Generic placeholder image

Current Pharmaceutical Design

Editor-in-Chief

ISSN (Print): 1381-6128
ISSN (Online): 1873-4286

Review Article

Insight into RNA-based Therapies for Ovarian Cancer

Author(s): Vahideh Keyvani, Reihaneh Alsadat Mahmoudian, Samaneh Mollazadeh, Nahid Kheradmand, Elnaz Ghorbani, Majid Khazaei*, Ibrahim Saeed Al-Hayawi, Seyed Mahdi Hassanian, Gordon A. Ferns, Amir Avan* and Kazem Anvari*

Volume 29, Issue 34, 2023

Published on: 01 November, 2023

Page: [2692 - 2701] Pages: 10

DOI: 10.2174/0113816128270476231023052228

Price: $65

Abstract

Ovarian cancer (OC) is one of the most common malignancies in women and is associated with poor outcomes. The treatment for OC is often associated with resistance to therapies and hence this has stimulated the search for alternative therapeutic approaches, including RNA-based therapeutics. However, this approach has some challenges that include RNA degradation. To solve this critical issue, some novel delivery systems have been proposed. In current years, there has been growing interest in the improvement of RNAbased therapeutics as a promising approach to target ovarian cancer and improve patient outcomes. This paper provides a practical insight into the use of RNA-based therapeutics in ovarian cancers, highlighting their potential benefits, challenges, and current research progress. RNA-based therapeutics offer a novel and targeted approach to treat ovarian cancer by exploiting the unique characteristics of RNA molecules. By targeting key oncogenes or genes responsible for drug resistance, siRNAs can effectively inhibit tumor growth and sensitize cancer cells to conventional therapies. Furthermore, messenger RNA (mRNA) vaccines have emerged as a revolutionary tool in cancer immunotherapy. MRNA vaccines can be designed to encode tumor-specific antigens, stimulating the immune system to distinguish and eliminate ovarian cancer cells. A nano-based delivery platform improves the release of loaded RNAs to the target location and reduces the off-target effects. Additionally, off-target effects and immune responses triggered by RNA molecules necessitate careful design and optimization of these therapeutics. Several preclinical and clinical researches have shown promising results in the field of RNA-based therapeutics for ovarian cancer. In a preclinical study, siRNA-mediated silencing of the poly (ADP-ribose) polymerase 1 (PARP1) gene, involved in DNA repair, sensitized ovarian cancer cells to PARP inhibitors, leading to enhanced therapeutic efficacy. In clinical trials, mRNA-based vaccines targeting tumor-associated antigens have demonstrated safety and efficacy in stimulating immune responses in ovarian cancer patients. In aggregate, RNA-based therapeutics represent a promising avenue for the therapy of ovarian cancers. The ability to specifically target oncogenes or stimulate immune responses against tumor cells holds great potential for improving patient outcomes. However, further research is needed to address challenges related to delivery, permanence, and off-target effects. Clinical trials assessing the care and effectiveness of RNAbased therapeutics in larger patient cohorts are warranted. With continued advancements in the field, RNAbased therapeutics have the potential to develop the management of ovarian cancer and provide new hope for patients.

Keywords: Ovarian cancer, RNA-based therapeutics, nanocarriers, siRNA, mRNA vaccine, nano-based delivery.

[1]
Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2018; 68(6): 394-424.
[http://dx.doi.org/10.3322/caac.21492] [PMID: 30207593]
[2]
Sant M, Chirlaque Lopez MD, Agresti R, et al. Survival of women with cancers of breast and genital organs in Europe 1999-2007: Results of the EUROCARE-5 study. Eur J Cancer 2015; 51(15): 2191-205.
[http://dx.doi.org/10.1016/j.ejca.2015.07.022] [PMID: 26421822]
[3]
Lukanović D, Herzog M, Kobal B, Černe K. The contribution of copper efflux transporters ATP7A and ATP7B to chemoresistance and personalized medicine in ovarian cancer. Biomed Pharmacother 2020; 129: 110401.
[http://dx.doi.org/10.1016/j.biopha.2020.110401] [PMID: 32570116]
[4]
Brett MR, Brett MR, Jennifer BP, Thomas AS, Jennifer BP, Thomas AS. Epidemiology of ovarian cancer: A review. Cancer Biol Med 2017; 14(1): 9-32.
[http://dx.doi.org/10.20892/j.issn.2095-3941.2016.0084] [PMID: 28443200]
[5]
Alberts B, Johnson A, Lewis J, Raff M, Roberts K, Walter P. The extracellular matrix of animals. Molecular Biology of the Cell. 4th ed. Garland Science 2002.
[6]
Colombo N, Sessa C, du Bois A, et al. ESMO-ESGO consensus conference recommendations on ovarian cancer: Pathology and molecular biology, early and advanced stages, borderline tumours and recurrent disease. Ann Oncol 2019; 30(5): 672-705.
[http://dx.doi.org/10.1093/annonc/mdz062] [PMID: 31046081]
[7]
Cornelison R, Llaneza D, Landen C. Emerging therapeutics to overcome chemoresistance in epithelial ovarian cancer: A mini-review. Int J Mol Sci 2017; 18(10): 2171.
[http://dx.doi.org/10.3390/ijms18102171] [PMID: 29057791]
[8]
Liu X, Chan D, Ngan H. Mechanisms of chemoresistance in human ovarian cancer at a glance. Gynecol Obstet 2012; 2: 3.
[9]
Zahreddine H, Borden KLB. Mechanisms and insights into drug resistance in cancer. Front Pharmacol 2013; 4: 28.
[http://dx.doi.org/10.3389/fphar.2013.00028] [PMID: 23504227]
[10]
Škof E, Cerar O. Ovarian cancer: Resistance to treatment. ME DI CIN SKI RAZ GLE DI 2015; 60(1): 55-64.
[11]
Vodnik L. Ovarian cancer: Resistance to treatment. Med Razgl 2021; 60(1): 55-64.
[12]
Ashique S, Sandhu NK, Chawla V, Chawla PA. Targeted drug delivery: Trends and perspectives. Curr Drug Deliv 2021; 18(10): 1435-55.
[http://dx.doi.org/10.2174/1567201818666210609161301] [PMID: 34151759]
[13]
Ashique S, Almohaywi B, Haider N, et al. siRNA-based nanocarriers for targeted drug delivery to control breast cancer. Adv Cancer Biol - Metastasis 2022; 4: 100047.
[http://dx.doi.org/10.1016/j.adcanc.2022.100047]
[14]
Ashique S, Upadhyay A, Kumar N, Chauhan S, Mishra N. Metabolic syndromes responsible for cervical cancer and advancement of nanocarriers for efficient targeted drug delivery- A review. Adv Cancer Bio 2022; 4: 100041.
[http://dx.doi.org/10.1016/j.adcanc.2022.100041]
[15]
Blagden SP. Harnessing pandemonium: The clinical implications of tumor heterogeneity in ovarian cancer. Front Oncol 2015; 5: 149.
[http://dx.doi.org/10.3389/fonc.2015.00149] [PMID: 26175968]
[16]
Hibbs K, Skubitz KM, Pambuccian SE, et al. Differential gene expression in ovarian carcinoma: Identification of potential biomarkers. Am J Pathol 2004; 165(2): 397-414.
[http://dx.doi.org/10.1016/S0002-9440(10)63306-8] [PMID: 15277215]
[17]
Fletcher NM, Belotte J, Saed MG, et al. Specific point mutations in key redox enzymes are associated with chemoresistance in epithelial ovarian cancer. Free Radic Biol Med 2017; 102: 122-32.
[http://dx.doi.org/10.1016/j.freeradbiomed.2016.11.028] [PMID: 27890641]
[18]
Rojas V, Hirshfield K, Ganesan S, Rodriguez-Rodriguez L. Molecular characterization of epithelial ovarian cancer: Implications for diagnosis and treatment. Int J Mol Sci 2016; 17(12): 2113.
[http://dx.doi.org/10.3390/ijms17122113] [PMID: 27983698]
[19]
Ramus SJ, Vierkant RA, Johnatty SE, et al. Consortium analysis of 7 candidate SNPs for ovarian cancer. Int J Cancer 2008; 123(2): 380-8.
[http://dx.doi.org/10.1002/ijc.23448] [PMID: 18431743]
[20]
Noske A, Kaszubiak A, Weichert W, et al. Specific inhibition of AKT2 by RNA interference results in reduction of ovarian cancer cell proliferation: Increased expression of AKT in advanced ovarian cancer. Cancer Lett 2007; 246(1-2): 190-200.
[http://dx.doi.org/10.1016/j.canlet.2006.02.018] [PMID: 16584837]
[21]
Sethi G, Pathak HB, Zhang H, et al. An RNA interference lethality screen of the human druggable genome to identify molecular vulnerabilities in epithelial ovarian cancer. PLoS One 2012; 7(10): e47086.
[http://dx.doi.org/10.1371/journal.pone.0047086]
[22]
Keyvani V, Nezhad SRK, Moghbeli M, Mollazadeh S, Abbaszadegan MR. Isolation and eradication of ovarian CD44+ cancer stem cells via Notch signaling pathway mediated by ectopic silence of MAML1. Iran Red Crescent Med J 2022; 24(4).
[23]
Zeng P, Wagoner HA, Pescovitz OH, Steinmetz R. RNA interference (RNAi) for extracellular signal-regulated kinase 1 (ERK1) alone is sufficient to suppress cell viability in ovarian cancer cells. Cancer Biol Ther 2005; 4(9): 961-7.
[http://dx.doi.org/10.4161/cbt.4.9.1912] [PMID: 16138005]
[24]
Chen J, Liu X, Zhang J, Zhao Y. Targeting HMGB1 inhibits ovarian cancer growth and metastasis by lentivirus-mediated RNA interference. J Cell Physiol 2012; 227(11): 3629-38.
[http://dx.doi.org/10.1002/jcp.24069] [PMID: 22331597]
[25]
Zhang J, Zhou S, Tang L, et al. WAVE1 gene silencing via RNA interference reduces ovarian cancer cell invasion, migration and proliferation. Gynecol Oncol 2013; 130(2): 354-61.
[http://dx.doi.org/10.1016/j.ygyno.2013.05.005] [PMID: 23680521]
[26]
Stahel RA, Zangemeister-Wittke U. Antisense oligonucleotides for cancer therapy-an overview. Lung Cancer 2003; 41(S1): 81-8.
[http://dx.doi.org/10.1016/S0169-5002(03)00147-8] [PMID: 12867066]
[27]
McPhillips F, Mullen P, Monia BP, et al. Association of c-Raf expression with survival and its targeting with antisense oligonucleotides in ovarian cancer. Br J Cancer 2001; 85(11): 1753-8.
[http://dx.doi.org/10.1054/bjoc.2001.2139] [PMID: 11742498]
[28]
Popadiuk CM, Xiong J, Wells MG, et al. Antisense suppression of pygopus2 results in growth arrest of epithelial ovarian cancer. Clin Cancer Res 2006; 12(7): 2216-23.
[http://dx.doi.org/10.1158/1078-0432.CCR-05-2433] [PMID: 16609037]
[29]
Masanek U, Stammler G, Volm M. Modulation of multidrug resistance in human ovarian cancer cell lines by inhibition of P-glycoprotein 170 and PKC isoenzymes with antisense oligonucleotides. J Exp Ther Oncol 2002; 2(1): 37-41.
[http://dx.doi.org/10.1046/j.1359-4117.2002.01004.x] [PMID: 12415618]
[30]
Tang J, Li J, Zeng G, et al. Antisense oligonucleotide suppression of human IGF-1R inhibits the growth and survival of in vitro cultured epithelial ovarian cancer cells. J Ovarian Res 2013; 6(1): 71.
[http://dx.doi.org/10.1186/1757-2215-6-71] [PMID: 24103397]
[31]
Henri JL, Macdonald J, Strom M, Duan W, Shigdar S. Aptamers as potential therapeutic agents for ovarian cancer. Biochimie 2018; 145: 34-44.
[http://dx.doi.org/10.1016/j.biochi.2017.12.001] [PMID: 29224849]
[32]
Pi F, Zhang H, Li H, et al. RNA nanoparticles harboring annexin A2 aptamer can target ovarian cancer for tumor-specific doxorubicin delivery. Nanomedicine 2017; 13(3): 1183-93.
[http://dx.doi.org/10.1016/j.nano.2016.11.015] [PMID: 27890659]
[33]
Lamberti I, Scarano S, Esposito CL, et al. In vitro selection of RNA aptamers against CA125 tumor marker in ovarian cancer and its study by optical biosensing. Methods 2016; 97: 58-68.
[http://dx.doi.org/10.1016/j.ymeth.2015.10.022] [PMID: 26542762]
[34]
Zhu G, Zhang H, Jacobson O, et al. Combinatorial screening of DNA aptamers for molecular imaging of HER2 in cancer. Bioconjug Chem 2017; 28(4): 1068-75.
[http://dx.doi.org/10.1021/acs.bioconjchem.6b00746] [PMID: 28122449]
[35]
Zheng J, Zhao S, Yu X, Huang S, Liu HY. Simultaneous targeting of CD44 and EpCAM with a bispecific aptamer effectively inhibits intraperitoneal ovarian cancer growth. Theranostics 2017; 7(5): 1373-88.
[http://dx.doi.org/10.7150/thno.17826] [PMID: 28435472]
[36]
Yan RL, Qian XH, Xin XY, et al. Experimental study of anti-VEGF hairpin ribozyme gene inhibiting expression of VEGF and proliferation of ovarian cancer cells. Chin J Cancer 2002; 21(1): 39-44.
[PMID: 12500395]
[37]
Materna V, Liedert B, Thomale J, Lage H. Protection of platinum-DNA adduct formation and reversal of cisplatin resistance by anti-MRP2 hammerhead ribozymes in human cancer cells. Int J Cancer 2005; 115(3): 393-402.
[http://dx.doi.org/10.1002/ijc.20899] [PMID: 15688364]
[38]
Yang XK, Xing H, Gao QL, et al. Mdr-1 ribozyme in the reversal of multidrug resistance in human ovarian cancer. Zhonghua Zhong Liu Za Zhi 2003; 25(5): 425-8.
[PMID: 14575561]
[39]
Tekur S, Ho SM. Ribozyme-mediated downregulation of human metallothionein IIa induces apoptosis in human prostate and ovarian cancer cell lines. Mol Carcinog 2002; 33(1): 44-55.
[http://dx.doi.org/10.1002/mc.10017]
[40]
Worku T, Bhattarai D, Ayers D, et al. Long non-coding RNAs: The new horizon of gene regulation in ovarian cancer. Cell Physiol Biochem 2017; 44(3): 948-66.
[http://dx.doi.org/10.1159/000485395] [PMID: 29179183]
[41]
Wu H, Shang X, Shi Y, et al. Genetic variants of lncRNA HOTAIR and risk of epithelial ovarian cancer among Chinese women. Oncotarget 2106; 7(27): 41047-52.
[http://dx.doi.org/10.18632/oncotarget.8535] [PMID: 27166268]
[42]
Qiu H, Wang X, Guo R, et al. HOTAIR rs920778 polymorphism is associated with ovarian cancer susceptibility and poor prognosis in a Chinese population. Future Oncol 2017; 13(4): 347-55.
[http://dx.doi.org/10.2217/fon-2016-0290] [PMID: 27690631]
[43]
Richards EJ, Permuth-Wey J, Li Y, et al. A functional variant in HOXA11-AS, a novel long non-coding RNA, inhibits the oncogenic phenotype of epithelial ovarian cancer. Oncotarget 2015; 6(33): 34745-57.
[http://dx.doi.org/10.18632/oncotarget.5784] [PMID: 26430965]
[44]
Hu X, Feng Y, Zhang D, et al. A functional genomic approach identifies FAL1 as an oncogenic long noncoding RNA that associates with BMI1 and represses p21 expression in cancer. Cancer Cell 2014; 26(3): 344-57.
[http://dx.doi.org/10.1016/j.ccr.2014.07.009] [PMID: 25203321]
[45]
Meryet-Figuière M, Lambert B, Gauduchon P, et al. An overview of long non-coding RNAs in ovarian cancers. Oncotarget 2016; 7(28): 44719-34.
[http://dx.doi.org/10.18632/oncotarget.8089] [PMID: 26992233]
[46]
Özeş AR, Miller DF, Özeş ON, et al. NF-κB-HOTAIR axis links DNA damage response, chemoresistance and cellular senescence in ovarian cancer. Oncogene 2016; 35(41): 5350-61.
[http://dx.doi.org/10.1038/onc.2016.75] [PMID: 27041570]
[47]
Zhou Y, Xu X, Lv H, et al. The long noncoding RNA MALAT-1 is highly expressed in ovarian cancer and induces cell growth and migration. PLoS One 2016; 11(5): e0155250.
[http://dx.doi.org/10.1371/journal.pone.0155250] [PMID: 27227769]
[48]
Liu S, Jiang X, Li W, Cao D, Shen K, Yang J. Inhibition of the long non-coding RNA MALAT1 suppresses tumorigenicity and induces apoptosis in the human ovarian cancer SKOV3 cell line. Oncol Lett 2016; 11(6): 3686-92.
[http://dx.doi.org/10.3892/ol.2016.4435] [PMID: 27313681]
[49]
Iorio MV, Visone R, Di Leva G, et al. MicroRNA signatures in human ovarian cancer. Cancer Res 2007; 67(18): 8699-707.
[http://dx.doi.org/10.1158/0008-5472.CAN-07-1936] [PMID: 17875710]
[50]
Dahiya N, Sherman-Baust CA, Wang TL, et al. MicroRNA expression and identification of putative miRNA targets in ovarian cancer. PLoS One 2008; 3(6): e2436.
[http://dx.doi.org/10.1371/journal.pone.0002436] [PMID: 18560586]
[51]
Ganesan S. Breaking satellite silence: Human satellite II RNA expression in ovarian cancer. J Clin Invest 2022; 132(16): e161981.
[http://dx.doi.org/10.1172/JCI161981] [PMID: 35968784]
[52]
Hou W, Zhang Y. Circ_0025033 promotes the progression of ovarian cancer by activating the expression of LSM4 via targeting miR-184. Pathol Res Pract 2021; 217: 153275.
[http://dx.doi.org/10.1016/j.prp.2020.153275] [PMID: 33285422]
[53]
Chishti N, Jagwani S, Dhamecha D, Jalalpure S, Dehghan MH. Preparation, optimization, and in vivo evaluation of nanoparticle-based formulation for pulmonary delivery of anticancer drug. Medicina 2019; 55(6): 294.
[http://dx.doi.org/10.3390/medicina55060294] [PMID: 31226865]
[54]
Tian Z, Liang G, Cui K, et al. Insight into the prospects for RNAi therapy of cancer. Front Pharmacol 2021; 12: 644718.
[http://dx.doi.org/10.3389/fphar.2021.644718] [PMID: 33796026]
[55]
Fluiter K, Mook OR, Baas F. The therapeutic potential of LNA- modified siRNAs: reduction of off-target effects by chemical modification of the siRNA sequence. Methods Mol Biol 2009; 487: 189-203.
[56]
Kawasaki AM, Casper MD, Freier SM, et al. Uniformly modified 2′-deoxy-2′-fluoro-phosphorothioate oligonucleotides as nuclease-resistant antisense compounds with high affinity and specificity for RNA targets. J Med Chem 1993; 36(7): 831-41.
[http://dx.doi.org/10.1021/jm00059a007] [PMID: 8464037]
[57]
Geary RS, Watanabe TA, Truong L, et al. Pharmacokinetic properties of 2′-O-(2-methoxyethyl)-modified oligonucleotide analogs in rats. J Pharmacol Exp Ther 2001; 296(3): 890-7.
[PMID: 11181921]
[58]
Sioud M, Furset G, Cekaite L. Suppression of immunostimulatory siRNA-driven innate immune activation by 2′-modified RNAs. Biochem Biophys Res Commun 2007; 361(1): 122-6.
[http://dx.doi.org/10.1016/j.bbrc.2007.06.177] [PMID: 17658482]
[59]
Boo SH, Kim YK. The emerging role of RNA modifications in the regulation of mRNA stability. Exp Mol Med 2020; 52(3): 400-8.
[http://dx.doi.org/10.1038/s12276-020-0407-z] [PMID: 32210357]
[60]
Mahmoudian RA, Fathi F, Farshchian M, Abbaszadegan MR. Construction and quantitative evaluation of a tissue-specific sleeping beauty by EDL2-specific transposase expression in esophageal squamous carcinoma cell line KYSE-30. Mol Biotechnol 2023; 65(3): 350-60.
[http://dx.doi.org/10.1007/s12033-022-00490-4] [PMID: 35474410]
[61]
Mahmoudian RA, Farshchian M, Abbaszadegan MR. Evaluation and optimization of lipofectamine 3000 reagents for transient gene expression in KYSE-30 esophagus cancer cell line. Arch Med Sci 2019; 5(4): 21.
[62]
Dlamini NG, Basson AK, Pullabhotla VSR. Optimization and application of bioflocculant passivated copper nanoparticles in the wastewater treatment. Int J Environ Res Public Health 2019; 16(12): 2185.
[http://dx.doi.org/10.3390/ijerph16122185] [PMID: 31226768]
[63]
Brunetti J, Piantini S, Fragai M, et al. A new NT4 peptide-based drug delivery system for cancer treatment. Molecules 2020; 25(5): 1088.
[http://dx.doi.org/10.3390/molecules25051088] [PMID: 32121130]
[64]
Dong X. Current strategies for brain drug delivery. Theranostics 2018; 8(6): 1481-93.
[http://dx.doi.org/10.7150/thno.21254] [PMID: 29556336]
[65]
Hong DS, Kurzrock R, Oh Y, et al. A phase 1 dose escalation, pharmacokinetic, and pharmacodynamic evaluation of eIF-4E antisense oligonucleotide LY2275796 in patients with advanced cancer. Clin Cancer Res 2011; 17(20): 6582-91.
[http://dx.doi.org/10.1158/1078-0432.CCR-11-0430] [PMID: 21831956]
[66]
Larrañeta E, Stewart S, Ervine M, Al-Kasasbeh R, Donnelly R. Hydrogels for hydrophobic drug delivery. Classification, synthesis and applications. J Funct Biomater 2018; 9(1): 13.
[http://dx.doi.org/10.3390/jfb9010013] [PMID: 29364833]
[67]
Whitehouse C, Solomon E. Current status of the molecular characterization of the ovarian cancer antigen CA125 and implications for its use in clinical screening. Gynecol Oncol 2003; 88(1): S152-7.
[http://dx.doi.org/10.1006/gyno.2002.6708] [PMID: 12586109]
[68]
Brigger I, Dubernet C, Couvreur P. Nanoparticles in cancer therapy and diagnosis. Adv Drug Deliv Rev 2012; 64: 24-36.
[http://dx.doi.org/10.1016/j.addr.2012.09.006] [PMID: 12204596]
[69]
Duncan R. The dawning era of polymer therapeutics. Nat Rev Drug Discov 2003; 2(5): 347-60.
[http://dx.doi.org/10.1038/nrd1088] [PMID: 12750738]
[70]
Devalapally H, Duan Z, Seiden MV, Amiji MM. Paclitaxel and ceramide co-administration in biodegradable polymeric nanoparticulate delivery system to overcome drug resistance in ovarian cancer. Int J Cancer 2007; 121(8): 1830-8.
[http://dx.doi.org/10.1002/ijc.22886] [PMID: 17557285]
[71]
Devalapally H, Duan Z, Seiden MV, Amiji MM. Modulation of drug resistance in ovarian adenocarcinoma by enhancing intracellular ceramide using tamoxifen-loaded biodegradable polymeric nanoparticles. Clin Cancer Res 2008; 14(10): 3193-203.
[http://dx.doi.org/10.1158/1078-0432.CCR-07-4973] [PMID: 18483388]
[72]
Jabr-Milane LS, van Vlerken LE, Yadav S, Amiji MM. Multi- functional nanocarriers to overcome tumor drug resistance. Cancer Treat Rev 2008; 34(7): 592-602.
[http://dx.doi.org/10.1016/j.ctrv.2008.04.003] [PMID: 18538481]
[73]
Sahay G, Alakhova DY, Kabanov AV. Endocytosis of nanomedicines. J Control Release 2010; 145(3): 182-95.
[http://dx.doi.org/10.1016/j.jconrel.2010.01.036] [PMID: 20226220]
[74]
Bansal T, Akhtar N, Jaggi M, Khar RK, Talegaonkar S. Novel formulation approaches for optimising delivery of anticancer drugs based on P-glycoprotein modulation. Drug Discov Today 2009; 14(21-22): 1067-74.
[http://dx.doi.org/10.1016/j.drudis.2009.07.010] [PMID: 19647803]
[75]
Elamanchili P, McEachern C, Burt H. Reversal of multidrug resistance by methoxypolyethylene glycol-block-polycaprolactone diblock copolymers through the inhibition of P-glycoprotein function. J Pharm Sci 2009; 98(3): 945-58.
[http://dx.doi.org/10.1002/jps.21479] [PMID: 18623213]
[76]
Zhang X, Chen J, Zheng Y, et al. Follicle-stimulating hormone peptide can facilitate paclitaxel nanoparticles to target ovarian carcinoma in vivo. Cancer Res 2009; 69(16): 6506-14.
[http://dx.doi.org/10.1158/0008-5472.CAN-08-4721] [PMID: 19638590]
[77]
Pantshwa JM, Kondiah PPD, Choonara YE, Marimuthu T, Pillay V. Nanodrug delivery systems for the treatment of ovarian cancer. Cancers 2020; 12(1): 213.
[http://dx.doi.org/10.3390/cancers12010213] [PMID: 31952210]
[78]
Shin H, Park SJ, Yim Y, et al. Recent advances in RNA therapeutics and RNA delivery systems based on nanoparticles. Adv Ther 2018; 1(7): 1800065.
[http://dx.doi.org/10.1002/adtp.201800065]
[79]
Akinc A, Goldberg M, Qin J, et al. Development of lipidoid-siRNA formulations for systemic delivery to the liver. Mol Ther 2009; 17(5): 872-9.
[http://dx.doi.org/10.1038/mt.2009.36] [PMID: 19259063]
[80]
García-Manrique P, Matos M, Gutiérrez G, Pazos C, Blanco-López MC. Therapeutic biomaterials based on extracellular vesicles: classification of bio-engineering and mimetic preparation routes. J Extracell Vesicles 2018; 7(1): 1422676.
[http://dx.doi.org/10.1080/20013078.2017.1422676] [PMID: 29372017]
[81]
Valadi H, Ekström K, Bossios A, Sjöstrand M, Lee JJ, Lötvall JO. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol 2007; 9(6): 654-9.
[http://dx.doi.org/10.1038/ncb1596] [PMID: 17486113]
[82]
Enriquez VA, Cleys ER, Da Silveira JC, Spillman MA, Winger QA, Bouma GJ. High LIN28A expressing ovarian cancer cells secrete exosomes that induce invasion and migration in HEK293 cells. Biomed Res Int 2015; 2015: 701390.
[http://dx.doi.org/10.1155/2015/701390]
[83]
Yin J, Yan X, Yao X, et al. Secretion of annexin A3 from ovarian cancer cells and its association with platinum resistance in ovarian cancer patients. J Cell Mol Med 2012; 16(2): 337-48.
[http://dx.doi.org/10.1111/j.1582-4934.2011.01316.x] [PMID: 21435174]
[84]
Khan S, Jutzy JMS, Aspe JR, McGregor DW, Neidigh JW, Wall NR. Survivin is released from cancer cells via exosomes. Apoptosis 2011; 16(1): 1-12.
[http://dx.doi.org/10.1007/s10495-010-0534-4] [PMID: 20717727]
[85]
Johnsen KB, Gudbergsson JM, Skov MN, Pilgaard L, Moos T, Duroux M. A comprehensive overview of exosomes as drug delivery vehicles - endogenous nanocarriers for targeted cancer therapy. Biochim Biophys Acta 2014; 1846(1): 75-87.
[PMID: 24747178]
[86]
Kooijmans SA, Vader P, van Dommelen SM, van Solinge WW, Schiffelers RM. Exosome mimetics: A novel class of drug delivery systems. Int J Nanomedicine 2012; 7: 1525-41.
[PMID: 22619510]
[87]
Liu H, Shen M, Zhao D, et al. The effect of triptolide-loaded exosomes on the proliferation and apoptosis of human ovarian cancer SKOV3 cells. Biomed Res Int 2019; 2019: 2595801.
[http://dx.doi.org/10.1155/2019/2595801]
[88]
Wang C, Guan W, Peng J, Chen Y, Xu G, Dou H. Gene/paclitaxel co-delivering nanocarriers prepared by framework-induced self-assembly for the inhibition of highly drug-resistant tumors. Acta Biomater 2020; 103: 247-58.
[http://dx.doi.org/10.1016/j.actbio.2019.12.015] [PMID: 31846802]
[89]
Markman M, Webster K, Zanotti K, Peterson G, Kulp B, Belinson J. Survival following the documentation of platinum and taxane resistance in ovarian cancer: A single institution experience involving multiple phase 2 clinical trials. Gynecol Oncol 2004; 93(3): 699-701.
[http://dx.doi.org/10.1016/j.ygyno.2004.03.023] [PMID: 15196867]
[90]
Mosleh-Shirazi S, Abbasi M, Shafiee M, Kasaee SR, Amani AM. Renal clearable nanoparticles: An expanding horizon for improving biomedical imaging and cancer therapy. Mater Today Commun 2021; 26: 102064.
[http://dx.doi.org/10.1016/j.mtcomm.2021.102064]
[91]
Tunç CÜ, Aydin O. Co-delivery of Bcl-2 siRNA and doxorubicin through gold nanoparticle-based delivery system for a combined cancer therapy approach. J Drug Deliv Sci Technol 2022; 74: 103603.
[http://dx.doi.org/10.1016/j.jddst.2022.103603]
[92]
Zhang J, Ding H, Zhang F, Xu Y, Liang W, Huang L. New trends in diagnosing and treating ovarian cancer using nanotechnology. Front Bioeng Biotechnol 2023; 11: 1160985.
[http://dx.doi.org/10.3389/fbioe.2023.1160985] [PMID: 37082219]
[93]
Ma CC, Wang ZL, Xu T, He ZY, Wei YQ. The approved gene therapy drugs worldwide: From 1998 to 2019. Biotechnol Adv 2020; 40: 107502.
[http://dx.doi.org/10.1016/j.biotechadv.2019.107502] [PMID: 31887345]
[94]
van den Brand D, Mertens V, Massuger LFAG, Brock R. siRNA in ovarian cancer - Delivery strategies and targets for therapy. J Control Release 2018; 283: 45-58.
[http://dx.doi.org/10.1016/j.jconrel.2018.05.012] [PMID: 29777795]
[95]
Gordon AN, Tonda M, Sun S, Rackoff W. Long-term survival advantage for women treated with pegylated liposomal doxorubicin compared with topotecan in a phase 3 randomized study of recurrent and refractory epithelial ovarian cancer. Gynecol Oncol 2004; 95(1): 1-8.
[http://dx.doi.org/10.1016/j.ygyno.2004.07.011] [PMID: 15385103]
[96]
Mutch DG, Orlando M, Goss T, et al. Randomized phase III trial of gemcitabine compared with pegylated liposomal doxorubicin in patients with platinum-resistant ovarian cancer. J Clin Oncol 2007; 25(19): 2811-8.
[http://dx.doi.org/10.1200/JCO.2006.09.6735] [PMID: 17602086]
[97]
Ferrandina G, Ludovisi M, Lorusso D, et al. Phase III trial of gemcitabine compared with pegylated liposomal doxorubicin in progressive or recurrent ovarian cancer. J Clin Oncol 2008; 26(6): 890-6.
[http://dx.doi.org/10.1200/JCO.2007.13.6606] [PMID: 18281662]
[98]
Lamb YN, Scott LJ. Liposomal irinotecan: A review in metastatic pancreatic adenocarcinoma. Drugs 2017; 77(7): 785-92.
[http://dx.doi.org/10.1007/s40265-017-0741-1] [PMID: 28401446]
[99]
Lim SA, Cox A, Tung M, Chung EJ. Clinical progress of nanomedicine-based RNA therapies. Bioact Mater 2022; 12: 203-13.
[http://dx.doi.org/10.1016/j.bioactmat.2021.10.018] [PMID: 35310381]
[100]
Schultheis B, Strumberg D, Santel A, et al. First-in-human phase I study of the liposomal RNA interference therapeutic Atu027 in patients with advanced solid tumors. J Clin Oncol 2014; 32(36): 4141-8.
[http://dx.doi.org/10.1200/JCO.2013.55.0376] [PMID: 25403217]
[101]
Zuckerman JE, Gritli I, Tolcher A, et al. Correlating animal and human phase Ia/Ib clinical data with CALAA-01, a targeted, polymer-based nanoparticle containing siRNA. Proc Natl Acad Sci 2014; 111(31): 11449-54.
[http://dx.doi.org/10.1073/pnas.1411393111] [PMID: 25049380]
[102]
Prakash TP, Allerson CR, Dande P, et al. Positional effect of chemical modifications on short interference RNA activity in mammalian cells. J Med Chem 2005; 48(13): 4247-53.
[http://dx.doi.org/10.1021/jm050044o] [PMID: 15974578]
[103]
Kaur T, Slavcev R, Wettig S. Addressing the challenge: Current and future directions in ovarian cancer therapy. Curr Gene Ther 2009; 9(6): 434-58.
[http://dx.doi.org/10.2174/156652309790031148] [PMID: 20021329]
[104]
Armstrong DK, Fleming GF, Markman M, Bailey HH. A phase I trial of intraperitoneal sustained-release paclitaxel microspheres (Paclimer®) in recurrent ovarian cancer: A gynecologic oncology group study. Gynecol Oncol 2006; 103(2): 391-6.
[http://dx.doi.org/10.1016/j.ygyno.2006.02.029] [PMID: 16626792]
[105]
Hatefi A, Amsden B. Biodegradable injectable in situ forming drug delivery systems. J Control Release 2002; 80(1-3): 9-28.
[http://dx.doi.org/10.1016/S0168-3659(02)00008-1] [PMID: 11943384]
[106]
Zahedi P, Yoganathan R, Piquette-Miller M, Allen C. Recent advances in drug delivery strategies for treatment of ovarian cancer. Expert Opin Drug Deliv 2012; 9(5): 567-83.
[http://dx.doi.org/10.1517/17425247.2012.665366] [PMID: 22452661]
[107]
Juliano R, Bauman J, Kang H, Ming X. Biological barriers to therapy with antisense and siRNA oligonucleotides. Mol Pharm 2009; 6(3): 686-95.
[http://dx.doi.org/10.1021/mp900093r] [PMID: 19397332]
[108]
Birmingham A, Anderson EM, Reynolds A, et al. 3′ UTR seed matches, but not overall identity, are associated with RNAi off-targets. Nat Methods 2006; 3(3): 199-204.
[http://dx.doi.org/10.1038/nmeth854] [PMID: 16489337]
[109]
Jackson AL, Bartz SR, Schelter J, et al. Expression profiling reveals off-target gene regulation by RNAi. Nat Biotechnol 2003; 21(6): 635-7.
[http://dx.doi.org/10.1038/nbt831] [PMID: 12754523]
[110]
Maduri S. Applicability of RNA interference in cancer therapy: Current status. Indian J Cancer 2015; 52(1): 11-21.
[http://dx.doi.org/10.4103/0019-509X.175598] [PMID: 26837960]
[111]
Scaggiante B, Dapas B, Farra R, et al. Improving siRNA bio-distribution and minimizing side effects. Curr Drug Metab 2011; 12(1): 11-23.
[http://dx.doi.org/10.2174/138920011794520017] [PMID: 21222588]
[112]
Halbur C, Choudhury N, Chen M, Kim JH, Chung EJ. siRNA-conjugated nanoparticles to treat ovarian cancer. SLAS Technol 2019; 24(2): 137-50.
[http://dx.doi.org/10.1177/2472630318816668] [PMID: 30616494]
[113]
Choung S, Kim YJ, Kim S, Park HO, Choi YC. Chemical modification of siRNAs to improve serum stability without loss of efficacy. Biochem Biophys Res Commun 2006; 342(3): 919-27.
[http://dx.doi.org/10.1016/j.bbrc.2006.02.049] [PMID: 16598842]
[114]
Bramsen JB, Kjems J. Engineering small interfering RNAs by strategic chemical modification. Methods Mol Biol 2013; 942: 87-109.
[115]
Soutschek J, Akinc A, Bramlage B, et al. Therapeutic silencing of an endogenous gene by systemic administration of modified siRNAs. Nature 2004; 432(7014): 173-8.
[http://dx.doi.org/10.1038/nature03121] [PMID: 15538359]
[116]
Nishina K, Unno T, Uno Y, et al. Efficient in vivo delivery of siRNA to the liver by conjugation of α-tocopherol. Mol Ther 2008; 16(4): 734-40.
[http://dx.doi.org/10.1038/mt.2008.14]
[117]
Wolfrum C, Shi S, Jayaprakash KN, et al. Mechanisms and optimization of in vivo delivery of lipophilic siRNAs. Nat Biotechnol 2007; 25(10): 1149-57.
[http://dx.doi.org/10.1038/nbt1339] [PMID: 17873866]
[118]
Zou S, Scarfo K, Nantz MH, Hecker JG. Lipid-mediated delivery of RNA is more efficient than delivery of DNA in non-dividing cells. Int J Pharm 2010; 389(1-2): 232-43.
[http://dx.doi.org/10.1016/j.ijpharm.2010.01.019] [PMID: 20080162]
[119]
Kosaka N, Iguchi H, Ochiya T. Circulating microRNA in body fluid: A new potential biomarker for cancer diagnosis and prognosis. Cancer Sci 2010; 101(10): 2087-92.
[http://dx.doi.org/10.1111/j.1349-7006.2010.01650.x] [PMID: 20624164]
[120]
Dickerson EB, Blackburn WH, Smith MH, Kapa LB, Lyon LA, McDonald JF. Chemosensitization of cancer cells by siRNA using targeted nanogel delivery. BMC Cancer 2010; 10(1): 10.
[http://dx.doi.org/10.1186/1471-2407-10-10] [PMID: 20064265]
[121]
Pan X, Thompson R, Meng X, Wu D, Xu L. Tumor-targeted RNA-interference: Functional non-viral nanovectors. Am J Cancer Res 2011; 1(1): 25-42.
[PMID: 21572539]
[122]
Jackson AL, Burchard J, Leake D, et al. Position-specific chemical modification of siRNAs reduces “off-target” transcript silencing. RNA 2006; 12(7): 1197-205.
[http://dx.doi.org/10.1261/rna.30706] [PMID: 16682562]
[123]
Liu Y, Franzen S. Factors determining the efficacy of nuclear delivery of antisense oligonucleotides by gold nanoparticles. Bioconjug Chem 2008; 19(5): 1009-16.
[http://dx.doi.org/10.1021/bc700421u] [PMID: 18393455]
[124]
Detzer A, Overhoff M, Wünsche W, et al. Increased RNAi is related to intracellular release of siRNA via a covalently attached signal peptide. RNA 2009; 15(4): 627-36.
[http://dx.doi.org/10.1261/rna.1305209] [PMID: 19228587]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy