Next Article in Journal
The Impact of Remote Ischemic Preconditioning on Arterial Stiffness and Heart Rate Variability in Patients with Angina Pectoris
Previous Article in Journal
Tele-Operated Echography and Remote Guidance for Performing Tele-Echography on Geographically Isolated Patients
Previous Article in Special Issue
Considering Exosomal miR-21 as a Biomarker for Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of microRNAs in the Diagnosis and Treatment of Pancreatic Adenocarcinoma

1
Department of Internal Medicine, School of Medicine, Wayne State University, Detroit, MI 48201, USA
2
Department of Oncology, Karmanos Cancer institute, Wayne State University, Detroit, MI 48201, USA
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2016, 5(6), 59; https://doi.org/10.3390/jcm5060059
Submission received: 31 March 2016 / Revised: 8 June 2016 / Accepted: 13 June 2016 / Published: 16 June 2016
(This article belongs to the Special Issue MicroRNAs: Novel Biomarkers and Therapeutic Targets for Human Cancers)

Abstract

:
Pancreatic ductal adenocarcinoma (PDAC) remains a very challenging malignancy. Disease is diagnosed in an advanced stage in the vast majority of patients, and PDAC cells are often resistant to conventional cytotoxic drugs. Targeted therapies have made no progress in the management of this disease, unlike other cancers. microRNAs (miRs) are small non-coding RNAs that regulate the expression of multitude number of genes by targeting their 3′-UTR mRNA region. Aberrant expression of miRNAs has been linked to the development of various malignancies, including PDAC. In PDAC, a series of miRs have been defined as holding promise for early diagnostics, as indicators of therapy resistance, and even as markers for therapeutic response in patients. In this mini-review, we present an update on the various different miRs that have been defined in PDAC biology.

1. Introduction

Pancreatic cancer is the fourth leading cause of cancer-related deaths in the United States, with 53,070 new cases expected in 2016, of which 41,780 are expected to die from disease [1]. Surgery remains the only potentially curative treatment. However, a majority of patients present with non-resectable disease; only 15%–20% are surgical candidates at the time of diagnosis [2]. Surgery has an overall morbidity and mortality of 24% and 5.3%, respectively [3]. Tumor size less than 3 cm, negative surgical resection margins, well-differentiated histology and absence of lymph node involvement are favorable prognostic indicators [4]. Following a pancreaticoduodenectomy (Whipple procedure), the five-year survival rate is 25%–30% for node-negative [5] and 10% for node-positive disease [6]. This can be explained, in part, by the tumor’s high resistance to chemotherapy, as well as its propensity to recur and metastasize early, which may be related to the persistence of cancer stem cells (CSCs). Gemcitabine remains a commonly used drug in this disease [7]. Nab-paclitaxel has recently been shown to add to the benefit of gemcitabine in patients with favorable performance status [8]. The combination of fluorouracil, leucovorin, irinotecan, and oxalipatin (FOLFIRINOX) was also shown to be superior to gemcitabine, but, due to its side effect profile, it is reserved for patients with good performance [9]. More recently, monotherapy with S-1, an oral fluoropyrimidine derivative, demonstrated noninferiority to gemcitabine [10].
In light of the disappointing statistics in the prognosis of pancreatic ductal adenocarcinoma (PDAC), early detection of malignant and premalignant lesions is key. Unfortunately, no effective screening tool has been identified to date [11]. The tumors markers carcinoembryonic antigen (CEA) and carbohydrate antigen 19-9 (CA 19-9) are neither sensitive nor specific for screening but are used to follow known disease if they were initially elevated [12,13].
microRNAs (miRNA) are small (19–25 nucleotides) non-coding ribonucleic acids (RNAs) that interact with messenger RNA (mRNA) and serve as negative regulators of gene expression [14,15] by binding to imperfect complementary regions in the 3′ untranslated region of the target messenger RNA (mRNAs), inhibiting their translation or leading to their degradation. They have been shown to influence cell differentiation, proliferation, and apoptosis [16]. They represent only 3% of the human genome, but regulate 20%–30% of the protein coding genes [17,18]. They were first described in C. elegans in 1993 [19], and have a tissue-specific expression that is modified in a number of different conditions, including malignancy. They have been profiled in many different malignancies including breast [20], lung [21], and colorectal cancer [22] and differential expression was detected with those malignancies, all of which have made miRNAs promising biomarkers. The aim of this review is to present the evidence on the utility of miRNA in the diagnosis, treatment, and prognosis of PDAC.

2. microRNA in PDAC Biology

An understanding of the processes that govern the development of PDAC is crucial as it sheds light on potential biomarkers of early diagnosis and rational systemic therapeutic approaches. Multiple mutations in the evolution of PDAC are influenced by miRNAs, which serve as tumor promoters or suppressors by silencing or promoting of downstream pathways [23].
Activating mutations in KRAS are present in more than 90% of PDAC [24]. miRNA-96, 126, and 217, all of which target KRAS, were found to be downregulated in PDAC compared to other noncancerous, as well as normal, pancreatic tissues [25,26,27]. Furthermore, re-expression of miR-96 and 217 suppressed KRAS activity and resulted in reduced tumor migration and invasion, suggesting their role as tumor suppressors [26,27]. Additionally, miR-217 overexpression phosphorylated AKT levels, suggesting that miR-217 also influences downstream signaling involving cell survival and proliferation [27]. In another study, Kent et al. showed that RAS-responsive element-binding protein (RREB1) repressed the expression of miR-143/145 by binding to the promoter of the cluster [28]. Interestingly, oncogenic KRAS G12D mutations induce expression of RREB1 in PDAC to check the expression of miR-143/145 cluster. As the miR-143/145 cluster expression targets RREB1 protein to inhibit a feed forward circuit of KRAS signals through RREB1, the KRAS (G12D) mediated overexpression of RREB1 simultaneously represses the miR143/145 cluster expression, resulting in promotion of KRAS mediated signaling. Loss of expression of let-7 family miRNAs was described for the first time by Torrisani et al. [29]. Expression of let-7 suppressed KRAS expression and mitogen-activated protein kinase activation (MAPK), and inhibited cell proliferation but failed to hinder tumor progression [29].
Inactivation of p53 occurs in 50%–75% of PDAC, predominantly through missense mutations in the TP53 tumor suppressor gene [30]. Several studies showed that mutant p53 regulates the transcription of certain miRNAs, and, subsequently, influence the expression of their target genes either by degrading their messenger RNA or by inhibiting their translation [31,32]. miR-15a, a known transcriptional target of p53, was shown to be downregulated in PDAC [33]. The overexpression of miR-15a downregulated WNT3A and FGF7, resulting in reduced proliferation and survival of pancreatic cancer cells [33]. p53 has also been shown to induce the expression of miR-200 and repress that of Zeb1 and Zeb2, both of which are known activators of epithelial to mesenchymal transformation (EMT) [34]. In chemoresistant pancreatic cancer cell lines, miR-200 family was downregulated, suggesting a deregulated p53 signaling in those cell lines [34]. Furthermore, upregulation of Zeb1 was associated with downregulation of the miR-200 family expression [35]. The overexpression of miR-200 family led to the downregulation of Jag1, a target of Zeb1 and a ligand of the Notch pathway [35]. p53 not only regulates the expression of certain miRs but also is in turn modulated by specific miRs. miR-491-5p inhibited the expression of both TP53 and Bcl-XL genes, as well as mitogenic signaling pathways, such as STAT3 and PI-3K/Akt, resulting in decreased cell proliferation and induction of apoptosis [36]. Furthermore, Neault showed that miR-137 targets KMD4A messenger RNA during Ras-induced senescence, a tumor suppressor response, and activates both p53 and retinoblastoma tumor suppressor pathways [37]. miR-137 levels were found to be significantly reduced in PDAC; restoring its expression inhibited proliferation and promoted senescence of pancreatic cancer cells [37].
Aberrations in the expression of the p16 genes have been described in PDAC [38]. Also known as cyclin dependent kinase inhibitor 2A, p16 functions as a tumor suppressor gene by regulating cell cycle and cellular senescence. Studies have shown the inhibitory role of miR-10b and -24 on the expression of p16 in malignancies other than pancreatic cancer [39,40]. Both miR-10 and -24 were overexpressed in pancreatic cancer [41,42].
The TGFβ/SMAD pathway has been implicated in EMT. Through binding with their receptors, transforming growth factor β (TGFβ) isoforms transduce the phosphorylation of SMAD2 and SMAD3, which in turn bind to SMAD4 and translocate to the nucleus, where they regulate the transcription of target genes [43]. Other SMADs include SMAD-1, SMAD-5, and SMAD-8, and are collectively referred to as R-SMAD. On the other hand, SMAD-6 and SMAD-7 are negative regulators of R-SMADs and referred to as I-SMADs, or inhibitory SMADs [44]. While TGFβ acts as a tumor suppressant in normal cells by inhibiting cell growth, in cancer cells, the TGFβ/SMAD axis is modified resulting in impaired mediation of growth arrest [45]. Overexpression of the messenger RNAs encoding for TGFβ was observed PDAC and was associated with poor prognosis [46]. There is evidence suggesting that various microRNAs are regulated by the TGFβ/SMAD pathway, while others serve as regulators of that same pathway. The 130a/301a/454 microRNA family regulates TGFβ signaling through suppressing SMAD-4 expression by directly binding to its 3’UTR sequence [47]. This cluster was found to be upregulated in PDAC [48]. In another study, miR-421 and -483-3p promoted PDAC progression through directly regulating the tumor suppressor DPC4/SMAD4 [49,50]. Furthermore, aberrant expression of miR-146a on dendritic cells from PDAC patients was observed, and repression of SMAD-4 resulted in impaired differentiation as well as inhibition of antigen presenting function of dendritic cells, suggesting a role of microRNAs in modulating the immune response in PDAC patients through regulating TGFβ/SMAD signaling [51]. Overexpression of miR-192 was associated with a reduction in the expression of SMAD-interacting protein 1 (SIP1) [52]. Through direct suppression of SMAD2 and SMAD3, miR-323-3p inhibited TGFβ signaling, resulting in decreased cell motility and metastasis [53].

3. microRNA in PDAC Diagnosis

Accumulating evidence is showing that miRNA profiles are cell-specific and tumor-specific [54,55]. miRNAs have been so far isolated from the pancreatic tissue, pancreatic juices, bile, stool, blood, plasma, and sera of patients with pancreatic cancer [56]. Circulating miRNAs, specifically, have several exceptionally appealing characteristics: they are abundant, they are strongly resistant to degradation or modification compared to protein or carbohydrate-based tumor markers, their isolation is non-invasive and their amplification is technically easy and inexpensive [57,58]. Several miRNA profiles were observed to discriminate pancreatic cancer from benign pancreatic pathology and healthy samples. Circulating miRNA-483-3p levels are overexpressed in PDAC compared to intrapapillary mucinous neoplasms and healthy controls, and plasma levels of miR-483-3p differentiated PDAC from intraductal papillary mucinous neoplasm (IPMN) with a sensitivity (Sn) of 43.8%, similar to that of CA19-9 (45%) [59]. Elevated serum miR-200a and -200b levels were associated with silencing of SIP1 and overexpression of E-cadherin in patients with pancreatic cancer and chronic pancreatitis compared to healthy controls [60]. Serum miR-200a and -200b distinguished patients with PDAC from healthy controls with a Sn and specificity (Sp) of 84.4% and 87.5% for miR-200a and 71.1% and 96.9% for miR-200b, respectively [60].
Compared to traditionally used markers, serum miR-1290 distinguished patients with low-stage pancreatic cancer from controls better than CA19-9 did, and it was also found to influence pancreatic cancer cell invasion capability [61]. miR-16 and -196a independently discriminated pancreatic cancer patients from those with chronic pancreatitis or healthy controls. When CA 19-9 was added to the analysis, the discrimination was more sensitive and specific compared to microRNA panel or CA19-9 alone, with a Sn of 92% and Sp 95.6% for the discrimination of pancreatic cancer from healthy controls, and 88.4% and 96.3% for discriminating pancreatic cancer from chronic pancreatitis [62].
Specific alterations in miRNA expression are also noted in metastatic disease. Singh et al. showed at least a two-fold downregulation of miRNA-205 compared to nonmetastatic disease [63]. On the other hand, miR-146a was upregulated. Diagnostic kits profiling differentially expressed miRNAs were investigated to distinguish benign, premalignant, and malignant pancreatic lesions [64]. Szafranska et al. developed the first miR diagnostic, miRInform Pancreas, which utilized miR-196a and -217 to differentiate chronic pancreatitis from PDAC; their diagnostic Sn and Sp were 95% [64]. Lee et al. identified a panel of four miRs (miR-21-5p, 485-3p, 708-5p, and 375) that distinguished PDAC from IPMN with a Sn and Sp or 95% and 85%, respectively [65].
Table 1 and Table 2 list miRs that were shown to be upregulated and downregulated, respectively, in patients with pancreatic cancer, compared to benign pancreatic pathology and/or healthy samples.

4. microRNA in Therapy

4.1. Role of miRNAs in PDAC Therapy Resistance

The poor prognosis of pancreatic cancer is in part attributed to the high resistance rates to conventional chemotherapy. Accumulating evidence shows that most solid tumors are composed of two portions: the bulk and the cancer stem cell population. The latter survive the initial chemotherapy and utilize their self-renewal capabilities to regenerate a secondary population of tumor cells that is resistance to therapy. This inherent characteristic of CSCs might be controlled by specific miRNAs [63]. Jung et al. detected differentially expressed miRNAs in CSCs, including miR-99a, miR-100, miR-125b, miR-192, and miR-429 [95]. Certain alterations in miRNA expression are associated with chemoresistance. miRNA-200 family expression downregulation was observed in gemcitabine-resistant pancreatic cancer cells [96]. The mechanisms through which miRNAs induce chemoresistance have been elucidated in some studies. Hamada et al. showed that miR-365 induced chemoresistance through directly targeting the adaptor protein Src Homology 2 Domain Containing 1 (SHC1) and apoptosis-promoting protein BAX. It also upregulated S100P and Inhibitor of DNA binding 2, both of which are cancer-promoting molecules [97]. On the other hand, miRNA-34 regulated Notch signaling, leading to reduction in pancreatic CSC population [97]. Another study showed that miR-1246 expression induced chemoresistance through downregulating CCNG2 [98].

4.2. Potential of miRNAs as PDAC Therapeutics

As miRNAs regulate multiple gene expressions and signaling pathways, miRNA-based therapies are at an advantage over single-gene therapy, and, at least hypothetically, targeting miRNAs is expected to produce more effective anti-cancer activities. To that goal, multiple approaches have been utilized in vitro and in vivo, aiming for the downregulation of oncogenic miRNAs and/or the restoration of tumor suppressor ones. Approaches included introducing a miR antagonist or use of an miR mimic agent [55]. Transfecting pancreatic CSCs with a miR-200c mimic decreased colony formation, invasion and chemoresistance of pancreatic CSCs by regulating EMT [99]. Lu et al. reached similar results with transfection of miR-200a [100]. On the same note, transfecting gemcitabine-resistant pancreatic cells with miRNA-205 and miR-7 reduced the expression of TUBB3 and Pak-1, respectively, and reduced the CSC population [63]. Administering complexed micelles of gemcitabine and the tumor suppressor miRNA-205 achieved significant inhibition of tumor growth in a pancreatic tumor model; immuno-histochemical analysis showed decreased tumor cell proliferation and increased apoptosis [101]. Transfection efficiency was >90%. In another study, targeting miR-21 with lentiviral vectors inhibited cell proliferation [102]. Pancreatic stellate cells (PSCs) represent the precursor cells for cancer-associated fibroblasts in pancreatic tumor stroma [103]. Kuninty et al. showed that suppressing miR-199a and -214 in PSCs abolished the PSC-driven pro-tumor effects and resulted in decreased tumor cell growth [103].
Using treatment with the demethylating agent 5-Aza-2′-deoxycytidine (5-Aza-dC) and HDAC inhibitor vorinostat (SAHA), Nalls et al. restored the expression of miR-34, a transcriptional target of p53, which induced apoptosis and inhibited cell cycle progression and epithelial to mesenchymal transition [104]. Systemic intravenous delivery with miR-34a and miR-143/145 nanovectors inhibited the growth of MiaPsCa-2 subcutaneous xenografts in mouse models; this was displayed even in the orthotopic setting [105]. Treatment with a synthetic (fluorinated) curcumin analogue, CDF, led to the downregulation of miR-21, restoration of miR-200 and tumor suppressor PTEN, and the killing of the CSC population, resulting in suppressed tumor growth [106]. This was previously observed in the work of Ali et al., as well as others [96,107,108,109,110,111]. Oral curcumin was well tolerated and showed some response in one phase II trial [112]. In another study, treatment with isoflavone or 3,3′-diindolylmethane (DIM) reversed the EMT, restored expression of the miRNA-200 family, and resensitized pancreatic cancer cells to gemcitabine [113].
Following miR expression patterns over the course of treatment provides a tool to monitor tumor burden, as well as the emergence of resistant strains of cancer cells, which would prompt modifying therapy [114]. In two studies, plasma levels of miR-18a and 221 dropped postoperatively in nine and eight patients, respectively [86,90]; furthermore, in one patient who had recurrence after surgery, miR-18a levels re-elevated with no similar change in the levels of CA19-9.

5. microRNAs as Prognostic Biomarkers

Evidence shows that certain miR profiles are associated with a more aggressive disease and worse survival. In a meta-analysis involving 1525 patients, overall and disease-free survivals were significantly shorter in patients with high tumoral miR-21 [115]. This was further shown in the work of Abue et al. [59]. Poor survival was also linked to high miR-155, 203, 222, and 10b, and low miR-34a levels [115]. Similarly, lower expression of miR-183 reduced survival compared to higher levels, and was significantly associated with tumor grade, metastasis, and TNM stage [116]. Overexpression of miR-1290 was also associated with worse outcomes [61].

6. Other Noncoding RNAs

Although miRNAs have gained a lot of praise as future biomarkers for PDAC, other less popular small noncoding RNAs (snRNAs), as well as long noncoding RNAs (lnRNAs), are also being studied as diagnostic and prognostic biomarkers. Circulating U2 snRNA identified PDAC from controls with high sensitivity and specificity [117]. Overexpression of lncRNAs HOTAIR, HULC, MALAT1, and PVT1 were observed in PDAC compared to non-cancerous controls, and was associated with more aggressive disease [118,119,120,121]. In another study, overexpression of lncRNA was associated with inhibition of cell proliferation [122].

7. Conclusions

Accumulating evidence supports the strong involvement of microRNAs in the pathogenesis of PDAC, highlighting their many different roles in the KRAS, p53, and TGFβ/SMAD pathways, among others. Whether it is their abundance, their resistance to degradation, the feasibility of isolating them noninvasively, or the ease of amplifying them, miRNAs represent appealing biomarkers that have so far been linked to the diagnosis, therapy, as well as the prognosis of PDAC. However, despite the many efforts that have occurred, a practical application to be used in the clinic is still lacking.

Acknowledgments

Work in the lab of A.S.A. is supported by NIH NCI 1R21CA188818-01A1. The authors acknowledge the support from Perri Foundation and SKY foundation.

Author Contributions

All authors are aware of the content of this manuscript. Maria Diab wrote significant portions of the text and all co-authors have read and edited the manuscript.

Conflicts of Interest

None of the authors have any conflicts of interest to disclose. This manuscript received no sources of funding.

References

  1. Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2016. CA: Cancer J. Clin. 2016, 66, 7–30. [Google Scholar] [CrossRef] [PubMed]
  2. Yeo, C.J.; Cameron, J.L. Prognostic factors in ductal pancreatic cancer. Langenbeck’s Arch. Surg. Deutsche Ges. Chir. 1998, 383, 129–133. [Google Scholar] [CrossRef]
  3. Benassai, G.; Mastrorilli, M.; Quarto, G.; Cappiello, A.; Giani, U.; Mosella, G. Survival after pancreaticoduodenectomy for ductal adenocarcinoma of the head of the pancreas. Chir. Ital. 2000, 52, 263–270. [Google Scholar] [PubMed]
  4. Yeo, C.J.; Cameron, J.L.; Sohn, T.A.; Lillemoe, K.D.; Pitt, H.A.; Talamini, M.A.; Hruban, R.H.; Ord, S.E.; Sauter, P.K.; Coleman, J.; et al. Six hundred fifty consecutive pancreaticoduodenectomies in the 1990s: Pathology, complications, and outcomes. Ann. Surg. 1997, 226, 248–257, discussion 257–260. [Google Scholar] [CrossRef] [PubMed]
  5. Trede, M.; Schwall, G.; Saeger, H.D. Survival after pancreatoduodenectomy. 118 consecutive resections without an operative mortality. Ann. Surg. 1990, 211, 447–458. [Google Scholar] [CrossRef] [PubMed]
  6. Kang, M.J.; Jang, J.Y.; Chang, Y.R.; Kwon, W.; Jung, W.; Kim, S.W. Revisiting the concept of lymph node metastases of pancreatic head cancer: Number of metastatic lymph nodes and lymph node ratio according to n stage. Ann. Surg. Oncol. 2014, 21, 1545–1551. [Google Scholar] [CrossRef] [PubMed]
  7. Heinemann, V.; Haas, M.; Boeck, S. Systemic treatment of advanced pancreatic cancer. Cancer Treat. Rev. 2012, 38, 843–853. [Google Scholar] [CrossRef] [PubMed]
  8. Von Hoff, D.D.; Ervin, T.; Arena, F.P.; Chiorean, E.G.; Infante, J.; Moore, M.; Seay, T.; Tjulandin, S.A.; Ma, W.W.; Saleh, M.N.; et al. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N. Engl. J. Med. 2013, 369, 1691–1703. [Google Scholar] [CrossRef] [PubMed]
  9. Conroy, T.; Desseigne, F.; Ychou, M.; Bouche, O.; Guimbaud, R.; Becouarn, Y.; Adenis, A.; Raoul, J.L.; Gourgou-Bourgade, S.; de la Fouchardiere, C.; et al. Folfirinox versus gemcitabine for metastatic pancreatic cancer. N. Engl. J. Med. 2011, 364, 1817–1825. [Google Scholar] [CrossRef] [PubMed]
  10. Ueno, H.; Ioka, T.; Ikeda, M.; Ohkawa, S.; Yanagimoto, H.; Boku, N.; Fukutomi, A.; Sugimori, K.; Baba, H.; Yamao, K.; et al. Randomized phase iii study of gemcitabine plus s-1, s-1 alone, or gemcitabine alone in patients with locally advanced and metastatic pancreatic cancer in japan and taiwan: Gest study. J. Clin. Oncol. 2013, 31, 1640–1648. [Google Scholar] [CrossRef] [PubMed]
  11. Ryan, D.P.; Hong, T.S.; Bardeesy, N. Pancreatic adenocarcinoma. N. Engl. J. Med. 2014, 371, 1039–1049. [Google Scholar] [CrossRef] [PubMed]
  12. DiMagno, E.P.; Reber, H.A.; Tempero, M.A. Aga technical review on the epidemiology, diagnosis, and treatment of pancreatic ductal adenocarcinoma. American gastroenterological association. Gastroenterology 1999, 117, 1464–1484. [Google Scholar] [CrossRef]
  13. Lamerz, R. Role of tumour markers, cytogenetics. Ann. Oncol. 1999, 10 (Suppl. 4), 145–149. [Google Scholar] [CrossRef] [PubMed]
  14. Galasso, M.; Sandhu, S.K.; Volinia, S. MicroRNA expression signatures in solid malignancies. Cancer J. (Sudbury Mass.) 2012, 18, 238–243. [Google Scholar] [CrossRef] [PubMed]
  15. Zhang, B.; Pan, X.; Cobb, G.P.; Anderson, T.A. MicroRNAs as oncogenes and tumor suppressors. Dev. Biol. 2007, 302, 1–12. [Google Scholar] [CrossRef] [PubMed]
  16. Iorio, M.V.; Croce, C.M. MicroRNAs in cancer: Small molecules with a huge impact. J. Clin. Oncol. 2009, 27, 5848–5856. [Google Scholar] [CrossRef] [PubMed]
  17. Bentwich, I.; Avniel, A.; Karov, Y.; Aharonov, R.; Gilad, S.; Barad, O.; Barzilai, A.; Einat, P.; Einav, U.; Meiri, E.; et al. Identification of hundreds of conserved and nonconserved human microRNAs. Nat. Genet. 2005, 37, 766–770. [Google Scholar] [CrossRef] [PubMed]
  18. Carthew, R.W. Gene regulation by microRNAs. Curr. Opin. Genet. Dev. 2006, 16, 203–208. [Google Scholar] [CrossRef] [PubMed]
  19. Lee, R.C.; Feinbaum, R.L.; Ambros, V. The c. Elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell 1993, 75, 843–854. [Google Scholar] [CrossRef]
  20. Iorio, M.V.; Ferracin, M.; Liu, C.G.; Veronese, A.; Spizzo, R.; Sabbioni, S.; Magri, E.; Pedriali, M.; Fabbri, M.; Campiglio, M.; et al. MicroRNA gene expression deregulation in human breast cancer. Cancer Res. 2005, 65, 7065–7070. [Google Scholar] [CrossRef] [PubMed]
  21. Johnson, S.M.; Grosshans, H.; Shingara, J.; Byrom, M.; Jarvis, R.; Cheng, A.; Labourier, E.; Reinert, K.L.; Brown, D.; Slack, F.J. Ras is regulated by the let-7 microRNA family. Cell 2005, 120, 635–647. [Google Scholar] [CrossRef] [PubMed]
  22. Michael, M.Z.; SM, O.C.; van Holst Pellekaan, N.G.; Young, G.P.; James, R.J. Reduced accumulation of specific microRNAs in colorectal neoplasia. Mol. Cancer Res. MCR 2003, 1, 882–891. [Google Scholar] [PubMed]
  23. Bhardwaj, A.; Arora, S.; Prajapati, V.K.; Singh, S.; Singh, A.P. Cancer “stemness”- regulating microRNAs: Role, mechanisms and therapeutic potential. Curr. Drug Targets 2013, 14, 1175–1184. [Google Scholar] [CrossRef] [PubMed]
  24. Almoguera, C.; Shibata, D.; Forrester, K.; Martin, J.; Arnheim, N.; Perucho, M. Most human carcinomas of the exocrine pancreas contain mutant c-k-ras genes. Cell 1988, 53, 549–554. [Google Scholar] [CrossRef]
  25. Jiao, L.R.; Frampton, A.E.; Jacob, J.; Pellegrino, L.; Krell, J.; Giamas, G.; Tsim, N.; Vlavianos, P.; Cohen, P.; Ahmad, R.; et al. MicroRNAs targeting oncogenes are down-regulated in pancreatic malignant transformation from benign tumors. PLoS ONE 2012, 7, e32068. [Google Scholar] [CrossRef] [PubMed]
  26. 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. [Google Scholar] [CrossRef] [PubMed]
  27. Zhao, W.G.; Yu, S.N.; Lu, Z.H.; Ma, Y.H.; Gu, Y.M.; Chen, J. The mir-217 microRNA functions as a potential tumor suppressor in pancreatic ductal adenocarcinoma by targeting kras. Carcinogenesis 2010, 31, 1726–1733. [Google Scholar] [CrossRef] [PubMed]
  28. Kent, O.A.; Chivukula, R.R.; Mullendore, M.; Wentzel, E.A.; Feldmann, G.; Lee, K.H.; Liu, S.; Leach, S.D.; Maitra, A.; Mendell, J.T. Repression of the mir-143/145 cluster by oncogenic ras initiates a tumor-promoting feed-forward pathway. Genes Dev. 2010, 24, 2754–2759. [Google Scholar] [CrossRef] [PubMed]
  29. Torrisani, J.; Bournet, B.; du Rieu, M.C.; Bouisson, M.; Souque, A.; Escourrou, J.; Buscail, L.; Cordelier, P. Let-7 microRNA transfer in pancreatic cancer-derived cells inhibits in vitro cell proliferation but fails to alter tumor progression. Hum. Gene Ther. 2009, 20, 831–844. [Google Scholar] [CrossRef] [PubMed]
  30. Scarpa, A.; Capelli, P.; Mukai, K.; Zamboni, G.; Oda, T.; Iacono, C.; Hirohashi, S. Pancreatic adenocarcinomas frequently show p53 gene mutations. Am. J. Pathol. 1993, 142, 1534–1543. [Google Scholar] [PubMed]
  31. Dong, P.; Karaayvaz, M.; Jia, N.; Kaneuchi, M.; Hamada, J.; Watari, H.; Sudo, S.; Ju, J.; Sakuragi, N. Mutant p53 gain-of-function induces epithelial-mesenchymal transition through modulation of the mir-130b-zeb1 axis. Oncogene 2013, 32, 3286–3295. [Google Scholar] [CrossRef] [PubMed]
  32. Neilsen, P.M.; Noll, J.E.; Mattiske, S.; Bracken, C.P.; Gregory, P.A.; Schulz, R.B.; Lim, S.P.; Kumar, R.; Suetani, R.J.; Goodall, G.J.; et al. Mutant p53 drives invasion in breast tumors through up-regulation of mir-155. Oncogene 2013, 32, 2992–3000. [Google Scholar] [CrossRef] [PubMed]
  33. Zhang, X.J.; Ye, H.; Zeng, C.W.; He, B.; Zhang, H.; Chen, Y.Q. Dysregulation of mir-15a and mir-214 in human pancreatic cancer. J. Hematol. Oncol. 2010, 3, 46. [Google Scholar] [CrossRef] [PubMed]
  34. Soubani, O.; Ali, A.S.; Logna, F.; Ali, S.; Philip, P.A.; Sarkar, F.H. Re-expression of mir-200 by novel approaches regulates the expression of pten and mt1-mmp in pancreatic cancer. Carcinogenesis 2012, 33, 1563–1571. [Google Scholar] [CrossRef] [PubMed]
  35. Brabletz, S.; Bajdak, K.; Meidhof, S.; Burk, U.; Niedermann, G.; Firat, E.; Wellner, U.; Dimmler, A.; Faller, G.; Schubert, J.; et al. The zeb1/mir-200 feedback loop controls notch signalling in cancer cells. EMBO J. 2011, 30, 770–782. [Google Scholar] [CrossRef] [PubMed]
  36. Guo, R.; Wang, Y.; Shi, W.Y.; Liu, B.; Hou, S.Q.; Liu, L. MicroRNA mir-491–5p targeting both tp53 and bcl-xl induces cell apoptosis in sw1990 pancreatic cancer cells through mitochondria mediated pathway. Molecules (Basel Switzerland) 2012, 17, 14733–14747. [Google Scholar] [CrossRef] [PubMed]
  37. Neault, M.; Mallette, F.A.; Richard, S. Mir-137 modulates a tumor suppressor network-inducing senescence in pancreatic cancer cells. Cell Rep. 2016, 14, 1966–1978. [Google Scholar] [CrossRef] [PubMed]
  38. Okamoto, A.; Demetrick, D.J.; Spillare, E.A.; Hagiwara, K.; Hussain, S.P.; Bennett, W.P.; Forrester, K.; Gerwin, B.; Serrano, M.; Beach, D.H.; et al. Mutations and altered expression of p16ink4 in human cancer. Proc. Natl. Acad. Sci. USA 1994, 91, 11045–11049. [Google Scholar] [CrossRef] [PubMed]
  39. Lal, A.; Kim, H.H.; Abdelmohsen, K.; Kuwano, Y.; Pullmann, R., Jr.; Srikantan, S.; Subrahmanyam, R.; Martindale, J.L.; Yang, X.; Ahmed, F.; et al. P16(ink4a) translation suppressed by mir-24. PLoS ONE 2008, 3, e1864. [Google Scholar] [CrossRef] [PubMed]
  40. Venkataraman, S.; Alimova, I.; Fan, R.; Harris, P.; Foreman, N.; Vibhakar, R. MicroRNA 128a increases intracellular ros level by targeting bmi-1 and inhibits medulloblastoma cancer cell growth by promoting senescence. PLoS ONE 2010, 5, e10748. [Google Scholar] [CrossRef] [PubMed]
  41. Nakata, K.; Ohuchida, K.; Mizumoto, K.; Kayashima, T.; Ikenaga, N.; Sakai, H.; Lin, C.; Fujita, H.; Otsuka, T.; Aishima, S.; et al. MicroRNA-10b is overexpressed in pancreatic cancer, promotes its invasiveness, and correlates with a poor prognosis. Surgery 2011, 150, 916–922. [Google Scholar] [CrossRef] [PubMed]
  42. Zhang, L.; Jamaluddin, M.S.; Weakley, S.M.; Yao, Q.; Chen, C. Roles and mechanisms of microRNAs in pancreatic cancer. World J. Surg. 2011, 35, 1725–1731. [Google Scholar] [CrossRef] [PubMed]
  43. Cano, C.E.; Motoo, Y.; Iovanna, J.L. Epithelial-to-mesenchymal transition in pancreatic adenocarcinoma. Sci. World J. 2010, 10, 1947–1957. [Google Scholar] [CrossRef] [PubMed]
  44. Rachagani, S.; Macha, M.A.; Heimann, N.; Seshacharyulu, P.; Haridas, D.; Chugh, S.; Batra, S.K. Clinical implications of miRNAs in the pathogenesis, diagnosis and therapy of pancreatic cancer. Adv. Drug Deliv. Rev. 2015, 81, 16–33. [Google Scholar] [CrossRef] [PubMed]
  45. Nicolas, F.J.; Hill, C.S. Attenuation of the tgf-beta-smad signaling pathway in pancreatic tumor cells confers resistance to tgf-beta-induced growth arrest. Oncogene 2003, 22, 3698–3711. [Google Scholar] [CrossRef] [PubMed]
  46. Friess, H.; Yamanaka, Y.; Buchler, M.; Ebert, M.; Beger, H.G.; Gold, L.I.; Korc, M. Enhanced expression of transforming growth factor beta isoforms in pancreatic cancer correlates with decreased survival. Gastroenterology 1993, 105, 1846–1856. [Google Scholar] [CrossRef]
  47. Liu, L.; Nie, J.; Chen, L.; Dong, G.; Du, X.; Wu, X.; Tang, Y.; Han, W. The oncogenic role of microRNA-130a/301a/454 in human colorectal cancer via targeting smad4 expression. PLoS ONE 2013, 8, e55532. [Google Scholar] [CrossRef] [PubMed]
  48. Chen, Z.; Chen, L.Y.; Dai, H.Y.; Wang, P.; Gao, S.; Wang, K. Mir-301a promotes pancreatic cancer cell proliferation by directly inhibiting bim expression. J. Cell. Biochem. 2012, 113, 3229–3235. [Google Scholar] [CrossRef] [PubMed]
  49. Hao, J.; Zhang, S.; Zhou, Y.; Hu, X.; Shao, C. MicroRNA 483–3p suppresses the expression of dpc4/smad4 in pancreatic cancer. FEBS Lett. 2011, 585, 207–213. [Google Scholar] [CrossRef] [PubMed]
  50. Hao, J.; Zhang, S.; Zhou, Y.; Liu, C.; Hu, X.; Shao, C. MicroRNA 421 suppresses dpc4/smad4 in pancreatic cancer. Biochem. Biophys. Res. Commun. 2011, 406, 552–557. [Google Scholar] [CrossRef] [PubMed]
  51. Du, J.; Wang, J.; Tan, G.; Cai, Z.; Zhang, L.; Tang, B.; Wang, Z. Aberrant elevated microRNA-146a in dendritic cells (dc) induced by human pancreatic cancer cell line bxpc-3-conditioned medium inhibits dc maturation and activation. Med. Oncol. (Northwood Lond. Engl.) 2012, 29, 2814–2823. [Google Scholar] [CrossRef] [PubMed]
  52. 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. [Google Scholar] [CrossRef] [PubMed]
  53. Wang, C.; Liu, P.; Wu, H.; Cui, P.; Li, Y.; Liu, Y.; Liu, Z.; Gou, S. MicroRNA-323–3p inhibits cell invasion and metastasis in pancreatic ductal adenocarcinoma via direct suppression of SMAD2 and SMAD3. Oncotarget 2016, 7, 14912–14924. [Google Scholar] [PubMed]
  54. Lu, J.; Getz, G.; Miska, E.A.; Alvarez-Saavedra, E.; Lamb, J.; Peck, D.; Sweet-Cordero, A.; Ebert, B.L.; Mak, R.H.; Ferrando, A.A.; et al. MicroRNA expression profiles classify human cancers. Nature 2005, 435, 834–838. [Google Scholar] [CrossRef] [PubMed]
  55. Rosenfeld, N.; Aharonov, R.; Meiri, E.; Rosenwald, S.; Spector, Y.; Zepeniuk, M.; Benjamin, H.; Shabes, N.; Tabak, S.; Levy, A.; et al. MicroRNAs accurately identify cancer tissue origin. Nat. Biotechnol. 2008, 26, 462–469. [Google Scholar] [CrossRef] [PubMed]
  56. Visani, M.; Acquaviva, G.; Fiorino, S.; Bacchi Reggiani, M.L.; Masetti, M.; Franceschi, E.; Fornelli, A.; Jovine, E.; Fabbri, C.; Brandes, A.A.; et al. Contribution of microRNA analysis to characterisation of pancreatic lesions: A review. J. Clinical Pathol. 2015, 68, 859–869. [Google Scholar] [CrossRef] [PubMed]
  57. Kishikawa, T.; Otsuka, M.; Ohno, M.; Yoshikawa, T.; Takata, A.; Koike, K. Circulating RNAs as new biomarkers for detecting pancreatic cancer. World J. Gastroenterol. 2015, 21, 8527–8540. [Google Scholar] [CrossRef] [PubMed]
  58. Schwarzenbach, H.; Nishida, N.; Calin, G.A.; Pantel, K. Clinical relevance of circulating cell-free microRNAs in cancer. Nat. Rev. Clin. Oncol. 2014, 11, 145–156. [Google Scholar] [CrossRef] [PubMed]
  59. Abue, M.; Yokoyama, M.; Shibuya, R.; Tamai, K.; Yamaguchi, K.; Sato, I.; Tanaka, N.; Hamada, S.; Shimosegawa, T.; Sugamura, K.; et al. Circulating mir-483–3p and mir-21 is highly expressed in plasma of pancreatic cancer. Int. J. Oncol. 2015, 46, 539–547. [Google Scholar] [CrossRef] [PubMed]
  60. Li, A.; Omura, N.; Hong, S.M.; 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. [Google Scholar] [CrossRef] [PubMed]
  61. Li, A.; Yu, J.; Kim, H.; Wolfgang, C.L.; Canto, M.I.; Hruban, R.H.; 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. [Google Scholar] [CrossRef] [PubMed]
  62. 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. [Google Scholar] [CrossRef] [PubMed]
  63. Singh, S.; Chitkara, D.; Kumar, V.; Behrman, S.W.; Mahato, R.I. MiRNA profiling in pancreatic cancer and restoration of chemosensitivity. Cancer Lett. 2013, 334, 211–220. [Google Scholar] [CrossRef] [PubMed]
  64. Szafranska-Schwarzbach, A.E.; Adai, A.T.; Lee, L.S.; Conwell, D.L.; Andruss, B.F. Development of a miRNA-based diagnostic assay for pancreatic ductal adenocarcinoma. Expert Rev. Mol. Diagn. 2011, 11, 249–257. [Google Scholar] [PubMed]
  65. Lee, L.S.; Szafranska-Schwarzbach, A.E.; Wylie, D.; Doyle, L.A.; Bellizzi, A.M.; Kadiyala, V.; Suleiman, S.; Banks, P.A.; Andruss, B.F.; Conwell, D.L. Investigating microRNA expression profiles in pancreatic cystic neoplasms. Clin. Transl. Gastroenterol. 2014, 5, e47. [Google Scholar] [CrossRef] [PubMed]
  66. Bloomston, M.; Frankel, W.L.; Petrocca, F.; Volinia, S.; Alder, H.; Hagan, J.P.; Liu, C.G.; Bhatt, D.; Taccioli, C.; Croce, C.M. MicroRNA expression patterns to differentiate pancreatic adenocarcinoma from normal pancreas and chronic pancreatitis. Jama 2007, 297, 1901–1908. [Google Scholar] [CrossRef] [PubMed]
  67. Szafranska, A.E.; Davison, T.S.; John, J.; Cannon, T.; Sipos, B.; Maghnouj, A.; Labourier, E.; Hahn, S.A. MicroRNA expression alterations are linked to tumorigenesis and non-neoplastic processes in pancreatic ductal adenocarcinoma. Oncogene 2007, 26, 4442–4452. [Google Scholar] [CrossRef] [PubMed]
  68. Szafranska, A.E.; Doleshal, M.; Edmunds, H.S.; Gordon, S.; Luttges, J.; Munding, J.B.; Barth, R.J., Jr.; Gutmann, E.J.; Suriawinata, A.A.; Marc Pipas, J.; et al. Analysis of microRNAs in pancreatic fine-needle aspirates can classify benign and malignant tissues. Clin. Chem. 2008, 54, 1716–1724. [Google Scholar] [CrossRef] [PubMed]
  69. Zhang, Y.; Li, M.; Wang, H.; Fisher, W.E.; Lin, P.H.; Yao, Q.; Chen, C. Profiling of 95 microRNAs in pancreatic cancer cell lines and surgical specimens by real-time pcr analysis. World J. Surg. 2009, 33, 698–709. [Google Scholar] [CrossRef] [PubMed]
  70. Lee, E.J.; Gusev, Y.; Jiang, J.; Nuovo, G.J.; Lerner, M.R.; Frankel, W.L.; Morgan, D.L.; Postier, R.G.; Brackett, D.J.; Schmittgen, T.D. Expression profiling identifies microRNA signature in pancreatic cancer. Int. J. Cancer 2007, 120, 1046–1054. [Google Scholar] [CrossRef] [PubMed]
  71. Park, J.K.; Henry, J.C.; Jiang, J.; Esau, C.; Gusev, Y.; Lerner, M.R.; Postier, R.G.; Brackett, D.J.; Schmittgen, T.D. Mir-132 and mir-212 are increased in pancreatic cancer and target the retinoblastoma tumor suppressor. Biochem. Biophys. Res. Commun. 2011, 406, 518–523. [Google Scholar] [CrossRef] [PubMed]
  72. Jamieson, N.B.; Morran, D.C.; Morton, J.P.; Ali, A.; Dickson, E.J.; Carter, C.R.; Sansom, O.J.; Evans, T.R.; McKay, C.J.; Oien, K.A. MicroRNA molecular profiles associated with diagnosis, clinicopathologic criteria, and overall survival in patients with resectable pancreatic ductal adenocarcinoma. Clin. Cancer Res. 2012, 18, 534–545. [Google Scholar] [CrossRef] [PubMed]
  73. Piepoli, A.; Tavano, F.; Copetti, M.; Mazza, T.; Palumbo, O.; Panza, A.; di Mola, F.F.; Pazienza, V.; Mazzoccoli, G.; Biscaglia, G.; et al. MiRNA expression profiles identify drivers in colorectal and pancreatic cancers. PLoS ONE 2012, 7, e33663. [Google Scholar] [CrossRef] [PubMed]
  74. Hong, T.H.; Park, I.Y. MicroRNA expression profiling of diagnostic needle aspirates from surgical pancreatic cancer specimens. Ann. Surg. Treat. Res. 2014, 87, 290–297. [Google Scholar] [CrossRef] [PubMed]
  75. Greither, T.; Grochola, L.F.; Udelnow, A.; Lautenschlager, C.; Wurl, P.; Taubert, H. Elevated expression of microRNAs 155, 203, 210 and 222 in pancreatic tumors is associated with poorer survival. Int. J. Cancer 2010, 126, 73–80. [Google Scholar] [CrossRef] [PubMed]
  76. Panarelli, N.C.; Chen, Y.T.; Zhou, X.K.; Kitabayashi, N.; Yantiss, R.K. MicroRNA expression aids the preoperative diagnosis of pancreatic ductal adenocarcinoma. Pancreas 2012, 41, 685–690. [Google Scholar] [CrossRef] [PubMed]
  77. Papaconstantinou, I.G.; Manta, A.; Gazouli, M.; Lyberopoulou, A.; Lykoudis, P.M.; Polymeneas, G.; Voros, D. Expression of microRNAs in patients with pancreatic cancer and its prognostic significance. Pancreas 2013, 42, 67–71. [Google Scholar] [CrossRef] [PubMed]
  78. Xue, Y.; Abou Tayoun, A.N.; Abo, K.M.; Pipas, J.M.; Gordon, S.R.; Gardner, T.B.; Barth, R.J., Jr.; Suriawinata, A.A.; Tsongalis, G.J. MicroRNAs as diagnostic markers for pancreatic ductal adenocarcinoma and its precursor, pancreatic intraepithelial neoplasm. Cancer Genet. 2013, 206, 217–221. [Google Scholar] [CrossRef] [PubMed]
  79. 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. [Google Scholar] [PubMed]
  80. Wang, J.; Raimondo, M.; Guha, S.; Chen, J.; Diao, L.; Dong, X.; Wallace, M.B.; Killary, A.M.; Frazier, M.L.; Woodward, T.A.; et al. Circulating microRNAs in pancreatic juice as candidate biomarkers of pancreatic cancer. J. Cancer 2014, 5, 696–705. [Google Scholar] [CrossRef] [PubMed]
  81. Schultz, N.A.; Dehlendorff, C.; Jensen, B.V.; Bjerregaard, J.K.; Nielsen, K.R.; Bojesen, S.E.; Calatayud, D.; Nielsen, S.E.; Yilmaz, M.; Hollander, N.H.; et al. MicroRNA biomarkers in whole blood for detection of pancreatic cancer. Jama 2014, 311, 392–404. [Google Scholar] [CrossRef] [PubMed]
  82. Wang, J.; Chen, J.; Chang, P.; LeBlanc, A.; Li, D.; Abbruzzesse, J.L.; Frazier, M.L.; Killary, A.M.; Sen, S. MicroRNAs in plasma of pancreatic ductal adenocarcinoma patients as novel blood-based biomarkers of disease. Cancer Prev. Res. (Philadelphia Pa.) 2009, 2, 807–813. [Google Scholar] [CrossRef] [PubMed]
  83. Ali, S.; Almhanna, K.; Chen, W.; Philip, P.A.; Sarkar, F.H. Differentially expressed miRNAs in the plasma may provide a molecular signature for aggressive pancreatic cancer. Am. J. Transl. Res. 2010, 3, 28–47. [Google Scholar] [PubMed]
  84. Ho, A.S.; Huang, X.; Cao, H.; Christman-Skieller, C.; Bennewith, K.; Le, Q.T.; Koong, A.C. Circulating mir-210 as a novel hypoxia marker in pancreatic cancer. Transl. Oncol. 2010, 3, 109–113. [Google Scholar] [CrossRef] [PubMed]
  85. LaConti, J.J.; Shivapurkar, N.; Preet, A.; Deslattes Mays, A.; Peran, I.; Kim, S.E.; Marshall, J.L.; Riegel, A.T.; Wellstein, A. Tissue and serum microRNAs in the kras(g12d) transgenic animal model and in patients with pancreatic cancer. PLoS ONE 2011, 6, e20687. [Google Scholar] [CrossRef] [PubMed]
  86. Morimura, R.; Komatsu, S.; Ichikawa, D.; Takeshita, H.; Tsujiura, M.; Nagata, H.; Konishi, H.; Shiozaki, A.; Ikoma, H.; Okamoto, K.; et al. Novel diagnostic value of circulating mir-18a in plasma of patients with pancreatic cancer. Br. J. Cancer 2011, 105, 1733–1740. [Google Scholar] [CrossRef] [PubMed]
  87. Chen, Q.; Yang, L.; Xiao, Y.; Zhu, J.; Li, Z. Circulating microRNA-182 in plasma and its potential diagnostic and prognostic value for pancreatic cancer. Med. Oncol. (Northwood Lond. Engl.) 2014, 31, 225. [Google Scholar] [CrossRef] [PubMed]
  88. Cote, G.A.; Gore, A.J.; McElyea, S.D.; Heathers, L.E.; 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Ganepola, G.A.; Rutledge, J.R.; Suman, P.; Yiengpruksawan, A.; Chang, D.H. Novel blood-based microRNA biomarker panel for early diagnosis of pancreatic cancer. World J. Gastrointest. Oncol. 2014, 6, 22–33. [Google Scholar] [CrossRef] [PubMed]
  90. Kawaguchi, T.; Komatsu, S.; Ichikawa, D.; Morimura, R.; Tsujiura, M.; Konishi, H.; Takeshita, H.; Nagata, H.; Arita, T.; Hirajima, S.; et al. Clinical impact of circulating mir-221 in plasma of patients with pancreatic cancer. Br. J. Cancer 2013, 108, 361–369. [Google Scholar] [CrossRef] [PubMed]
  91. Kojima, M.; Sudo, H.; Kawauchi, J.; Takizawa, S.; Kondou, S.; Nobumasa, H.; Ochiai, A. MicroRNA markers for the diagnosis of pancreatic and biliary-tract cancers. PLoS ONE 2015, 10, e0118220. [Google Scholar] [CrossRef] [PubMed]
  92. Liu, R.; Chen, X.; Du, Y.; Yao, W.; Shen, L.; Wang, C.; Hu, Z.; Zhuang, R.; Ning, G.; Zhang, C.; et al. Serum microRNA expression profile as a biomarker in the diagnosis and prognosis of pancreatic cancer. Clin. Chem. 2012, 58, 610–618. [Google Scholar] [CrossRef] [PubMed]
  93. Yang, J.Y.; Sun, Y.W.; Liu, D.J.; Zhang, J.F.; 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. [Google Scholar] [PubMed]
  94. Lin, M.S.; Chen, W.C.; Huang, J.X.; Gao, H.J.; Sheng, H.H. Aberrant expression of microRNAs in serum may identify individuals with pancreatic cancer. Int. J. Clin. Exp. Med. 2014, 7, 5226–5234. [Google Scholar] [PubMed]
  95. Jung, D.E.; Wen, J.; Oh, T.; Song, S.Y. Differentially expressed microRNAs in pancreatic cancer stem cells. Pancreas 2011, 40, 1180–1187. [Google Scholar] [CrossRef] [PubMed]
  96. Park, J.K.; Lee, E.J.; Esau, C.; Schmittgen, T.D. Antisense inhibition of microRNA-21 or -221 arrests cell cycle, induces apoptosis, and sensitizes the effects of gemcitabine in pancreatic adenocarcinoma. Pancreas 2009, 38, e190–e199. [Google Scholar] [CrossRef] [PubMed]
  97. Ji, Q.; Hao, X.; Zhang, M.; Tang, W.; Yang, M.; Li, L.; Xiang, D.; Desano, J.T.; Bommer, G.T.; Fan, D.; et al. MicroRNA mir-34 inhibits human pancreatic cancer tumor-initiating cells. PLoS ONE 2009, 4, e6816. [Google Scholar] [CrossRef] [PubMed]
  98. Hasegawa, S.; Eguchi, H.; Nagano, H.; Konno, M.; Tomimaru, Y.; Wada, H.; Hama, N.; Kawamoto, K.; Kobayashi, S.; Nishida, N.; et al. MicroRNA-1246 expression associated with ccng2-mediated chemoresistance and stemness in pancreatic cancer. Br. J. Cancer 2014, 111, 1572–1580. [Google Scholar] [CrossRef] [PubMed]
  99. Ma, C.; Huang, T.; Ding, Y.C.; Yu, W.; Wang, Q.; Meng, B.; Luo, S.X. MicroRNA-200c overexpression inhibits chemoresistance, invasion and colony formation of human pancreatic cancer stem cells. Int. J. Clin. Exp. Pathol. 2015, 8, 6533–6539. [Google Scholar] [PubMed]
  100. Lu, Y.; Lu, J.; Li, X.; Zhu, H.; Fan, X.; Zhu, S.; Wang, Y.; Guo, Q.; Wang, L.; Huang, Y.; et al. Mir-200a inhibits epithelial-mesenchymal transition of pancreatic cancer stem cell. BMC Cancer 2014, 14, 85. [Google Scholar] [CrossRef] [PubMed]
  101. Mittal, A.; Chitkara, D.; Behrman, S.W.; Mahato, R.I. Efficacy of gemcitabine conjugated and miRNA-205 complexed micelles for treatment of advanced pancreatic cancer. Biomaterials 2014, 35, 7077–7087. [Google Scholar] [CrossRef] [PubMed]
  102. Sicard, F.; Gayral, M.; Lulka, H.; Buscail, L.; Cordelier, P. Targeting mir-21 for the therapy of pancreatic cancer. Mol. Ther. 2013, 21, 986–994. [Google Scholar] [CrossRef] [PubMed]
  103. Kuninty, P.R.; Bojmar, L.; Tjomsland, V.; Larsson, M.; Storm, G.; Ostman, A.; Sandstrom, P.; Prakash, J. MicroRNA-199a and -214 as potential therapeutic targets in pancreatic stellate cells in pancreatic tumor. Oncotarget 2016, 7, 1949–2553. [Google Scholar] [CrossRef] [PubMed]
  104. Nalls, D.; Tang, S.N.; Rodova, M.; Srivastava, R.K.; Shankar, S. Targeting epigenetic regulation of mir-34a for treatment of pancreatic cancer by inhibition of pancreatic cancer stem cells. PLoS ONE 2011, 6, e24099. [Google Scholar] [CrossRef] [PubMed]
  105. Pramanik, D.; Campbell, N.R.; Karikari, C.; Chivukula, R.; Kent, O.A.; Mendell, J.T.; Maitra, A. Restitution of tumor suppressor microRNAs using a systemic nanovector inhibits pancreatic cancer growth in mice. Mol. Cancer Ther. 2011, 10, 1470–1480. [Google Scholar] [CrossRef] [PubMed]
  106. Bao, B.; Ali, S.; Kong, D.; Sarkar, S.H.; Wang, Z.; Banerjee, S.; Aboukameel, A.; Padhye, S.; Philip, P.A.; Sarkar, F.H. Anti-tumor activity of a novel compound-cdf is mediated by regulating mir-21, mir-200, and pten in pancreatic cancer. PLoS ONE 2011, 6, e17850. [Google Scholar] [CrossRef] [PubMed]
  107. Ali, S.; Ahmad, A.; Banerjee, S.; Padhye, S.; Dominiak, K.; Schaffert, J.M.; Wang, Z.; Philip, P.A.; Sarkar, F.H. Gemcitabine sensitivity can be induced in pancreatic cancer cells through modulation of mir-200 and mir-21 expression by curcumin or its analogue cdf. Cancer Res. 2010, 70, 3606–3617. [Google Scholar] [CrossRef] [PubMed]
  108. Giovannetti, E.; Funel, N.; Peters, G.J.; Del Chiaro, M.; Erozenci, L.A.; Vasile, E.; Leon, L.G.; Pollina, L.E.; Groen, A.; Falcone, A.; et al. MicroRNA-21 in pancreatic cancer: Correlation with clinical outcome and pharmacologic aspects underlying its role in the modulation of gemcitabine activity. Cancer Res. 2010, 70, 4528–4538. [Google Scholar] [CrossRef] [PubMed]
  109. Hwang, J.H.; Voortman, J.; Giovannetti, E.; Steinberg, S.M.; Leon, L.G.; Kim, Y.T.; Funel, N.; Park, J.K.; Kim, M.A.; Kang, G.H.; et al. Identification of microRNA-21 as a biomarker for chemoresistance and clinical outcome following adjuvant therapy in resectable pancreatic cancer. PLoS ONE 2010, 5, e10630. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. 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. [Google Scholar] [CrossRef] [PubMed]
  111. Wang, P.; Zhuang, L.; Zhang, J.; Fan, J.; Luo, J.; Chen, H.; Wang, K.; Liu, L.; Chen, Z.; Meng, Z. The serum mir-21 level serves as a predictor for the chemosensitivity of advanced pancreatic cancer, and mir-21 expression confers chemoresistance by targeting fasl. Mol. Oncol. 2013, 7, 334–345. [Google Scholar] [CrossRef] [PubMed]
  112. Dhillon, N.; Aggarwal, B.B.; Newman, R.A.; Wolff, R.A.; Kunnumakkara, A.B.; Abbruzzese, J.L.; Ng, C.S.; Badmaev, V.; Kurzrock, R. Phase ii trial of curcumin in patients with advanced pancreatic cancer. Clin. Cancer Res. 2008, 14, 4491–4499. [Google Scholar] [CrossRef] [PubMed]
  113. Li, Y.; VandenBoom, T.G., 2nd; Kong, D.; Wang, Z.; Ali, S.; Philip, P.A.; Sarkar, F.H. Up-regulation of mir-200 and let-7 by natural agents leads to the reversal of epithelial-to-mesenchymal transition in gemcitabine-resistant pancreatic cancer cells. Cancer Res. 2009, 69, 6704–6712. [Google Scholar] [CrossRef] [PubMed]
  114. Vietsch, E.E.; van Eijck, C.H.; Wellstein, A. Circulating DNA and micro-RNA in patients with pancreatic cancer. Pancreat. Disord. Ther. 2015, 5. [Google Scholar] [CrossRef]
  115. Frampton, A.E.; Krell, J.; Jamieson, N.B.; Gall, T.M.; Giovannetti, E.; Funel, N.; Mato Prado, M.; Krell, D.; Habib, N.A.; Castellano, L.; et al. MicroRNAs with prognostic significance in pancreatic ductal adenocarcinoma: A meta-analysis. Eur. J. Cancer 2015, 51, 1389–1404. [Google Scholar] [CrossRef] [PubMed]
  116. Zhou, L.; Zhang, W.G.; Wang, D.S.; Tao, K.S.; Song, W.J.; Dou, K.F. MicroRNA-183 is involved in cell proliferation, survival and poor prognosis in pancreatic ductal adenocarcinoma by regulating bmi-1. Oncol. Rep. 2014, 32, 1734–1740. [Google Scholar] [CrossRef] [PubMed]
  117. Baraniskin, A.; Nopel-Dunnebacke, S.; Ahrens, M.; Jensen, S.G.; Zollner, H.; Maghnouj, A.; Wos, A.; Mayerle, J.; Munding, J.; Kost, D.; et al. Circulating u2 small nuclear RNA fragments as a novel diagnostic biomarker for pancreatic and colorectal adenocarcinoma. Int. J. Cancer 2013, 132, E48–E57. [Google Scholar] [CrossRef] [PubMed]
  118. Huang, C.; Yu, W.; Wang, Q.; Cui, H.; Wang, Y.; Zhang, L.; Han, F.; Huang, T. Increased expression of the lncRNA pvt1 is associated with poor prognosis in pancreatic cancer patients. Minerva Med. 2015, 106, 143–149. [Google Scholar] [PubMed]
  119. Kim, K.; Jutooru, I.; Chadalapaka, G.; Johnson, G.; Frank, J.; Burghardt, R.; Kim, S.; Safe, S. Hotair is a negative prognostic factor and exhibits pro-oncogenic activity in pancreatic cancer. Oncogene 2013, 32, 1616–1625. [Google Scholar] [CrossRef] [PubMed]
  120. Pang, E.J.; Yang, R.; Fu, X.B.; Liu, Y.F. Overexpression of long non-coding RNA malat1 is correlated with clinical progression and unfavorable prognosis in pancreatic cancer. Tumour Biol. 2015, 36, 2403–2407. [Google Scholar] [CrossRef] [PubMed]
  121. Peng, W.; Gao, W.; Feng, J. Long noncoding RNA hulc is a novel biomarker of poor prognosis in patients with pancreatic cancer. Med. Oncol. (Northwood Lond. Engl.) 2014, 31, 346. [Google Scholar] [CrossRef] [PubMed]
  122. Lu, X.; Fang, Y.; Wang, Z.; Xie, J.; Zhan, Q.; Deng, X.; Chen, H.; Jin, J.; Peng, C.; Li, H.; et al. Downregulation of gas5 increases pancreatic cancer cell proliferation by regulating cdk6. Cell Tissue Res. 2013, 354, 891–896. [Google Scholar] [CrossRef] [PubMed]
Table 1. miRNAs upregulated in pancreatic ductal adenocarcinoma (PDAC) compared to benign pancreatic pathology and/or healthy pancreas.
Table 1. miRNAs upregulated in pancreatic ductal adenocarcinoma (PDAC) compared to benign pancreatic pathology and/or healthy pancreas.
miRNASourceReference
miR-10a, miR-10b, miR-146a, miR-204, miR-372PDAC tissue[41]
miR-16, miR-21, miR-155, miR-181a, miR-181b, miR-196a, miR-210plasma[62]
miR-155, miR-181a, miR-181b, miR--181b-1, miR-181c, miR-181d, miR-21, miR-221PDAC tissue[66]
miR-196a, miR-196b, miR-203, miR-210, miR-222PDAC tissue[67]
miR-196a, miR-155, miR-143, miR-145, miR-223, miR-31PDAC tissue[68]
miR-196a, miR-221, miR-222, miR-15b, miR-95, miR-186, miR-190, miR-200bPDAC tissue[69]
miR-221, miR-181a, miR-181c, miR-155, miR-21, miR-100PDAC tissue[70]
miR-132, miR-212PDAC tissue[71]
miR-223, miR-143, miR-27a, miR-21, let-7i, miR-145, miR-142-5p, miR-142-3p, miR-10a, miR-150, miR-214, miR-107, miR-146b, miR-100, miR-23a, miR-199a-5p, miR-222, miR-155, miR-103, miR-221, miR34a, miR130a, miR-331-3p, miR-24, miR-505PDAC tissue[72]
miR-107, miR-103, miR-23a, miR-1207-5p, miR-125a-5p, miR-140-5p, miR-221, miR-143, miR-146, let-7, let-7d, let-7e, miR-145, miR-199b-3p, miR-199a-3p, miR-138-1, miR-92b, miR-181, miR-1246, miR-31, miR-155, miR-26a, miR-17, miR-23b, miR-24, miR-500, miR-331-3p, miR-939PDAC tissue[73]
miR-196a, miR-200a, miR-21, miR-27a, miR-146aPDAC tissue[74]
miR-155, miR-203, miR-210, miR-222PDAC tissue[75]
miR-21, miR-221, miR-100, miR-155, miR-181b, miR-196aPDAC tissue[76]
miR-21, miR-210, miR-221, miR-222, miR-155PDAC tissue[77]
miR-21, miR-196aPDAC tissue[78]
miR-21, miR-155pancreatic juice[79]
miR-205, miR-210, miR-492, miR-1247pancreatic juice[80]
miR-26b, miR-34a, miR-122, miR-126, miR-145, miR-150, miR-223, miR-505, miR-636, miR-885-5pwhole blood[81]
miR-483-3p, miR-21plasma[59]
miR-21, 210, 155, 196aplasma[82]
miR-21plasma[83]
miR-210plasma[84]
miR-100a, miR-10plasma[85]
miR-18aplasma[86]
miR-182plasma[87]
miR-10b, miR-30c, miR-106b, miR-132, miR-155, miR-181a, miR-181b, miR-196a, miR-212plasma[88]
miR-642b, miR-885-5p, miR-22plasma[89]
miR-221plasma[90]
miR-200a, 200bserum[60]
miR-24, miR-134, miR-146a, miR-378, miR-484, miR-628-3p, miR-1290, miR-1825serum[61]
miR-6826-5p, mi-6757-5p, miR-miR-3131, miR-1343-3p, serum[91]
miR-20a, miR-21, miR-24, miR-25, miR-99a, miR-185, miR-191serum[92]
miR-10b, miR-30c, miR-106b, miR-155, miR-181a, miR-196a, miR-212bile[88]
miR-21, miR-155stool[93]
Table 2. miRNAs downregulated in PDAC compared to benign pancreatic pathology and/or healthy pancreas.
Table 2. miRNAs downregulated in PDAC compared to benign pancreatic pathology and/or healthy pancreas.
miRNASourceReference
miR-148a, miR-148b, miR-375PDAC tissue[66]
miR-216, miR-217, miR-375PDAC tissue[67]
miR-96, miR-130b, miR-148a, miR-217, miR-375PDAC tissue[68]
miR-375PDAC tissue[69]
miR-30d, miR-381, miR-29c, miR-30a, miR-874, miR-324-3p, miR-33b, miR-30c-1, miR-139-3p, miR-887, miR-141, miR-575, miR-28-3p, miR-665, miR-494, miR- 617, miR-564, miR-217, miR-130b, miR-148a, miR-708, miR-648, miR-148b, miR-345, miR216aPDAC tissue[72]
miR-1254, miR-559, miR-1274a, let-7f-1PDAC tissue[73]
miR-217, miR-20a, miR-96PDAC tissue[74]
miR-216, miR-217PDAC tissue[75]
miR-31, miR-122, miR-145, miR-146aPDAC tissue[77]
miR-148a, miR-217PDAC tissue[78]
let-7d, miR-146aplasma[83]
miR-375plasma[90]
miR-6075, miR-4294, miR-6880-5p, miR-6799-5p, miR-125a-3p, miR-4530, miR-6836-3p, miR-4634, miR-7114-5p, miR-4476serum[91]
miR-492, miR-663aserum[94]
miR-216stool[93]

Share and Cite

MDPI and ACS Style

Diab, M.; Muqbil, I.; Mohammad, R.M.; Azmi, A.S.; Philip, P.A. The Role of microRNAs in the Diagnosis and Treatment of Pancreatic Adenocarcinoma. J. Clin. Med. 2016, 5, 59. https://doi.org/10.3390/jcm5060059

AMA Style

Diab M, Muqbil I, Mohammad RM, Azmi AS, Philip PA. The Role of microRNAs in the Diagnosis and Treatment of Pancreatic Adenocarcinoma. Journal of Clinical Medicine. 2016; 5(6):59. https://doi.org/10.3390/jcm5060059

Chicago/Turabian Style

Diab, Maria, Irfana Muqbil, Ramzi M. Mohammad, Asfar S. Azmi, and Philip A. Philip. 2016. "The Role of microRNAs in the Diagnosis and Treatment of Pancreatic Adenocarcinoma" Journal of Clinical Medicine 5, no. 6: 59. https://doi.org/10.3390/jcm5060059

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop