannakakis A, Alexiou P, Hasegawa K. Genomic and epigenetic alterations deregulate microRNA expression in human epithelial ovarian cancer. Proc Natl Acad Sci USA. 2008;105:7004-7009.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 382]  [Cited by in F6Publishing: 402]  [Article Influence: 25.1]  [Reference Citation Analysis (0)]
106.  Esquela-Kerscher A, Slack FJ. Oncomirs - microRNAs with a role in cancer. Nat Rev Cancer. 2006;6:259-269.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5384]  [Cited by in F6Publishing: 5489]  [Article Influence: 304.9]  [Reference Citation Analysis (0)]
107.  Deng S, Calin GA, Croce CM, Coukos G, Zhang L. Mechanisms of microRNA deregulation in human cancer. Cell Cycle. 2008;7:2643-2646.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 228]  [Cited by in F6Publishing: 244]  [Article Influence: 24.4]  [Reference Citation Analysis (0)]
108.  Breving K, Esquela-Kerscher A. The complexities of microRNA regulation: mirandering around the rules. Int J Biochem Cell Biol. 2010;42:1316-1329.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 160]  [Cited by in F6Publishing: 175]  [Article Influence: 11.7]  [Reference Citation Analysis (0)]
109.  Deng W, Cao X, Chen J, Zhang Z, Yu Q, Wang Y, Shao G, Zhou J, Gao X, Yu J. MicroRNA Replacing Oncogenic Klf4 and c-Myc for Generating iPS Cells via Cationized Pleurotus eryngii Polysaccharide-based Nanotransfection. ACS Appl Mater Interfaces. 2015;7:18957-18966.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 14]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
110.  He L, He X, Lowe SW, Hannon GJ. microRNAs join the p53 network--another piece in the tumour-suppression puzzle. Nat Rev Cancer. 2007;7:819-822.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 414]  [Cited by in F6Publishing: 409]  [Article Influence: 24.1]  [Reference Citation Analysis (0)]
111.  Chang TC, Yu D, Lee YS, Wentzel EA, Arking DE, West KM, Dang CV, Thomas-Tikhonenko A, Mendell JT. Widespread microRNA repression by Myc contributes to tumorigenesis. Nat Genet. 2008;40:43-50.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 973]  [Cited by in F6Publishing: 1013]  [Article Influence: 59.6]  [Reference Citation Analysis (0)]
112.  Lomberk GA, Urrutia R. The Triple-Code Model for Pancreatic Cancer: Cross Talk Among Genetics, Epigenetics, and Nuclear Structure. Surg Clin North Am. 2015;95:935-952.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 19]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
113.  Santosh B, Varshney A, Yadava PK. Non-coding RNAs: biological functions and applications. Cell Biochem Funct. 2015;33:14-22.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 98]  [Cited by in F6Publishing: 107]  [Article Influence: 10.7]  [Reference Citation Analysis (0)]
114.  Rodríguez-Vicente AE, Díaz MG, Hernández-Rivas JM. Chronic lymphocytic leukemia: a clinical and molecular heterogenous disease. Cancer Genet. 2013;206:49-62.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 50]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
115.  Shi L, Jackstadt R, Siemens H, Li H, Kirchner T, Hermeking H. p53-induced miR-15a/16-1 and AP4 form a double-negative feedback loop to regulate epithelial-mesenchymal transition and metastasis in colorectal cancer. Cancer Res. 2014;74:532-542.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 101]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
116.  Vos S, Vesuna F, Raman V, van Diest PJ, van der Groep P. miRNA expression patterns in normal breast tissue and invasive breast cancers of BRCA1 and BRCA2 germ-line mutation carriers. Oncotarget. 2015;6:32115-32137.  [PubMed]  [DOI]  [Cited in This Article: ]
117.  Wu M, Jolicoeur N, Li Z, Zhang L, Fortin Y, L’Abbe D, Yu Z, Shen SH. Genetic variations of microRNAs in human cancer and their effects on the expression of miRNAs. Carcinogenesis. 2008;29:1710-1716.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 120]  [Cited by in F6Publishing: 128]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
118.  Calin GA, Sevignani C, Dumitru CD, Hyslop T, Noch E, Yendamuri S, Shimizu M, Rattan S, Bullrich F, Negrini M. Human microRNA genes are frequently located at fragile sites and genomic regions involved in cancers. Proc Natl Acad Sci USA. 2004;101:2999-3004.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2908]  [Cited by in F6Publishing: 3030]  [Article Influence: 151.5]  [Reference Citation Analysis (0)]
119.  Zhang L, Huang J, Yang N, Greshock J, Megraw MS, Giannakakis A, Liang S, Naylor TL, Barchetti A, Ward MR. microRNAs exhibit high frequency genomic alterations in human cancer. Proc Natl Acad Sci USA. 2006;103:9136-9141.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 785]  [Cited by in F6Publishing: 780]  [Article Influence: 43.3]  [Reference Citation Analysis (0)]
120.  Mehrotra M, Luthra R, Ravandi F, Sargent RL, Barkoh BA, Abraham R, Mishra BM, Medeiros LJ, Patel KP. Identification of clinically important chromosomal aberrations in acute myeloid leukemia by array-based comparative genomic hybridization. Leuk Lymphoma. 2014;55:2538-2548.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 12]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
121.  Eldai H, Periyasamy S, Al Qarni S, Al Rodayyan M, Muhammed Mustafa S, Deeb A, Al Sheikh E, Afzal M, Johani M, Yousef Z. Novel genes associated with colorectal cancer are revealed by high resolution cytogenetic analysis in a patient specific manner. PLoS One. 2013;8:e76251.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 38]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
122.  Pallasch CP, Patz M, Park YJ, Hagist S, Eggle D, Claus R, Debey-Pascher S, Schulz A, Frenzel LP, Claasen J. miRNA deregulation by epigenetic silencing disrupts suppression of the oncogene PLAG1 in chronic lymphocytic leukemia. Blood. 2009;114:3255-3264.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 117]  [Cited by in F6Publishing: 125]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
123.  Hammond SM. An overview of microRNAs. Adv Drug Deliv Rev. 2015;87:3-14.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 808]  [Cited by in F6Publishing: 980]  [Article Influence: 108.9]  [Reference Citation Analysis (0)]
124.  Lee Y, Ahn C, Han J, Choi H, Kim J, Yim J, Lee J, Provost P, Rådmark O, Kim S. The nuclear RNase III Drosha initiates microRNA processing. Nature. 2003;425:415-419.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3513]  [Cited by in F6Publishing: 3520]  [Article Influence: 167.6]  [Reference Citation Analysis (0)]
125.  Yi R, Qin Y, Macara IG, Cullen BR. Exportin-5 mediates the nuclear export of pre-microRNAs and short hairpin RNAs. Genes Dev. 2003;17:3011-3016.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1970]  [Cited by in F6Publishing: 1963]  [Article Influence: 93.5]  [Reference Citation Analysis (0)]
126.  Lund E, Güttinger S, Calado A, Dahlberg JE, Kutay U. Nuclear export of microRNA precursors. Science. 2004;303:95-98.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1918]  [Cited by in F6Publishing: 1830]  [Article Influence: 91.5]  [Reference Citation Analysis (0)]
127.  Bohnsack MT, Czaplinski K, Gorlich D. Exportin 5 is a RanGTP-dependent dsRNA-binding protein that mediates nuclear export of pre-miRNAs. RNA. 2004;10:185-191.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 946]  [Cited by in F6Publishing: 957]  [Article Influence: 47.9]  [Reference Citation Analysis (0)]
128.  Davis CD, Ross SA. Evidence for dietary regulation of microRNA expression in cancer cells. Nutr Rev. 2008;66:477-482.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 78]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
129.  Wang WL, Chatterjee N, Chittur SV, Welsh J, Tenniswood MP. Effects of 1α,25 dihydroxyvitamin D3 and testosterone on miRNA and mRNA expression in LNCaP cells. Mol Cancer. 2011;10:58.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 94]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
130.  Kutmon M, Coort SL, de Nooijer K, Lemmens C, Evelo CT. Integrative network-based analysis of mRNA and microRNA expression in 1,25-dihydroxyvitamin D3-treated cancer cells. Genes Nutr. 2015;10:484.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 7]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
131.  He WA, Calore F, Londhe P, Canella A, Guttridge DC, Croce CM. Microvesicles containing miRNAs promote muscle cell death in cancer cachexia via TLR7. Proc Natl Acad Sci USA. 2014;111:4525-4529.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 164]  [Cited by in F6Publishing: 176]  [Article Influence: 17.6]  [Reference Citation Analysis (0)]
132.  Soares RJ, Cagnin S, Chemello F, Silvestrin M, Musaro A, De Pitta C, Lanfranchi G, Sandri M. Involvement of microRNAs in the regulation of muscle wasting during catabolic conditions. J Biol Chem. 2014;289:21909-21925.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 105]  [Cited by in F6Publishing: 113]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
133.  Wang N, Xia S, Chen K, Xiang X, Zhu A. Genetic alteration regulated by microRNAs in biliary tract cancers. Crit Rev Oncol Hematol. 2015;96:262-273.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 11]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
134.  Wang WS, Liu LX, Li GP, Chen Y, Li CY, Jin DY, Wang XL. Combined serum CA19-9 and miR-27a-3p in peripheral blood mononuclear cells to diagnose pancreatic cancer. Cancer Prev Res (Phila). 2013;6:331-338.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 65]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
135.  Yu S, Lu Z, Liu C, Meng Y, Ma Y, Zhao W, Liu J, Yu J, Chen J. miRNA-96 suppresses KRAS and functions as a tumor suppressor gene in pancreatic cancer. Cancer Res. 2010;70:6015-6025.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 249]  [Cited by in F6Publishing: 262]  [Article Influence: 18.7]  [Reference Citation Analysis (0)]
136.  Li A, Omura N, Hong SM, Vincent A, Walter K, Griffith M, Borges M, Goggins M. 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 227]  [Cited by in F6Publishing: 245]  [Article Influence: 17.5]  [Reference Citation Analysis (0)]
137.  Ho AS, Huang X, Cao H, Christman-Skieller C, Bennewith K, Le QT, Koong AC. Circulating miR-210 as a Novel Hypoxia Marker in Pancreatic Cancer. Transl Oncol. 2010;3:109-113.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 209]  [Cited by in F6Publishing: 219]  [Article Influence: 16.8]  [Reference Citation Analysis (0)]
138.  Schultz NA, Werner J, Willenbrock H, Roslind A, Giese N, Horn T, Wøjdemann M, Johansen JS. MicroRNA expression profiles associated with pancreatic adenocarcinoma and ampullary adenocarcinoma. Mod Pathol. 2012;25:1609-1622.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 103]  [Cited by in F6Publishing: 119]  [Article Influence: 9.9]  [Reference Citation Analysis (0)]