Roles of circRNAs in the tumour microenvironment
Tóm tắt
Tài liệu tham khảo
Zhang QY, Wang FX, Jia KK, Kong LD. Natural Product Interventions for Chemotherapy and Radiotherapy-Induced Side Effects. Front Pharmacol. 2018;9:1253. https://doi.org/10.3389/fphar.2018.01253.
Hanahan D, Coussens LM. Accessories to the crime: functions of cells recruited to the tumor microenvironment. Cancer Cell. 2012;21(3):309–22. https://doi.org/10.1016/j.ccr.2012.02.022.
Balkwill FR, Capasso M, Hagemann T. The tumor microenvironment at a glance. J Cell Sci. 2012;125(Pt 23):5591–6. https://doi.org/10.1242/jcs.116392.
Chen Y, Liu X, Yuan H, Yang Z, von Roemeling CA, Qie Y, et al. Therapeutic Remodeling of the Tumor Microenvironment Enhances Nanoparticle Delivery. Adv Sci (Weinh). 2019;6(5):1802070. https://doi.org/10.1002/advs.201802070.
Ocana MC, Martinez-Poveda B, Quesada AR, Medina MA. Metabolism within the tumor microenvironment and its implication on cancer progression: An ongoing therapeutic target. Med Res Rev. 2019;39(1):70–113. https://doi.org/10.1002/med.21511.
He Y, Liu S, Mattei J, Bunn PA Jr, Zhou C, Chan D. The combination of anti-KIR monoclonal antibodies with anti-PD-1/PD-L1 monoclonal antibodies could be a critical breakthrough in overcoming tumor immune escape in NSCLC. Drug Des Devel Ther. 2018;12:981–6. https://doi.org/10.2147/dddt.S163304.
Hameed S, Bhattarai P, Dai Z. Nanotherapeutic approaches targeting angiogenesis and immune dysfunction in tumor microenvironment. Sci China Life Sci. 2018;61(4):380–91. https://doi.org/10.1007/s11427-017-9256-1.
Patel A, Sant S. Hypoxic tumor microenvironment: Opportunities to develop targeted therapies. Biotechnol Adv. 2016;34(5):803–12. https://doi.org/10.1016/j.biotechadv.2016.04.005.
Bronisz A, Godlewski J, Wallace JA, Merchant AS, Nowicki MO, Mathsyaraja H, et al. Reprogramming of the tumour microenvironment by stromal PTEN-regulated miR-320. Nat Cell Biol. 2011;14(2):159–67. https://doi.org/10.1038/ncb2396.
Aprelikova O, Yu X, Palla J, Wei BR, John S, Yi M, et al. The role of miR-31 and its target gene SATB2 in cancer-associated fibroblasts. Cell Cycle. 2010;9(21):4387–98. https://doi.org/10.4161/cc.9.21.13674.
Chou J, Shahi P, Werb Z. microRNA-mediated regulation of the tumor microenvironment. Cell Cycle. 2013;12(20):3262–71. https://doi.org/10.4161/cc.26087.
Wu K, Zhao Z, Liu K, Zhang J, Li G, Wang L. Long noncoding RNA lnc-sox5 modulates CRC tumorigenesis by unbalancing tumor microenvironment. Cell Cycle. 2017;16(13):1295–301. https://doi.org/10.1080/15384101.2017.1317416.
Lin YH, Wu MH, Yeh CT, Lin KH. Long Non-Coding RNAs as Mediators of Tumor Microenvironment and Liver Cancer Cell Communication. Int J Mol Sci. Int J Mol Sci. 2018;19(12):3742. https://doi.org/10.3390/ijms19123742.
Paget S. The distribution of secondary growths in cancer of the breast. 1889. Cancer Metastasis Rev. 1989;8(2):98–101.
Martin M, Wei H, Lu T. Targeting microenvironment in cancer therapeutics. Oncotarget. 2016;7(32):52575–83. https://doi.org/10.18632/oncotarget.9824.
Altorki NK, Markowitz GJ, Gao D, Port JL, Saxena A, Stiles B, et al. The lung microenvironment: an important regulator of tumour growth and metastasis. Nat Rev Cancer. 2019;19(1):9–31. https://doi.org/10.1038/s41568-018-0081-9.
Danan M, Schwartz S, Edelheit S, Sorek R. Transcriptome-wide discovery of circular RNAs in Archaea. Nucleic Acids Res. 2012;40(7):3131–42. https://doi.org/10.1093/nar/gkr1009.
Ye CY, Chen L, Liu C, Zhu QH, Fan L. Widespread noncoding circular RNAs in plants. New Phytol. 2015;208(1):88–95. https://doi.org/10.1111/nph.13585.
Lu T, Cui L, Zhou Y, Zhu C, Fan D, Gong H, et al. Transcriptome-wide investigation of circular RNAs in rice. Rna. 2015;21(12):2076–87. https://doi.org/10.1261/rna.052282.115.
Fan X, Zhang X, Wu X, Guo H, Hu Y, Tang F, et al. Single-cell RNA-seq transcriptome analysis of linear and circular RNAs in mouse preimplantation embryos. Genome Biol. 2015;16:148. https://doi.org/10.1186/s13059-015-0706-1.
Shen Y, Guo X, Wang W. Identification and characterization of circular RNAs in zebrafish. FEBS Lett. 2017;591(1):213–20. https://doi.org/10.1002/1873-3468.12500.
Wang PL, Bao Y, Yee MC, Barrett SP, Hogan GJ, Olsen MN, et al. Circular RNA is expressed across the eukaryotic tree of life. PLoS One. 2014;9(6):e90859. https://doi.org/10.1371/journal.pone.0090859.
Li X, Yang L, Chen LL. The Biogenesis, Functions, and Challenges of Circular RNAs. Mol Cell. 2018;71(3):428–42. https://doi.org/10.1016/j.molcel.2018.06.034.
Dong R, Ma XK, Chen LL, Yang L. Increased complexity of circRNA expression during species evolution. RNA Biol. 2017;14(8):1064–74. https://doi.org/10.1080/15476286.2016.1269999.
Salzman J, Chen RE, Olsen MN, Wang PL, Brown PO. Cell-type specific features of circular RNA expression. PLoS Genet. 2013;9(9):e1003777. https://doi.org/10.1371/journal.pgen.1003777.
Nicolet BP, Engels S, Aglialoro F, van den Akker E, von Lindern M, Wolkers MC. Circular RNA expression in human hematopoietic cells is widespread and cell-type specific. Nucleic Acids Res. 2018;46(16):8168–80. https://doi.org/10.1093/nar/gky721.
Zhang P, Zuo Z, Shang W, Wu A, Bi R, Wu J, et al. Identification of differentially expressed circular RNAs in human colorectal cancer. Tumour Biol. 2017;39(3):1010428317694546. https://doi.org/10.1177/1010428317694546.
Westholm JO, Miura P, Olson S, Shenker S, Joseph B, Sanfilippo P, et al. Genome-wide analysis of drosophila circular RNAs reveals their structural and sequence properties and age-dependent neural accumulation. Cell Rep. 2014;9(5):1966–80. https://doi.org/10.1016/j.celrep.2014.10.062.
Zheng XB, Zhang M, Xu MQ. Detection and characterization of ciRS-7: a potential promoter of the development of cancer. Neoplasma. 2017;64(3):321–8. https://doi.org/10.4149/neo_2017_301.
Wang R, Zhang S, Chen X, Li N, Li J, Jia R, et al. CircNT5E Acts as a Sponge of miR-422a to Promote Glioblastoma Tumorigenesis. Cancer Res. 2018;78(17):4812–25. https://doi.org/10.1158/0008-5472.Can-18-0532.
Abdelmohsen K, Panda AC, Munk R, Grammatikakis I, Dudekula DB, De S, et al. Identification of HuR target circular RNAs uncovers suppression of PABPN1 translation by CircPABPN1. RNA Biol. 2017;14(3):361–9. https://doi.org/10.1080/15476286.2017.1279788.
Yang ZG, Awan FM, Du WW, Zeng Y, Lyu J. Wu, et al. The Circular RNA Interacts with STAT3, Increasing Its Nuclear Translocation and Wound Repair by Modulating Dnmt3a and miR-17 Function. Mol Ther. 2017;25(9):2062–74. https://doi.org/10.1016/j.ymthe.2017.05.022.
Zang J, Lu D, Xu A. The interaction of circRNAs and RNA binding proteins: An important part of circRNA maintenance and function. J Neurosci Res. 2018. https://doi.org/10.1002/jnr.24356.
Du WW, Yang W, Chen Y, Wu ZK, Foster FS, Yang Z, et al. Foxo3 circular RNA promotes cardiac senescence by modulating multiple factors associated with stress and senescence responses. Eur Heart J. 2017;38(18):1402–12. https://doi.org/10.1093/eurheartj/ehw001.
Du WW, Fang L, Yang W, Wu N, Awan FM, Yang Z, et al. Induction of tumor apoptosis through a circular RNA enhancing Foxo3 activity. Cell Death Differ. 2017;24(2):357–70. https://doi.org/10.1038/cdd.2016.133.
Zheng X, Chen L, Zhou Y, Wang Q, Zheng Z, Xu B, et al. A novel protein encoded by a circular RNA circPPP1R12A promotes tumor pathogenesis and metastasis of colon cancer via Hippo-YAP signaling. Mol Cancer. 2019;18(1):47. https://doi.org/10.1186/s12943-019-1010-6.
Yang Q, Du WW, Wu N, Yang W, Awan FM, Fang L, et al. A circular RNA promotes tumorigenesis by inducing c-myc nuclear translocation. Cell Death Differ. 2017;24(9):1609–20. https://doi.org/10.1038/cdd.2017.86.
Bach DH, Lee SK, Sood AK. Circular RNAs in Cancer. Mol Ther Nucleic Acids. 2019;16:118–29. https://doi.org/10.1016/j.omtn.2019.02.005.
Barbalat R, Ewald SE, Mouchess ML, Barton GM. Nucleic acid recognition by the innate immune system. Annu Rev Immunol. 2011;29:185–214. https://doi.org/10.1146/annurev-immunol-031210-101340.
Wu J, Chen ZJ. Innate immune sensing and signaling of cytosolic nucleic acids. Annu Rev Immunol. 2014;32:461–88. https://doi.org/10.1146/annurev-immunol-032713-120156.
Iurescia S, Fioretti D, Rinaldi M. Nucleic Acid Sensing Machinery: Targeting Innate Immune System for Cancer Therapy. Recent Pat Anticancer Drug Discov. 2018;13(1):2–17. https://doi.org/10.2174/1574892812666171030163804.
Bourquin C, Pommier A, Hotz C. Harnessing the immune system to fight cancer with Toll-like receptor and RIG-I-like receptor agonists. Pharmacol Res. 2019;104192. https://doi.org/10.1016/j.phrs.2019.03.001.
Maybruck BT, Pfannenstiel LW, Diaz-Montero M, Gastman BR. Tumor-derived exosomes induce CD8(+) T cell suppressors. J Immunother Cancer. 2017;5(1):65. https://doi.org/10.1186/s40425-017-0269-7.
Li P, Liu C, Yu Z, Wu M. New Insights into Regulatory T Cells: Exosome- and Non-Coding RNA-Mediated Regulation of Homeostasis and Resident Treg Cells. Front Immunol. 2016;7:574. https://doi.org/10.3389/fimmu.2016.00574.
Dai X, Chen C, Yang Q, Xue J, Chen X, Sun B, et al. Exosomal circRNA_100284 from arsenite-transformed cells, via microRNA-217 regulation of EZH2, is involved in the malignant transformation of human hepatic cells by accelerating the cell cycle and promoting cell proliferation. Cell Death Dis. 2018;9(5):454. https://doi.org/10.1038/s41419-018-0485-1.
Dou Y, Cha DJ, Franklin JL, Higginbotham JN, Jeppesen DK, Weaver AM, et al. Circular RNAs are down-regulated in KRAS mutant colon cancer cells and can be transferred to exosomes. Sci Rep. 2016;6:37982. https://doi.org/10.1038/srep37982.
Weng Q, Chen M, Li M, Zheng YF, Shao G, Fan W, et al. Global microarray profiling identified hsa_circ_0064428 as a potential immune-associated prognosis biomarker for hepatocellular carcinoma. J Med Genet. 2019;56(1):32–8. https://doi.org/10.1136/jmedgenet-2018-105440.
Zhang XL, Xu LL, Wang F. Hsa_circ_0020397 regulates colorectal cancer cell viability, apoptosis and invasion by promoting the expression of the miR-138 targets TERT and PD-L1. Cell Biol Int. 2017;41(9):1056–64. https://doi.org/10.1002/cbin.10826.
Shi MH, Xing YF, Zhang ZL, Huang JA, Chen YJ. Effect of soluble PD-L1 released by lung cancer cells in regulating the function of T lymphocytes. Zhonghua Zhong Liu Za Zhi. 2013;35(2):85–8. https://doi.org/10.3760/cma.j.issn.0253-3766.2013.02.002.
Xu C, Fillmore CM, Koyama S, Wu H, Zhao Y, Chen Z, et al. Loss of Lkb1 and Pten leads to lung squamous cell carcinoma with elevated PD-L1 expression. Cancer Cell. 2014;25(5):590–604. https://doi.org/10.1016/j.ccr.2014.03.033.
Gevensleben H, Dietrich D, Golletz C, Steiner S, Jung M, Thiesler T, et al. The Immune Checkpoint Regulator PD-L1 Is Highly Expressed in Aggressive Primary Prostate Cancer. Clin Cancer Res. 2016;22(8):1969–77. https://doi.org/10.1158/1078-0432.Ccr-15-2042.
Passardi A, Canale M, Valgiusti M, Ulivi P. Immune Checkpoints as a Target for Colorectal Cancer Treatment. Int J Mol Sci. 2017;18(6). https://doi.org/10.3390/ijms18061324.
McCall NS, Dicker AP, Lu B. Beyond Concurrent Chemoradiation: The Emerging Role of PD-1/PD-L1 Inhibitors in Stage III Lung Cancer. Clin Cancer Res. 2018;24(6):1271–6. https://doi.org/10.1158/1078-0432.Ccr-17-3269.
Sonpavde G. PD-1 and PD-L1 Inhibitors as Salvage Therapy for Urothelial Carcinoma. N Engl J Med. 2017;376(11):1073–4. https://doi.org/10.1056/NEJMe1701182.
Li D, Wang X, Yang M, Kan Q, Duan Z. miR3609 sensitizes breast cancer cells to adriamycin by blocking the programmed death-ligand 1 immune checkpoint. Exp Cell Res. 2019;380(1):20–8. https://doi.org/10.1016/j.yexcr.2019.03.025.
Xie WB, Liang LH, Wu KG, Wang LX, He X, Song C, et al. MiR-140 Expression Regulates Cell Proliferation and Targets PD-L1 in NSCLC. Cell Physiol Biochem. 2018;46(2):654–63. https://doi.org/10.1159/000488634.
Kim R, Emi M, Tanabe K. Cancer immunoediting from immune surveillance to immune escape. Immunology. 2007;121(1):1–14. https://doi.org/10.1111/j.1365-2567.2007.02587.x.
Xu B, Chen L, Li J, Zheng X, Shi L, Wu C, et al. Prognostic value of tumor infiltrating NK cells and macrophages in stage II+III esophageal cancer patients. Oncotarget. 2016;7(46):74904–16. https://doi.org/10.18632/oncotarget.12484.
Ma Y, Zhang C, Zhang B, Yu H, Yu Q. circRNA of AR-suppressed PABPC1 91 bp enhances the cytotoxicity of natural killer cells against hepatocellular carcinoma via upregulating UL16 binding protein 1. Oncol Lett. 2019;17(1):388–97. https://doi.org/10.3892/ol.2018.9606.
Ou ZL, Luo Z, Wei W, Liang S, Gao TL, Lu YB. Hypoxia-induced shedding of MICA and HIF1A-mediated immune escape of pancreatic cancer cells from NK cells: role of circ_0000977/miR-153 axis. RNA Biol. 2019:1–12. https://doi.org/10.1080/15476286.2019.1649585.
Li J, Li H, Lv X, Yang Z, Gao M, Bi Y, et al. Diagnostic performance of circular RNAs in human cancers: A systematic review and meta-analysis. Mol Genet Genomic Med. 2019:e749. https://doi.org/10.1002/mgg3.749.
Claesson-Welsh L, Welsh M. VEGFA and tumour angiogenesis. J Intern Med. 2013;273(2):114–27. https://doi.org/10.1111/joim.12019.
Cao W, Zhao Y, Wang L, Huang X. Circ0001429 regulates progression of bladder cancer through binding miR-205-5p and promoting VEGFA expression. Cancer Biomark. 2019;25(1):101–13. https://doi.org/10.3233/cbm-182380.
Meng Q, Li S, Liu Y, Zhang S, Jin J, Zhang Y, et al. Circular RNA circSCAF11 Accelerates the Glioma Tumorigenesis through the miR-421/SP1/VEGFA Axis. Mol Ther Nucleic Acids. 2019;17:669–77. https://doi.org/10.1016/j.omtn.2019.06.022.
Yang P, Qiu Z, Jiang Y, Dong L, Yang W, Gu C, et al. Silencing of cZNF292 circular RNA suppresses human glioma tube formation via the Wnt/beta-catenin signaling pathway. Oncotarget. 2016;7(39):63449–55. https://doi.org/10.18632/oncotarget.11523.
Boeckel JN, Jae N, Heumuller AW, Chen W, Boon RA, Stellos K, et al. Identification and Characterization of Hypoxia-Regulated Endothelial Circular RNA. Circ Res. 2015;117(10):884–90. https://doi.org/10.1161/circresaha.115.306319.
Li Y, Zheng F, Xiao X, Xie F, Tao D, Huang C, et al. CircHIPK3 sponges miR-558 to suppress heparanase expression in bladder cancer cells. EMBO Rep. 2017;18(9):1646–59. https://doi.org/10.15252/embr.201643581.
Barbagallo D, Caponnetto A, Brex D, Mirabella F, Barbagallo C, Lauretta G, et al. CircSMARCA5 Regulates VEGFA mRNA Splicing and Angiogenesis in Glioblastoma Multiforme Through the Binding of SRSF1. Cancers (Basel). 2019;11(2). https://doi.org/10.3390/cancers11020194.
Gilbert MR, Dignam JJ, Armstrong TS, Wefel JS, Blumenthal DT, Vogelbaum MA, et al. A randomized trial of bevacizumab for newly diagnosed glioblastoma. N Engl J Med. 2014;370(8):699–708. https://doi.org/10.1056/NEJMoa1308573.
Li CY, Ma L, Yu B. Circular RNA hsa_circ_0003575 regulates oxLDL induced vascular endothelial cells proliferation and angiogenesis. Biomed Pharmacother. 2017;95:1514–9. https://doi.org/10.1016/j.biopha.2017.09.064.
Liang G, Liu Z, Tan L, Su AN, Jiang WG, Gong C. HIF1alpha-associated circDENND4C Promotes Proliferation of Breast Cancer Cells in Hypoxic Environment. Anticancer Res. 2017;37(8):4337–43. https://doi.org/10.21873/anticanres.11827.
Ren S, Liu J, Feng Y, Li Z, He L, Li L, et al. Knockdown of circDENND4C inhibits glycolysis, migration and invasion by up-regulating miR-200b/c in breast cancer under hypoxia. J Exp Clin Cancer Res. 2019;38(1):388. https://doi.org/10.1186/s13046-019-1398-2.
Zhang H, Zhu L, Bai M, Liu Y, Zhan Y, Deng T, et al. Exosomal circRNA derived from gastric tumor promotes white adipose browning by targeting the miR-133/PRDM16 pathway. Int J Cancer. 2019;144(10):2501–15. https://doi.org/10.1002/ijc.31977.
Li P, Chen H, Chen S, Mo X, Li T, Xiao B, et al. Circular RNA 0000096 affects cell growth and migration in gastric cancer. Br J Cancer. 2017;116(5):626–33. https://doi.org/10.1038/bjc.2016.451.
He C, Huang C, Zhou R, Yu H. CircLMNB1 promotes colorectal cancer by regulating cell proliferation, apoptosis and epithelial-mesenchymal transition. Onco Targets Ther. 2019;12:6349–59. https://doi.org/10.2147/ott.S204741.
Yang J, Cong X, Ren M, Sun H, Liu T, Chen G, et al. Circular RNA hsa_circRNA_0007334 is Predicted to Promote MMP7 and COL1A1 Expression by Functioning as a miRNA Sponge in Pancreatic Ductal Adenocarcinoma. J Oncol. 2019;2019:7630894. https://doi.org/10.1155/2019/7630894.
Yu J, Xu QG, Wang ZG, Yang Y, Zhang L, Ma JZ, et al. Circular RNA cSMARCA5 inhibits growth and metastasis in hepatocellular carcinoma. J Hepatol. 2018;68(6):1214–27. https://doi.org/10.1016/j.jhep.2018.01.012.
Lee Y, El Andaloussi S, Wood MJ. Exosomes and microvesicles: extracellular vesicles for genetic information transfer and gene therapy. Hum Mol Genet. 2012;21(R1):R125–34. https://doi.org/10.1093/hmg/dds317.
Shao H, Im H, Castro CM, Breakefield X, Weissleder R, Lee H. New Technologies for Analysis of Extracellular Vesicles. Chem Rev. 2018;118(4):1917–50. https://doi.org/10.1021/acs.chemrev.7b00534.
Balaj L, Lessard R, Dai L, Cho YJ, Pomeroy SL, Breakefield XO, et al. Tumour microvesicles contain retrotransposon elements and amplified oncogene sequences. Nat Commun. 2011;2:180. https://doi.org/10.1038/ncomms1180.
Kahlert C, Melo SA, Protopopov A, Tang J, Seth S, Koch M, et al. Identification of double-stranded genomic DNA spanning all chromosomes with mutated KRAS and p53 DNA in the serum exosomes of patients with pancreatic cancer. J Biol Chem. 2014;289(7):3869–75. https://doi.org/10.1074/jbc.C113.532267.
Tetta C, Ghigo E, Silengo L, Deregibus MC, Camussi G. Extracellular vesicles as an emerging mechanism of cell-to-cell communication. Endocrine. 2013;44(1):11–9. https://doi.org/10.1007/s12020-012-9839-0.
Xu Z, Li P, Fan L, Wu M. The Potential Role of circRNA in Tumor Immunity Regulation and Immunotherapy. Front Immunol. 2018;9:9. https://doi.org/10.3389/fimmu.2018.00009.
Wang M, Zhao J, Zhang L, Wei F, Lian Y, Wu Y, et al. Role of tumor microenvironment in tumorigenesis. J Cancer. 2017;8(5):761–73. https://doi.org/10.7150/jca.17648.
Zhang H, Deng T, Ge S, Liu Y, Bai M, Zhu K, et al. Exosome circRNA secreted from adipocytes promotes the growth of hepatocellular carcinoma by targeting deubiquitination-related USP7. Oncogene. 2019;38(15):2844–59. https://doi.org/10.1038/s41388-018-0619-z.
Chen W, Jiang J, Xia W, Huang J. Tumor-Related Exosomes Contribute to Tumor-Promoting Microenvironment: An Immunological Perspective. J Immunol Res. 2017;2017:1073947. https://doi.org/10.1155/2017/1073947.
Roma-Rodrigues C, Raposo LR, Cabral R, Paradinha F, Baptista PV, Fernandes AR. Tumor Microenvironment Modulation via Gold Nanoparticles Targeting Malicious Exosomes: Implications for Cancer Diagnostics and Therapy. Int J Mol Sci. 2017;18(1). https://doi.org/10.3390/ijms18010162.
McGee HM, Jiang D, Soto-Pantoja DR, Nevler A, Giaccia AJ, Woodward WA. Targeting the Tumor Microenvironment in Radiation Oncology: Proceedings from the 2018 ASTRO-AACR Research Workshop. Clin Cancer Res. 2019;25(10):2969–74. https://doi.org/10.1158/1078-0432.Ccr-18-3781.
Karar J, Maity A. Modulating the tumor microenvironment to increase radiation responsiveness. Cancer Biol Ther. 2009;8(21):1994–2001. https://doi.org/10.4161/cbt.8.21.9988.
Chen T, Luo J, Gu Y, Huang J, Luo Q, Yang Y. Comprehensive analysis of circular RNA profiling in AZD9291-resistant non-small cell lung cancer cell lines. Thorac Cancer. 2019;10(4):930–41. https://doi.org/10.1111/1759-7714.13032.
Xiong W, Ai YQ, Li YF, Ye Q, Chen ZT, Qin JY, et al. Microarray Analysis of Circular RNA Expression Profile Associated with 5-Fluorouracil-Based Chemoradiation Resistance in Colorectal Cancer Cells. Biomed Res Int. 2017;2017:8421614. https://doi.org/10.1155/2017/8421614.
Shao F, Huang M, Meng F, Huang Q. Circular RNA Signature Predicts Gemcitabine Resistance of Pancreatic Ductal Adenocarcinoma. Front Pharmacol. 2018;9:584. https://doi.org/10.3389/fphar.2018.00584.
Sang Y, Chen B, Song X, Li Y, Liang Y, Han D, et al. circRNA_0025202 Regulates Tamoxifen Sensitivity and Tumor Progression via Regulating the miR-182-5p/FOXO3a Axis in Breast. Cancer. Mol Ther. 2019. https://doi.org/10.1016/j.ymthe.2019.05.011.
Su H, Lin F, Deng X, Shen L, Fang Y, Fei Z, et al. Profiling and bioinformatics analyses reveal differential circular RNA expression in radioresistant esophageal cancer cells. J Transl Med. 2016;14(1):225. https://doi.org/10.1186/s12967-016-0977-7.
Horsman MR, Mortensen LS, Petersen JB, Busk M, Overgaard J. Imaging hypoxia to improve radiotherapy outcome. Nat Rev Clin Oncol. 2012;9(12):674–87. https://doi.org/10.1038/nrclinonc.2012.171.
Ishii H, Iwatsuki M, Ieta K, Ohta D, Haraguchi N, Mimori K, et al. Cancer stem cells and chemoradiation resistance. Cancer Science. 2008;99(10):1871–7. https://doi.org/10.1111/j.1349-7006.2008.00914.x.
Cheng CC, Chao WT, Liao CC, Shih JH, Lai YS, Hsu YH, et al. The Roles Of Angiogenesis And Cancer Stem Cells In Sorafenib Drug Resistance In Hepatocellular Carcinoma. Oncotargets Therapy. 2019;12:8217–27. https://doi.org/10.2147/Ott.S217468.
Wu Y, Zhang Y, Niu M, Shi Y, Liu H, Yang D, et al. Whole-Transcriptome Analysis of CD133+CD144+ Cancer Stem Cells Derived from Human Laryngeal Squamous Cell Carcinoma Cells. Cell Physiol Biochem. 2018;47(4):1696–710. https://doi.org/10.1159/000490992.
Jafari GF. Circular RNA in Saliva. Adv Exp Med Biol. 2018;1087:131–9. https://doi.org/10.1007/978-981-13-1426-1_11.
Lu R, Shao Y, Ye G, Xiao B, Guo J. Low expression of hsa_circ_0006633 in human gastric cancer and its clinical significances. Tumour Biol. 2017;39(6):1010428317704175. https://doi.org/10.1177/1010428317704175.
Yin WB, Yan MG, Fang X, Guo JJ, Xiong W, Zhang RP. Circulating circular RNA hsa_circ_0001785 acts as a diagnostic biomarker for breast cancer detection. Clin Chim Acta. 2018;487:363–8. https://doi.org/10.1016/j.cca.2017.10.011.
Zhao Q, Chen S, Li T, Xiao B, Zhang X. Clinical values of circular RNA 0000181 in the screening of gastric cancer. J Clin Lab Anal. 2018;32(4):e22333. https://doi.org/10.1002/jcla.22333.
Chen S, Li T, Zhao Q, Xiao B, Guo J. Using circular RNA hsa_circ_0000190 as a new biomarker in the diagnosis of gastric cancer. Clin Chim Acta. 2017;466:167–71. https://doi.org/10.1016/j.cca.2017.01.025.
Wang Q, Chen J, Wang A, Sun L, Qian L, Zhou X, et al. Differentially expressed circRNAs in melanocytes and melanoma cells and their effect on cell proliferation and invasion. Oncol Rep. 2018;39(4):1813–24. https://doi.org/10.3892/or.2018.6263.
Wang T, Wang X, Du Q, Wu N, Liu X, Chen Y, et al. The circRNA circP4HB promotes NSCLC aggressiveness and metastasis by sponging miR-133a-5p. Biochem Biophys Res Commun. 2019;513(4):904–11. https://doi.org/10.1016/j.bbrc.2019.04.108.
Jin P, Huang Y, Zhu P, Zou Y, Shao T, Wang O. CircRNA circHIPK3 serves as a prognostic marker to promote glioma progression by regulating miR-654/IGF2BP3 signaling. Biochem Biophys Res Commun. 2018;503(3):1570–4. https://doi.org/10.1016/j.bbrc.2018.07.081.
Chen B, Wei W, Huang X, Xie X, Kong Y, Dai D, et al. circEPSTI1 as a Prognostic Marker and Mediator of Triple-Negative Breast Cancer Progression. Theranostics. 2018;8(14):4003–15. https://doi.org/10.7150/thno.24106.
Weng W, Wei Q, Toden S, Yoshida K, Nagasaka T, Fujiwara T, et al. Circular RNA ciRS-7-A Promising Prognostic Biomarker and a Potential Therapeutic Target in Colorectal Cancer. Clin Cancer Res. 2017;23(14):3918–28. https://doi.org/10.1158/1078-0432.Ccr-16-2541.
Nie WB, Zhao LM, Guo R, Wang MX, Ye FG. Circular RNA circ-NT5C2 acts as a potential novel biomarker for prognosis of osteosarcoma. Eur Rev Med Pharmacol Sci. 2018;22(19):6239–44. https://doi.org/10.26355/eurrev_201810_16030.
Ou ZL, Luo Z, Wei W, Liang S, Gao TL, Lu YB. Hypoxia-induced shedding of MICA and HIF1A-mediated immune escape of pancreatic cancer cells from NK cells: role of circ_0000977/miR-153 axis. RNA Biol. 2019;16(11):1592–603. https://doi.org/10.1080/15476286.2019.1649585.