Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Mar 4.
Published in final edited form as: J Pancreatol. 2019 Dec;2(4):147–151. doi: 10.1097/jp9.0000000000000029

Development of microRNA-based therapy for pancreatic cancer

Andrew Fesler 1, Jingfang Ju 1,*
PMCID: PMC7055707  NIHMSID: NIHMS1065325  PMID: 32133215

Abstract

Despite extensive research efforts on diagnosis and treatment, pancreatic ductal adenocarcinoma (PDAC) remains a devastating disease and the third leading cause of cancer-related death in the United States. Resistance to current therapeutic approaches is a major reason for the poor survival of pancreatic patients. In order to overcome this major challenge and improve patient outcomes, we are in desperate need of novel therapeutic approaches. PDAC chemoresistance mechanisms are complex and multifaceted. Novel therapeutics must be equipped to deal with this challenge. microRNAs (miRNAs) have emerged as strong candidates to fill this role due to their multitargeted function. miRNAs have been shown to have important roles in pancreatic cancer resistance. In this review, we summarize the recent advancement in miRNA research related to PDAC therapeutic resistance mechanisms and the potential of miRNAs as therapeutic agents for future clinical management of PDAC.

Keywords: Chemotherapy, Gemcitabine, miRNA, Pancreatic ductal adenocarcinoma, Resistance

Background

In the past several decades we have made some great progress in treating several cancers; however, there is still much more that needs to be done to have a dramatic impact on patient survival. Pancreatic ductal adenocarcinoma (PDAC) in particular remains a considerable challenge. Several factors including typically late presentation, early metastasis, and resistance to chemotherapy, all contribute to PDAC being the third deadliest cancer in the United States.[1,2] While currently patients are typically treated with surgical resection, adjuvant chemotherapy, and radiotherapy, many patients experience postoperative recurrence.[3,4] For the past 20 years, gemcitabine has been the standard first-line chemotherapeutic used to treat pancreatic cancer.[5] Over this time, the efficacy of different therapeutic regiments based around gemcitabine, have been tested including combination of gemcitabine with several other agents such as 5-fluorouracil (5-FU), oxaliplatin, cisplatin, and capecitabine.[69] Despite these efforts, the impact on patient survival has remained limited, due in part to the various resistance mechanism involved in PDAC cells. Some PDAC patients will have intrinsic resistance and never respond to therapy, while others will have acquired resistance that will develop during the course of treatment leading to recurrence.[10] Since resistance is such a major issue for treating PDAC patients, extensive research efforts have been aimed at understanding the mechanisms involved in PDAC chemoresistance. These efforts have demonstrated that PDAC chemoresistance is multifaceted. PDAC cells have altered expression of several important genes such as KRAS, p53, and BCL-2 among others. These cells also have changes in important signaling pathways such as Notch and Hedgehog.[1013] Factors in the tumor microenvironment, secreted by tumor-associated fibroblasts, also contribute to resistance.[1417] The presence of PDAC stem cells also contributes to resistance, as these cells have several characteristics that make them highly resistant to chemotherapy. These cells are slowly dividing, making them more resistant than rapidly dividing cells. These cells also have a very plastic nature, and altered metabolism that allows them to cope with the challenge of therapy.[18] The need for novel therapeutics is clear based on the poor survival for PDAC patients, only 3% for patients with metastatic disease.[1] The fact that various mechanism contribute to resistance makes microRNA (miRNA) an ideal candidate to combat this challenge as their multitargeted function may be more challenging for cancer cells to develop resistance.

miRNAs are short (18–22 nucleotides) noncoding RNAs that regulate gene expression by base pairing primarily to the 3′-untranslated region of target messenger RNA (mRNA) to inhibit translation or induce mRNA degradation. Each miRNA is able to target several different target genes, allowing them to inhibit the expression of a network of targets. Depending on the target genes they inhibit, miRNAs may promote cancer progression and resistance (onco-miRNAs) or inhibit progression and resistance (tumor suppressor miRNAs). In the past decade, it has become clear that miRNAs have important roles in all cancers including PDAC. We have also come to appreciate the potential for manipulating miRNAs for therapeutic intervention.[19]

miRNAs functions in PDAC resistance mechanisms

It is now understood that miRNAs have important functions in PDAC and dysregulation of their expression may play a role in cancer development as well as resistance. miRNAs regulate tumorigenesis, cell cycle control, apoptosis, proliferation, chemoresistance, invasion, and metastasis.[19] There are plenty of examples of the important functions miRNAs have in PDAC resistance. Some miRNAs promote chemoresistance in PDAC. miR-21 is one of the best characterized onco-miRs in many cancers. In PDAC high expression of miR-21 has been shown to inhibit the effectiveness of gemcitabine. High expression is also associated with shorter patient survival. These effects of miR-21 are a result of its targeting phosphatase and tensin homolog, and suppression of miR-21 results in cell cycle arrest, apoptosis, and enhanced sensitivity to chemotherapy.[2022] In addition to miR-21, several other miRNAs have been shown to promote resistance in PDAC. miR-320a, promotes resistance to 5-FU in PDAC by targeting programmed cell death domain 4.[23] miR-221–3p also increases PDAC resistance to 5-FU by targeting Retinoblastoma 1 (RB1). In addition to promoting chemoresistance, by targeting RB1 miR-221–3p promotes cell proliferation, migration invasion, and epithelial-to-mesenchymal transition (EMT).[24] miR-106a is another miRNA that promotes chemoresistance. In the case of miR-106a, it is expressed in exosomes from cancer-associated fibroblasts.[25] miR-155 also has chemoresistance-associated functions in PDAC involving exosomes. Gemcitabine treatment promotes miR-155 expression which induces increased production of exosomes and inhibition of apoptosis to promote resistance. miR-155 is also packaged in exosomes promoting resistance in nearby cells.[26] In PDAC stem cells, the miR-30 family promotes invasion and migration.[27]

While there are many miRNAs that promote resistance in PDAC, there are also many other miRNAs that enhance PDAC chemosensitivity. Typically, expression of these miRNA is reduced in PDAC and restoration of their expression helps to combat chemoresistance. miR-34 is a well-studied tumor suppressor miRNA that is regulated by p53. In PDAC loss of miR-34 is associated with enrichment of stem like cells, while restoration of miR-34 inhibits growth and enhances sensitivity to gemcitabine.[28,29] Similar to miR-34, miR-205 has also been shown to inhibit PDAC stem cells and promote sensitivity to gemcitabine.[30] Various studies have also shown that the miR-200 as well as the let-7 family are reduced in gemcitabine resistant cells and expression of these miRNAs promote chemosensitivity.[31] Several groups have also demonstrated the importance of miR-506 in combating chemoresistance in PDAC. Through the regulation of several different targets, miR-506 promotes apoptosis, induces cell cycle arrest and enhances chemosensitivity.[32,33] miR-15a is another miRNA with important roles in PDAC resistance. miR-15a suppresses the growth of chemoresistant PDAC cells and inhibits EMT by targeting WNT3A, FGF7 as well as BMI-1.[34,35] These are just some examples of the important functions of miRNAs in PDAC resistance (Table 1). Clearly depending on the miRNA, it may either help to promote or combat resistance. Either by inhibiting onco-miRs or reintroducing tumor suppressor miRNAs, we can leverage these functions for therapeutic effects.

Table 1.

miRNAs with roles in chemoresistance.

miRNAs that promote chemoresistance
miRNAs that promote chemosensitivity
miRNA Target gene miRNA Target gene
miR-21[20,21] PTEN,[20] RECK[20] miR-34[26,27] Notch,[27] BCL2[27]
miR-320a[22] PDCD4[22] miR-205[28] ZEB1[28]
miR-221–3p[23] RB1[23] miR-200[29] ZEB1[29]
miR-106a[24] TP53INP1[24] miR-506[30,31] SPHK1,[30] PIM3[31]
miR-155[25] TP53INP1[25] miR-15a[32,33,44] WNT3A,[32] FGF7,[32] BMI-1,[33,44] Wee1,[44] Chk1,[44] Yap-1[44]

Chk1 = checkpoint kinase 1, miRNA = microRNA.

Therapeutic potential of miRNAs in PDAC

There are several strategies to develop miRNA-based therapeutics in PDAC. One is to suppress onco-miRNAs via antisense or antagomir-based approaches, the other is to restore and replace tumor suppressor miRNAs.[36] Because miRNA can impact of multiple targets and pathways, miRNA-based therapy will have a potential to overcome the complex resistance mechanism in cancer.[37] However, with regard to the therapeutic development, there are several major hurdles (e.g., delivery and poor pharmacokinetics) that needs to be overcome.[38] A number of strategies have been developed to overcome such hurdles. Various nano-vehicles and nanoparticles have been developed and tested.[3941] However, many of these delivery vehicles cause host toxicity. Various improvements have also been developed to enhance stability of anti-miR by modification of the backbone with 2′-O-methyl group, phosphothioate, or with the locked nucleic acid.[42]

We have recently developed a novel miRNA replacement strategy by integrating the therapeutic power of chemotherapeutic agents such as 5-FU with tumor suppressor miRNAs in PDAC. It is not just a simple combination of 5-FU with tumor suppressor miR, as 5-FU alone can still have major side effects in patients, but rather, we incorporate 5-FU into tumor suppressor miR by replacing Uracil (U) with 5-FU in the guide strand. We initially developed this approach using miR-129 and miR-15a in colon cancer.[43,44] By incorporating 5-FU into miR-129 and miR-15a, we created a potent new miRNA-based drug with the power of tumor suppressive function of miRNA together with 5-FU (Fig. 1). We demonstrated these modified miNRA have some unique features such as enhanced stability and potency, with no observed host toxicity. 5-FU-modified miRNA can also be delivered to tumor cells vehicle free. This is a major advancement that may help to overcome the bottleneck of delivery that hampers nucleic acid-based therapeutics.

Figure 1.

Figure 1.

5-FU-miR-15a combined the tumor suppressive effects of miR-15a with those of 5-FU. 5-FU-miR-15a is incorporated into the RISC complex resulting in suppression of miR-15a’s target genes, Wee1, Chk1, BMI-1, and Yap-1. Suppression of these targets induces cell cycle arrest, apoptosis, and suppresses EMT. When the 5-FU-mIR-15a breaks down 5-FU is released which will lead to DNA damage and cell death. In addition the modification of the miRNA may have some novel effects resulting from the unique modification as well as its breakdown products. Chk1 = checkpoint kinase 1, EMT = epithelial-to-mesenchymal transition, 5-FU = 5-fluorouracil, miRNA = microRNA, RISC = RNA-induced silencing complex.

Our recent studies demonstrated miR-15a functions as a tumor suppressor in PDAC in vitro by inhibiting cell proliferation and impacts cell cycle control.[45] We have demonstrated that 5-FU-modified miR-15a is a potent inhibitor for PDAC both in vitro and in vivo and improves survival, either alone or in combination with gemcitabine. The effects of miR-15a are mediated through the regulation of several important target genes (Wee1, checkpoint kinase 1 [Chk1], BMI-1, and Yap-1). All of these targets are elevated in PDAC and many are good target candidates for therapeutic development in PDAC. B lymphoma Mo-MLV insertion region 1 is an oncogene associated with poor prognosis.[46,47] Yes-associated protein 1 is crucial in promoting pancreatic tumorigenesis as well as invasion, migration, and chemosensitivity.[4850] Wee1 and Chk1 are 2 key G2/M checkpoint regulators, which can affect Cdc2 activity.[51,52] These targets have been recognized as candidates for therapeutic development by the pharmaceutical industry.[51,53,54] The regulation of all of these important targets combined with the effectiveness of the 5-FU incorporation makes 5-FU-modified miR-15a an intriguing candidate for PDAC therapy.

In addition to reintroducing tumor suppressor miRNAs, suppression of onco-miRNAs also has potential. By inhibiting these miRNAs expression of their target genes can be restored which may have therapeutic effects. A recent study has demonstrated the potential of anti-miR-21. Delivery of antimiR-21 was shown to be effective in patient derived PDAC organoids and patient derived xenograft models using tandem peptide pTP-iRGD.[55]

Conclusion

There are many mechanism that contribute to both the intrinsic and acquired resistance seen in PDAC. The adaptive nature of these cells makes overcoming resistance a moving target. This is highlighted by the fact that efforts focusing on single protein coding targets have yielded little progress on PDAC drug development in the past decades. In the face of this challenge, miRNA therapeutics may be excellent therapeutic candidates. Their ability to quickly modulate the expression of multiple targets gives them the unique ability to regulate several pathways at once. This should make it more difficult for cells to adapt and overcome miRNA therapy. As a result, a number of miRNAs have shown great potential as novel therapeutics in PDAC. With advancement in miRNA research and therapeutic development, we expect that tumor suppressive miRNAs, including 5-FUmodified miRNAs will allow us to better manage clinical treatment and enhance survival and minimize toxicity for PDAC patients.

Acknowledgments

The authors apologize to those whose work has not been included in this review due to scope and space limitations. This study was supported, in part, by the National Institute of Health/ National Cancer Institute R01CA15501904 (JJ).

Footnotes

Conflicts of interest

The authors declare no conflicts of interest.

References

  • [1].Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA Cancer J Clin 2019;69:7–34. [DOI] [PubMed] [Google Scholar]
  • [2].Li D, Xie K, Wolff R, et al. Pancreatic cancer. Lancet 2004;363:1049–1057. [DOI] [PubMed] [Google Scholar]
  • [3].Wagner M, Redaelli C, Lietz M, et al. Curative resection is the single most important factor determining outcome in patients with pancreatic adenocarcinoma. Br J Surg 2004;91:586–594. [DOI] [PubMed] [Google Scholar]
  • [4].Matsuno S, Egawa S, Fukuyama S, et al. Pancreatic cancer registry in Japan: 20 years of experience. Pancreas 2004;28:219–230. [DOI] [PubMed] [Google Scholar]
  • [5].Burris HA 3rd, Moore MJ, Andersen J, et al. Improvements in survival and clinical benefit with gemcitabine as first-line therapy for patients with advanced pancreas cancer: a randomized trial. J Clin Oncol 1997;15:2403–2413. [DOI] [PubMed] [Google Scholar]
  • [6].Berlin JD, Catalano P, Thomas JP, et al. Phase III study of gemcitabine in combination with fluorouracil versus gemcitabine alone in patients with advanced pancreatic carcinoma: Eastern Cooperative Oncology Group Trial E2297. J Clin Oncol 2002;20:3270–3275. [DOI] [PubMed] [Google Scholar]
  • [7].Heinemann V, Quietzsch D, Gieseler F, et al. Randomized phase III trial of gemcitabine plus cisplatin compared with gemcitabine alone in advanced pancreatic cancer. J Clin Oncol 2006;24:3946–3952. [DOI] [PubMed] [Google Scholar]
  • [8].Louvet C, Labianca R, Hammel P, et al. Gemcitabine in combination with oxaliplatin compared with gemcitabine alone in locally advanced or metastatic pancreatic cancer: results of a GERCOR and GISCAD phase III trial. J Clin Oncol 2005;23:3509–3516. [DOI] [PubMed] [Google Scholar]
  • [9].Herrmann R, Bodoky G, Ruhstaller T, et al. Gemcitabine plus capecitabine compared with gemcitabine alone in advanced pancreatic cancer: a randomized, multicenter, phase III trial of the Swiss Group for Clinical Cancer Research and the Central European Cooperative Oncology Group. J Clin Oncol 2007;25:2212–2217. [DOI] [PubMed] [Google Scholar]
  • [10].Wang Z, Li Y, Ahmad A, et al. Pancreatic cancer: understanding and overcoming chemoresistance. Nat Rev Gastroenterol Hepatol 2011;8:27–33. [DOI] [PubMed] [Google Scholar]
  • [11].Drakaki A, Iliopoulos D. MicroRNA-gene signaling pathways in pancreatic cancer. Biomed J 2013;36:200–208. [DOI] [PubMed] [Google Scholar]
  • [12].Yu S, Lu Z, Liu C, et al. miRNA-96 suppresses KRAS and functions as a tumor suppressor gene in pancreatic cancer. Cancer Res 2010;70:6015–6025. [DOI] [PubMed] [Google Scholar]
  • [13].Pai P, Rachagani S, Are C, et al. Prospects of miRNA-based therapy for pancreatic cancer. Curr Drug Targets 2013;14:1101–1109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [14].Jacobetz MA, Chan DS, Neesse A, et al. Hyaluronan impairs vascular function and drug delivery in a mouse model of pancreatic cancer. Gut 2013;62:112–120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [15].Provenzano PP, Cuevas C, Chang AE, et al. Enzymatic targeting of the stroma ablates physical barriers to treatment of pancreatic ductal adenocarcinoma. Cancer Cell 2012;21:418–429. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [16].Rhim AD, Oberstein PE, Thomas DH, et al. Stromal elements act to restrain, rather than support, pancreatic ductal adenocarcinoma. Cancer Cell 2014;25:735–747. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [17].Biffi G, Oni TE, Spielman B, et al. IL1-induced JAK/STAT signaling is antagonized by TGFbeta to shape CAF heterogeneity in pancreatic ductal adenocarcinoma. Cancer Discov 2019;9:282–301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [18].Biancur DE, Kimmelman AC. The plasticity of pancreatic cancer metabolism in tumor progression and therapeutic resistance. Biochim Biophys Acta Rev Cancer 2018;1870:67–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [19].Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 2004;116:281–297. [DOI] [PubMed] [Google Scholar]
  • [20].Giovannetti E, Funel N, Peters GJ, et al. MicroRNA-21 in pancreatic cancer: correlation with clinical outcome and pharmacologic aspects underlying its role in the modulation of gemcitabine activity. Cancer Res 2010;70:4528–4538. [DOI] [PubMed] [Google Scholar]
  • [21].Park JK, Lee EJ, Esau C, et al. 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. [DOI] [PubMed] [Google Scholar]
  • [22].Hwang JH, Voortman J, Giovannetti E, et al. Identification of microRNA-21 as a biomarker for chemoresistance and clinical outcome following adjuvant therapy in resectable pancreatic cancer. PLoS One 2010;5:e10630. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [23].Wang W, Zhao L, Wei X, et al. MicroRNA-320a promotes 5-FU resistance in human pancreatic cancer cells. Sci Rep 2016;6:27641. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [24].Zhao L, Zou D, Wei X, et al. MiRNA-221–3p desensitizes pancreatic cancer cells to 5-fluorouracil by targeting RB1. Tumour Biol 2016; 37:16053–16063. [DOI] [PubMed] [Google Scholar]
  • [25].Fang Y, Zhou W, Rong Y, et al. Exosomal miRNA-106b from cancer-associated fibroblast promotes gemcitabine resistance in pancreatic cancer. Exp Cell Res 2019;383:111543. [DOI] [PubMed] [Google Scholar]
  • [26].Mikamori M, Yamada D, Eguchi H, et al. MicroRNA-155 controls exosome synthesis and promotes gemcitabine resistance in pancreatic ductal adenocarcinoma. Sci Rep 2017;7:42339. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [27].Tsukasa K, Ding Q, Miyazaki Y, et al. miR-30 family promotes migratory and invasive abilities in CD133(+) pancreatic cancer stem-like cells. Hum Cell 2016;29:130–137. [DOI] [PubMed] [Google Scholar]
  • [28].Rokhlin OW, Scheinker VS, Taghiyev AF, et al. MicroRNA-34 mediates AR-dependent p53-induced apoptosis in prostate cancer. Cancer Biol Ther 2008;7:1288–1296. [DOI] [PubMed] [Google Scholar]
  • [29].Ji Q, Hao X, Zhang M, et al. MicroRNA miR-34 inhibits human pancreatic cancer tumor-initiating cells. PLoS One 2009;4:e6816. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [30].Chaudhary AK, Mondal G, Kumar V, et al. Chemosensitization and inhibition of pancreatic cancer stem cell proliferation by overexpression of microRNA-205. Cancer Lett 2017;402:1–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [31].Li Y, VandenBoom TG 2nd, Kong D, et al. Up-regulation of miR-200 and let-7 by natural agents leads to the reversal of epithelial-to-mesenchymal transition in gemcitabine-resistant pancreatic cancer cells. Cancer Res 2009;69:6704–6712. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [32].Li J, Wu H, Li W, et al. Downregulated miR-506 expression facilitates pancreatic cancer progression and chemoresistance via SPHK1/Akt/NF-kappaB signaling. Oncogene 2016;35:5501–5514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [33].Du J, Zheng X, Cai S, et al. MicroRNA-506 participates in pancreatic cancer pathogenesis by targeting PIM3. Mol Med Rep 2015;12:5121–5126. [DOI] [PubMed] [Google Scholar]
  • [34].Zhang XJ, Ye H, Zeng CW, et al. Dysregulation of miR-15a and miR-214 in human pancreatic cancer. J Hematol Oncol 2010;3:46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [35].Guo S, Xu X, Tang Y, et al. miR-15a inhibits cell proliferation and epithelial to mesenchymal transition in pancreatic ductal adenocarcinoma by down-regulating Bmi-1 expression. Cancer Lett 2014;344: 40–46. [DOI] [PubMed] [Google Scholar]
  • [36].Fesler A, Guo S, Liu H, et al. Overcoming chemoresistance in cancer stem cells with the help of microRNAs in colorectal cancer. Epigenomics 2017;9:793–796. [DOI] [PubMed] [Google Scholar]
  • [37].Fesler A, Liu H, Wu N, et al. Autophagy regulated by miRNAs in colorectal cancer progression and resistance. Cancer Transl Med 2017;3:96–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [38].Garzon R, Marcucci G, Croce CM. Targeting microRNAs in cancer: rationale, strategies and challenges. Nat Rev Drug Discov 2010;9:775–789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [39].Rai K, Takigawa N, Ito S, et al. Liposomal delivery of microRNA-7-expressing plasmid overcomes epidermal growth factor receptor tyrosine kinase inhibitor-resistance in lung cancer cells. Mol Cancer Ther 2011;10:1720–1727. [DOI] [PubMed] [Google Scholar]
  • [40].Griveau A, Bejaud J, Anthiya S, et al. Silencing of miR-21 by locked nucleic acid-lipid nanocapsule complexes sensitize human glioblastoma cells to radiation-induced cell death. Int J Pharm 2013;454: 765–774. [DOI] [PubMed] [Google Scholar]
  • [41].Yang YP, Chien Y, Chiou GY, et al. Inhibition of cancer stem cell-like properties and reduced chemoradioresistance of glioblastoma using microRNA145 with cationic polyurethane-short branch PEI. Biomaterials 2012;33:1462–1476. [DOI] [PubMed] [Google Scholar]
  • [42].Pecot CV, Calin GA, Coleman RL, et al. RNA interference in the clinic: challenges and future directions. Nat Rev Cancer 2011;11:59–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [43].Wu N, Fesler A, Liu H, et al. Development of novel miR-129 mimics with enhanced efficacy to eliminate chemoresistant colon cancer stem cells. Oncotarget 2018;9:8887–8897. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [44].Fesler A, Liu H, Ju J. Modified miR-15a has therapeutic potential for improving treatment of advanced stage colorectal cancer through inhibition of BCL2, BMI1, YAP1 and DCLK1. Oncotarget 2018;9: 2367–2383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [45].Guo S, Fesler A, Huang W, et al. Functional significance and therapeutic potential of mir-15a mimic in pancreatic ductal adenocarcinoma. Cold Spring Harbor Lab bioRxiv 2019;716738. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [46].Wu C, Zheng X, Li X, et al. Reduction of gastric cancer proliferation and invasion by miR-15a mediated suppression of Bmi-1 translation. Oncotarget 2016;7:14522–14536. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [47].Wang MC, Li CL, Cui J, et al. BMI-1, a promising therapeutic target for human cancer. Oncology Lett 2015;10:583–588. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [48].Wang P, Wei D, Zhang H, et al. PKCι and YAP1 are crucial in promoting pancreatic tumorigenesis. Oncotarget 2018;9:32736–32750. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [49].Kapoor A, Yao W, Ying H, et al. Yap1 activation enables bypass of oncogenic Kras addiction in pancreatic cancer. Cell 2014;158:185–197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [50].Gruber R, Panayiotou R, Nye E, et al. YAP1 and TAZ control pancreatic cancer initiation in mice by direct up-regulation of JAK-STAT3 signaling. Gastroenterology 2016;151:526–539. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [51].Chung S, Vail PJ, Witkiewicz AK, et al. Coordinately targeting cell cycle checkpoint functions in integrated models of pancreatic cancer. Clin Cancer Res 2019;25:2290–2304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [52].Parsels LA, Parsels JD, Tanska DM, et al. The contribution of DNA replication stress marked by high-intensity, pan-nuclear γH2AX staining to chemosensitization by CHK1 and WEE1 inhibitors. Cell Cycle 2018;17:1076–1086. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [53].Pilie PG, Tang C, Mills GB, et al. State-of-the-art strategies for targeting the DNA damage response in cancer. Nat Rev Clin Oncol 2019;16:81–104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [54].Rajeshkumar NV, De Oliveira E, Ottenhof N, et al. MK-1775, a potent Wee1 inhibitor, synergizes with gemcitabine to achieve tumor regressions, selectively in p53-deficient pancreatic cancer xenografts. Clin Cancer Res 2011;17:2799–2806. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [55].Gilles ME, Hao L, Huang L, et al. Personalized RNA medicine for pancreatic cancer. Clin Cancer Res 2018;24:1734–1747. [DOI] [PubMed] [Google Scholar]

RESOURCES