Nucleic Acid Drugs—Current Status, Issues, and Expectations for Exosomes
Abstract
:Simple Summary
Abstract
1. Introduction
2. Advantages of Nucleic Acid Drugs
3. Classes of Nucleic Acid Drugs
3.1. Inhibition Type
3.1.1. Antisense Oligonucleotides
3.1.2. siRNAs
3.2. Splice Switching Type
3.3. Editing Type
3.4. Augmentation Type
3.5. Replacement Type
3.5.1. miRNA Mimics
3.5.2. mRNA Drugs
4. Current Status of Clinical Application of Nucleic Acid Drugs
4.1. Target Tissues of Nucleic Acid Drugs
4.2. Competition for the Same Target
5. Challenges in Nucleic Acid Drugs
5.1. Pharmacokinetics, Delivery Efficiency, and Safety of Nucleic Acid Drugs
5.2. Challenges in the Field of Cancer Therapeutics
6. Overcoming Challenges Associated with Nucleic Acid Drugs by Using Exosomes
6.1. Delivery of Nucleic Acids
6.2. Approaches for Multifactorial Diseases
6.3. Pharmacodynamic (PD) Marker of Nucleic Acid Therapeutics
7. Conclusions and Perspectives
Funding
Conflicts of Interest
References
- Farkona, S.; Diamandis, E.P.; Blasutig, I.M. Cancer immunotherapy: The beginning of the end of cancer? BMC Med. 2016, 14, 73. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Stephenson, M.L.; Zamecnik, P.C. Inhibition of Rous sarcoma viral RNA translation by a specific oligodeoxyribonucleotide. Proc. Natl. Acad. Sci. USA 1978, 75, 285–288. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hüttenhofer, A.; Schattner, P.; Polacek, N. Non-coding RNAs: Hope or hype? Trends Genet. 2005, 21, 289–297. [Google Scholar] [CrossRef] [PubMed]
- Ransohoff, J.D.; Wei, Y.; Khavari, P.A. The functions and unique features of long intergenic non-coding RNA. Nat. Rev. Mol. Cell Biol. 2018, 19, 143–157. [Google Scholar] [CrossRef]
- Fox, A.H.; Nakagawa, S.; Hirose, T.; Bond, C.S. Paraspeckles: Where long noncoding RNA meets phase separation. Trends Biochem. Sci. 2018, 43, 124–135. [Google Scholar] [CrossRef] [Green Version]
- Tam, V.; Patel, N.; Turcotte, M.; Bossé, Y.; Paré, G.; Meyre, D. Benefits and limitations of genome-wide association studies. Nat. Rev. Genet. 2019, 20, 467–484. [Google Scholar] [CrossRef]
- Heilbron, K.; Mozaffari, S.V.; Vacic, V.; Yue, P.; Wang, W.; Shi, J.; Jubb, A.M.; Pitts, S.J.; Wang, X. Advancing drug discovery using the power of the human genome. J. Pathol. 2021, 254, 418–429. [Google Scholar] [CrossRef]
- Kim, J.; Hu, C.; Moufawad El Achkar, C.; Black, L.E.; Douville, J.; Larson, A.; Pendergast, M.K.; Goldkind, S.F.; Lee, E.A.; Kuniholm, A.; et al. Patient-customized oligonucleotide therapy for a rare genetic disease. N. Engl. J. Med. 2019, 381, 1644–1652. [Google Scholar] [CrossRef]
- Moderna, Inc. Moderna Announces First Participant Dosed in NIH-led Phase 1 Study of mRNA Vaccine (mRNA-1273) against Novel Coronavirus. 16 March 2020 at 12:42 pm EDT. Available online: https://investors.modernatx.com/news-releases/news-release-details/moderna-announces-first-participant-dosed-nih-led-phase-1-study (accessed on 19 July 2021).
- Zhu, G.; Chen, X. Aptamer-based targeted therapy. Adv. Drug Deliv. Rev. 2018, 134, 65–78. [Google Scholar] [CrossRef]
- Klinman, D.M.; Klaschik, S.; Sato, T.; Tross, D. CpG oligonucleotides as adjuvants for vaccines targeting infectious diseases. Adv. Drug Deliv. Rev. 2009, 61, 248–255. [Google Scholar] [CrossRef]
- Krützfeldt, J.; Rajewsky, N.; Braich, R.; Rajeev, K.G.; Tuschl, T.; Manoharan, M.; Stoffel, M. Silencing of microRNAs in vivo with ‘antagomirs’. Nature 2005, 438, 685–689. [Google Scholar] [CrossRef] [PubMed]
- Si, M.L.; Zhu, S.; Wu, H.; Lu, Z.; Wu, F.; Mo, Y.Y. miR-21-mediated tumor growth. Oncogene 2007, 26, 2799–2803. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wu, H.; Lima, W.F.; Zhang, H.; Fan, A.; Sun, H.; Crooke, S.T. Determination of the role of the human RNase H1 in the pharmacology of DNA-like antisense drugs. J. Biol. Chem. 2004, 279, 17181–17189. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Eckstein, F. Phosphorothioates, essential components of therapeutic oligonucleotides. Nucleic Acid Ther. 2014, 24, 374–387. [Google Scholar] [CrossRef]
- Prakash, T.P. An overview of sugar-modified oligonucleotides for antisense therapeutics. Chem. Biodivers. 2011, 8, 1616–1641. [Google Scholar] [CrossRef]
- Obika, S.; Nanbu, D.; Hari, Y.; Morio, K.; In, Y.; Ishida, T.; Imanishi, T. Synthesis of 2′-O, 4′-C-methyleneuridine and -cytidine. Novel bicyclic nucleosides having a fixed c3-endo sugar puckering. Tetrahedron Lett. 1997, 38, 8735–8738. [Google Scholar] [CrossRef]
- Morita, K.; Hasegawa, C.; Kaneko, M.; Tsutsumi, S.; Sone, J.; Ishikawa, T.; Imanishi, T.; Koizumi, M. 2′-O, 4′-C-ethylene-bridged nucleic acids (ENA): Highly nuclease-resistant and thermodynamically stable oligonucleotides for antisense drug. Bioorg. Med. Chem. Lett. 2002, 12, 73–76. [Google Scholar] [CrossRef]
- Seth, P.P.; Vasquez, G.; Allerson, C.A.; Berdeja, A.; Gaus, H.; Kinberger, G.A.; Prakash, T.P.; Migawa, M.T.; Bhat, B.; Swayze, E.E. Synthesis and biophysical evaluation of 2′, 4′-constrained 2′O-methoxyethyl and 2′, 4′-constrained 2′O-ethyl nucleic acid analogues. J. Org. Chem. 2010, 75, 1569–1581. [Google Scholar] [CrossRef]
- Altmann, K.H.; Fabbro, D.; Dean, N.M.; Geiger, T.; Monia, B.P.; Müller, M.; Nicklin, P. Second-generation antisense oligonucleotides: Structure-activity relationships and the design of improved signal-transduction inhibitors. Biochem. Soc. Trans. 1996, 24, 630–637. [Google Scholar] [CrossRef]
- Yasuhara, H.; Baba, T.; Ono, F.; Miyata, K.; Miyake, K.; Seth, P.P.; Low, A.; Yoshida, M.; Bennett, C.F.; Kataoka, K.; et al. DNA/RNA heteroduplex oligonucleotide for highly efficient gene silencing. Nat. Commun. 2015, 6, 7969. [Google Scholar]
- Hara, R.I.; Yoshioka, K.; Yokota, T. DNA-RNA heteroduplex oligonucleotide for highly efficient gene silencing. Methods Mol. Biol. 2020, 2176, 113–119. [Google Scholar] [PubMed]
- Perry, C.M.; Balfour, J.A. Fomivirsen. Drugs 1999, 57, 375–380. [Google Scholar] [CrossRef]
- Hair, P.; Cameron, F.; McKeage, K. Mipomersen sodium: First global approval. Drugs 2013, 73, 487–493. [Google Scholar] [CrossRef] [PubMed]
- Keam, S.J. Inotersen: First global approval. Drugs 2018, 78, 1371–1376. [Google Scholar] [CrossRef] [PubMed]
- Elbashir, S.M.; Harborth, J.; Lendeckel, W.; Yalcin, A.; Weber, K.; Tuschl, T. Duplexes of 21-nucleotide RNAs mediate RNA interference in cultured mammalian cells. Nature 2001, 411, 494–498. [Google Scholar] [CrossRef] [PubMed]
- Rana, T.M. Illuminating the silence: Understanding the structure and function of small RNAs. Nat. Rev. Mol. Cell Biol. 2007, 8, 23–36. [Google Scholar] [CrossRef] [PubMed]
- Ui-Tei, K.; Naito, Y.; Takahashi, F.; Haraguchi, T.; Ohki-Hamazaki, H.; Juni, A.; Ueda, R.; Saigo, K. Guidelines for the selection of highly effective siRNA sequences for mammalian and chick RNA interference. Nucleic Acids Res. 2004, 32, 936–948. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Reynolds, A.; Leake, D.; Boese, Q.; Scaringe, S.; Marshall, W.S.; Khvorova, A. Rational siRNA design for RNA interference. Nat. Biotechnol. 2004, 22, 326–330. [Google Scholar] [CrossRef]
- Chiu, Y.L.; Rana, T.M. siRNA function in RNAi: A chemical modification analysis. RNA 2003, 9, 1034–1048. [Google Scholar] [CrossRef] [Green Version]
- Allerson, C.R.; Sioufi, N.; Jarres, R.; Prakash, T.P.; Naik, N.; Berdeja, A.; Wanders, L.; Griffey, R.H.; Swayze, E.E.; Bhat, B. Fully 2′-modified oligonucleotide duplexes with improved in vitro potency and stability compared to unmodified small interfering RNA. J. Med. Chem. 2005, 48, 901–904. [Google Scholar] [CrossRef]
- Hoy, S.M. Patisiran: First global approval. Drugs 2018, 78, 1625–1631. [Google Scholar] [CrossRef]
- Arechavala-Gomeza, V.; Khoo, B.; Aartsma-Rus, A. Splicing modulation therapy in the treatment of genetic diseases. Appl. Clin. Genet. 2014, 7, 245–252. [Google Scholar] [PubMed] [Green Version]
- Lim, K.R.; Maruyama, R.; Yokota, T. Eteplirsen in the treatment of Duchenne muscular dystrophy. Drug Des. Devel. Ther. 2017, 11, 533–545. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hoy, S.M. Nusinersen: First global approval. Drugs 2017, 77, 473–479. [Google Scholar] [CrossRef] [PubMed]
- Neil, E.E.; Bisaccia, E.K. Nusinersen: A novel antisense oligonucleotide for the treatment of spinal muscular atrophy. J. Pediatr. Pharmacol. Ther. 2019, 24, 194–203. [Google Scholar] [CrossRef] [PubMed]
- Summerton, J.; Weller, D. Morpholino antisense oligomers: Design, preparation, and properties. Antisense Nucleic Acid Drug Dev. 1997, 7, 187–195. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bass, B.L. RNA editing by adenosine deaminases that act on RNA. Annu. Rev. Biochem. 2002, 71, 817–846. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Peng, Z.; Cheng, Y.; Tan, B.C.; Kang, L.; Tian, Z.; Zhu, Y.; Zhang, W.; Liang, Y.; Hu, X.; Tan, X.; et al. Comprehensive analysis of RNA-Seq data reveals extensive RNA editing in a human transcriptome. Nat. Biotechnol. 2012, 30, 253–260. [Google Scholar] [CrossRef]
- Fukuda, M.; Umeno, H.; Nose, K.; Nishitarumizu, A.; Noguchi, R.; Nakagawa, H. Construction of a guide-RNA for site-directed RNA mutagenesis utilising intracellular A-to-I RNA editing. Sci. Rep. 2017, 7, 41478. [Google Scholar] [CrossRef]
- Wettengel, J.; Reautschnig, P.; Geisler, S.; Kahle, P.J.; Stafforst, T. Harnessing human ADAR2 for RNA repair—Recoding a PINK1 mutation rescues mitophagy. Nucleic Acids Res. 2017, 45, 2797–2808. [Google Scholar] [CrossRef] [Green Version]
- Katrekar, D.; Chen, G.; Meluzzi, D.; Ganesh, A.; Worlikar, A.; Shih, Y.R.; Varghese, S.; Mali, P. In Vivo RNA editing of point mutations via RNA-guided adenosine deaminases. Nat. Methods 2019, 16, 239–242. [Google Scholar] [CrossRef]
- Stafforst, T.; Schneider, M.F. An RNA-deaminase conjugate selectively repairs point mutations. Angew. Chem. Int. Ed. Engl. 2012, 51, 11166–11169. [Google Scholar] [CrossRef]
- Cox, D.B.T.; Gootenberg, J.S.; Abudayyeh, O.O.; Franklin, B.; Kellner, M.J.; Joung, J.; Zhang, F. RNA editing with CRISPR-Cas13. Science 2017, 358, 1019–1027. [Google Scholar] [CrossRef] [Green Version]
- Cideciyan, A.V.; Jacobson, S.G.; Ho, A.C.; Garafalo, A.V.; Roman, A.J.; Sumaroka, A.; Krishnan, A.K.; Swider, M.; Schwartz, M.R.; Girach, A. Durable vision improvement after a single treatment with antisense oligonucleotide sepofarsen: A case report. Nat. Med. 2021, 27, 785–789. [Google Scholar] [CrossRef]
- Li, L.C.; Okino, S.T.; Zhao, H.; Pookot, D.; Place, R.F.; Urakami, S.; Enokida, H.; Dahiya, R. Small dsRNAs induce transcriptional activation in human cells. Proc. Natl. Acad. Sci. USA 2006, 103, 17337–17342. [Google Scholar] [CrossRef] [Green Version]
- Voutila, J.; Reebye, V.; Roberts, T.C.; Protopapa, P.; Andrikakou, P.; Blakey, D.C.; Habib, R.; Huber, H.; Saetrom, P.; Rossi, J.J.; et al. Development and mechanism of small activating RNA targeting CEBPA, a novel therapeutic in clinical trials for liver cancer. Mol. Ther. 2017, 25, 2705–2714. [Google Scholar] [CrossRef] [Green Version]
- Reebye, V.; Huang, K.W.; Lin, V.; Jarvis, S.; Cutilas, P.; Dorman, S.; Ciriello, S.; Andrikakou, P.; Voutila, J.; Saetrom, P.; et al. Gene activation of CEBPA using saRNA: Preclinical studies of the first in human saRNA drug candidate for liver cancer. Oncogene 2018, 37, 3216–3228. [Google Scholar] [CrossRef]
- Sarker, D.; Plummer, R.; Meyer, T.; Sodergren, M.H.; Basu, B.; Chee, C.E.; Huang, K.W.; Palmer, D.H.; Ma, Y.T.; Evans, T.R.J.; et al. MTL-CEBPA, a small activating RNA therapeutic upregulating C/EBP-alpha, in patients with advanced liver cancer: A first-in-human, multicenter, open-label, phase I trial. Clin. Cancer Res. 2020, 26, 3936–3946. [Google Scholar] [CrossRef]
- Hosseinahli, N.; Aghapour, M.; Duijf, P.H.G.; Baradaran, B. Treating cancer with microRNA replacement therapy: A literature review. J. Cell Physiol. 2018, 233, 5574–5588. [Google Scholar] [CrossRef]
- He, L.; He, X.; Lim, L.P.; de Stanchina, E.; Xuan, Z.; Liang, Y.; Xue, W.; Zender, L.; Magnus, J.; Ridzon, D.; et al. A microRNA component of the p53 tumour suppressor network. Nature 2007, 447, 1130–1134. [Google Scholar] [CrossRef] [Green Version]
- Chang, T.C.; Wentzel, E.A.; Kent, O.A.; Ramachandran, K.; Mullendore, M.; Lee, K.H.; Feldmann, G.; Yamakuchi, M.; Ferlito, M.; Lowenstein, C.J.; et al. Transactivation of miR-34a by p53 broadly influences gene expression and promotes apoptosis. Mol. Cell 2007, 26, 745–752. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Beg, M.S.; Brenner, A.J.; Sachdev, J.; Borad, M.; Kang, Y.K.; Stoudemire, J.; Smith, S.; Bader, A.G.; Kim, S.; Hong, D.S. Phase I study of MRX34, a liposomal miR-34a mimic, administered twice weekly in patients with advanced solid tumors. Investig. New Drugs 2017, 35, 180–188. [Google Scholar] [CrossRef] [PubMed]
- Hong, D.S.; Kang, Y.K.; Borad, M.; Sachdev, J.; Ejadi, S.; Lim, H.Y.; Brenner, A.J.; Park, K.; Lee, J.L.; Kim, T.Y.; et al. Phase 1 study of MRX34, a liposomal miR-34a mimic, in patients with advanced solid tumours. Br. J. Cancer 2020, 122, 1630–1637. [Google Scholar] [CrossRef]
- Gallant-Behm, C.L.; Piper, J.; Lynch, J.M.; Seto, A.G.; Hong, S.J.; Mustoe, T.A.; Maari, C.; Pestano, L.A.; Dalby, C.M.; Jackson, A.L.; et al. A MicroRNA-29 mimic (Remlarsen) represses extracellular matrix expression and fibroplasia in the skin. J. Investig. Dermatol. 2019, 139, 1073–1081. [Google Scholar] [CrossRef] [PubMed]
- Van Zandwijk, N.; Pavlakis, N.; Kao, S.C.; Linton, A.; Boyer, M.J.; Clarke, S.; Huynh, Y.; Chrzanowska, A.; Fulham, M.J.; Bailey, D.L.; et al. Safety and activity of microRNA-loaded minicells in patients with recurrent malignant pleural mesothelioma: A first-in-man, phase 1, open-label, dose-escalation study. Lancet Oncol. 2017, 18, 1386–1396. [Google Scholar] [CrossRef]
- Sahin, U.; Karikó, K.; Türeci, Ö. mRNA-based therapeutics—Developing a new class of drugs. Nat. Rev. Drug Discov. 2014, 13, 759–780. [Google Scholar] [CrossRef]
- Wolff, J.A.; Malone, R.W.; Williams, P.; Chong, W.; Acsadi, G.; Jani, A.; Felgner, P.L. Direct gene transfer into mouse muscle in vivo. Science 1990, 247, 1465–1468. [Google Scholar] [CrossRef]
- Yamamoto, A.; Kormann, M.; Rosenecker, J.; Rudolph, C. Current prospects for mRNA gene delivery. Eur. J. Pharm. Biopharm. 2009, 71, 484–489. [Google Scholar] [CrossRef]
- Sahin, U.; Derhovanessian, E.; Miller, M.; Kloke, B.P.; Simon, P.; Löwer, M.; Bukur, V.; Tadmor, A.D.; Luxemburger, U.; Schrörs, B.; et al. Personalized RNA mutanome vaccines mobilize poly-specific therapeutic immunity against cancer. Nature 2017, 547, 222–226. [Google Scholar] [CrossRef]
- Food and Drug Administration. Pfizer COVID-19 Vaccine Emergency Use Authorization. 2020. Available online: https://www.fda.gov/emergency-preparedness-and-response/coronavirus-disease-2019-covid-19/pfizer-biontech-covid-19-vaccine (accessed on 19 July 2021).
- Food and Drug Administration. Moderna COVID-19 Vaccine Emergency Use Authorization. 2020. Available online: https://www.fda.gov/emergency-preparedness-and-response/coronavirus-disease-2019-covid-19/Moderna-covid-19-vaccine (accessed on 19 July 2021).
- Karikó, K.; Muramatsu, H.; Welsh, F.A.; Ludwig, J.; Kato, H.; Akira, S.; Weissman, D. Incorporation of pseudouridine into mRNA yields superior nonimmunogenic vector with increased translational capacity and biological stability. Mol. Ther. 2008, 16, 1833–1840. [Google Scholar] [CrossRef]
- Furuichi, Y.; LaFiandra, A.; Shatkin, A.J. 5′-terminal structure and mRNA stability. Nature 1977, 266, 235–239. [Google Scholar] [CrossRef]
- Stepinski, J.; Waddell, C.; Stolarski, R.; Darzynkiewicz, E.; Rhoads, R.E. Synthesis and properties of mRNAs containing the novel “anti-reverse” cap analogs 7-methyl (3′-O-methyl) GpppG and 7-methyl (3′-deoxy) GpppG. RNA 2001, 7, 1486–1495. [Google Scholar]
- Gustafsson, C.; Govindarajan, S.; Minshull, J. Codon bias and heterologous protein expression. Trends Biotechnol. 2004, 22, 346–353. [Google Scholar] [CrossRef]
- Mockey, M.; Gonçalves, C.; Dupuy, F.P.; Lemoine, F.M.; Pichon, C.; Midoux, P. mRNA transfection of dendritic cells: Synergistic effect of ARCA mRNA capping with Poly (A) chains in cis and in trans for a high protein expression level. Biochem. Biophys. Res. Commun. 2006, 340, 1062–1068. [Google Scholar] [CrossRef]
- Geary, R.S. Antisense oligonucleotide pharmacokinetics and metabolism. Expert Opin. Drug Metab. Toxicol. 2009, 5, 381–391. [Google Scholar] [CrossRef]
- Braasch, D.A.; Paroo, Z.; Constantinescu, A.; Ren, G.; Oz, O.K.; Mason, R.P.; Corey, D.R. Biodistribution of phosphodiester and phosphorothioate siRNA. Bioorg. Med. Chem. Lett. 2004, 14, 1139–1143. [Google Scholar] [CrossRef]
- Haché, M.; Swoboda, K.J.; Sethna, N.; Farrow-Gillespie, A.; Khandji, A.; Xia, S.; Bishop, K.M. Intrathecal injections in children with spinal muscular atrophy: Nusinersen clinical trial experience. J. Child. Neurol. 2016, 31, 899–906. [Google Scholar] [CrossRef] [Green Version]
- Molitoris, B.A.; Dagher, P.C.; Sandoval, R.M.; Campos, S.B.; Ashush, H.; Fridman, E.; Brafman, A.; Faerman, A.; Atkinson, S.J.; Thompson, J.D.; et al. siRNA targeted to p53 attenuates ischemic and cisplatin-induced acute kidney injury. J. Am. Soc. Nephrol. 2009, 20, 1754–1764. [Google Scholar] [CrossRef] [Green Version]
- Thompson, J.D.; Kornbrust, D.J.; Foy, J.W.; Solano, E.C.; Schneider, D.J.; Feinstein, E.; Molitoris, B.A.; Erlich, S. Toxicological and pharmacokinetic properties of chemically modified siRNAs targeting p53 RNA following intravenous administration. Nucleic Acid Ther. 2012, 22, 255–264. [Google Scholar] [CrossRef]
- Demirjian, S.; Ailawadi, G.; Polinsky, M.; Bitran, D.; Silberman, S.; Shernan, S.K.; Burnier, M.; Hamilton, M.; Squiers, E.; Erlich, S.; et al. Safety and tolerability study of an intravenously administered small interfering ribonucleic acid (siRNA) post on-pump cardiothoracic surgery in patients at risk of acute kidney injury. Kidney Int. Rep. 2017, 2, 836–843. [Google Scholar] [CrossRef] [Green Version]
- Oyama, S.; Yamamoto, T.; Yamayoshi, A. Recent advances in the delivery carriers and chemical conjugation strategies for nucleic acid drugs. Cancers 2021, 13, 3881. [Google Scholar] [CrossRef]
- Rietwyk, S.; Peer, D. Next-generation lipids in RNA interference therapeutics. ACS Nano 2017, 11, 7572–7586. [Google Scholar] [CrossRef] [PubMed]
- Shi, B.; Keough, E.; Matter, A.; Leander, K.; Young, S.; Carlini, E.; Sachs, A.B.; Tao, W.; Abrams, M.; Howell, B.; et al. Biodistribution of small interfering RNA at the organ and cellular levels after lipid nanoparticle-mediated delivery. J. Histochem. Cytochem. 2011, 59, 727–740. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Akinc, A.; Querbes, W.; De, S.; Qin, J.; Frank-Kamenetsky, M.; Jayaprakash, K.N.; Jayaraman, M.; Rajeev, K.G.; Cantley, W.L.; Dorkin, J.R.; et al. Targeted delivery of RNAi therapeutics with endogenous and exogenous ligand-based mechanisms. Mol. Ther. 2010, 18, 1357–1364. [Google Scholar] [CrossRef] [PubMed]
- Zhang, X.; Goel, V.; Robbie, G.J. Pharmacokinetics of Patisiran, the first approved RNA interference therapy in patients with hereditary transthyretin-mediated amyloidosis. J. Clin. Pharmacol. 2019, 60, 573–585. [Google Scholar] [CrossRef] [Green Version]
- Endsley, A.N.; Ho, R.J. Design and characterization of novel peptide-coated lipid nanoparticles for targeting anti-HIV drug to CD4 expressing cells. AAPS J. 2012, 14, 225–235. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Suk, J.S.; Xu, Q.; Kim, N.; Hanes, J.; Ensign, L.M. PEGylation as a strategy for improving nanoparticle-based drug and gene delivery. Adv. Drug Deliv. Rev. 2016, 99, 28–51. [Google Scholar] [CrossRef] [Green Version]
- Sato, Y.; Murase, K.; Kato, J.; Kobune, M.; Sato, T.; Kawano, Y.; Takimoto, R.; Takada, K.; Miyanishi, K.; Matsunaga, T.; et al. Resolution of liver cirrhosis using vitamin A-coupled liposomes to deliver siRNA against a collagen-specific chaperone. Nat. Biotechnol. 2008, 26, 431–442. [Google Scholar] [CrossRef]
- Bahl, K.; Senn, J.J.; Yuzhakov, O.; Bulychev, A.; Brito, L.A.; Hassett, K.J.; Laska, M.E.; Smith, M.; Almarsson, Ö.; Thompson, J.; et al. Preclinical and clinical demonstration of immunogenicity by mRNA vaccines against H10N8 and H7N9 influenza viruses. Mol. Ther. 2017, 25, 1316–1327. [Google Scholar] [CrossRef] [Green Version]
- Richner, J.M.; Himansu, S.; Dowd, K.A.; Butler, S.L.; Salazar, V.; Fox, J.M.; Julander, J.G.; Tang, W.W.; Shresta, S.; Pierson, T.C.; et al. Modified mRNA vaccines protect against Zika virus infection. Cell 2017, 168, 1114–1125. [Google Scholar] [CrossRef] [Green Version]
- Weide, B.; Carralot, J.P.; Reese, A.; Scheel, B.; Eigentler, T.K.; Hoerr, I.; Rammensee, H.G.; Garbe, C.; Pascolo, S. Results of the first phase I/II clinical vaccination trial with direct injection of mRNA. J. Immunother. 2008, 31, 180–188. [Google Scholar] [CrossRef]
- Diken, M.; Kreiter, S.; Selmi, A.; Britten, C.M.; Huber, C.; Türeci, Ö.; Sahin, U. Selective uptake of naked vaccine RNA by dendritic cells is driven by macropinocytosis and abrogated upon DC maturation. Gene. Ther. 2011, 18, 702–708. [Google Scholar] [CrossRef] [Green Version]
- Selmi, A.; Vascotto, F.; Kautz-Neu, K.; Türeci, Ö.; Sahin, U.; von Stebut, E.; Diken, M.; Kreiter, S. Uptake of synthetic naked RNA by skin-resident dendritic cells via macropinocytosis allows antigen expression and induction of T-cell responses in mice. Cancer Immunol. Immunother. 2016, 65, 1075–1083. [Google Scholar] [CrossRef]
- Spiess, M. The asialoglycoprotein receptor: A model for endocytic transport receptors. Biochemistry 1990, 29, 10009–10018. [Google Scholar] [CrossRef]
- Nair, J.K.; Willoughby, J.L.; Chan, A.; Charisse, K.; Alam, M.R.; Wang, Q.; Hoekstra, M.; Kandasamy, P.; Kel’in, A.V.; Milstein, S.; et al. Multivalent N-acetylgalactosamine-conjugated siRNA localizes in hepatocytes and elicits robust RNAi-mediated gene silencing. J. Am. Chem. Soc. 2014, 136, 16958–16961. [Google Scholar] [CrossRef] [Green Version]
- Prakash, T.P.; Graham, M.J.; Yu, J.; Carty, R.; Low, A.; Chappell, A.; Schmidt, K.; Zhao, C.; Aghajan, M.; Murray, H.F.; et al. Targeted delivery of antisense oligonucleotides to hepatocytes using triantennary N-acetyl galactosamine improves potency 10-fold in mice. Nucleic Acids Res. 2014, 42, 8796–8807. [Google Scholar] [CrossRef] [Green Version]
- Scott, L.J. Givosiran: First approval. Drugs 2020, 80, 335–339. [Google Scholar] [CrossRef]
- Scott, L.J.; Keam, S.J. Lumasiran: First approval. Drugs 2021, 81, 277–282. [Google Scholar] [CrossRef]
- Johnson, S.M.; Connelly, S.; Fearns, C.; Powers, E.T.; Kelly, J.W. The transthyretin amyloidoses: From delineating the molecular mechanism of aggregation linked to pathology to a regulatory-agency-approved drug. J. Mol. Biol. 2012, 421, 185–203. [Google Scholar] [CrossRef] [Green Version]
- Adams, D.; Gonzalez-Duarte, A.; O′Riordan, W.D.; Yang, C.C.; Ueda, M.; Kristen, A.V.; Tournev, I.; Schmidt, H.H.; Coelho, T.; Berk, J.L.; et al. Patisiran, an RNAi therapeutic, for hereditary transthyretin amyloidosis. N. Engl. J. Med. 2018, 379, 11–21. [Google Scholar] [CrossRef]
- Benson, M.D.; Waddington-Cruz, M.; Berk, J.L.; Polydefkis, M.; Dyck, P.J.; Wang, A.K.; Planté-Bordeneuve, V.; Barroso, F.A.; Merlini, G.; Obici, L.; et al. Inotersen treatment for patients with hereditary transthyretin amyloidosis. N. Engl. J. Med. 2018, 379, 22–31. [Google Scholar] [CrossRef] [PubMed]
- Heo, Y.A. Golodirsen: First approval. Drugs 2020, 80, 329–333. [Google Scholar] [CrossRef] [PubMed]
- Dhillon, S. Viltolarsen: First approval. Drugs 2020, 80, 1027–1031. [Google Scholar] [CrossRef] [PubMed]
- Clemens, P.R.; Rao, V.K.; Connolly, A.M.; Harper, A.D.; Mah, J.K.; Smith, E.C.; McDonald, C.M.; Zaidman, C.M.; Morgenroth, L.P.; Osaki, H.; et al. Safety, tolerability, and efficacy of viltolarsen in boys with Duchenne muscular dystrophy amenable to exon 53 skipping: A phase 2 randomized clinical trial. JAMA Neurol. 2020, 77, 982–991. [Google Scholar] [CrossRef] [PubMed]
- Muntoni, F.; Frank, D.; Sardone, V.; Morgan, J.; Schnell, F.; Charleston, J.; Desjardins, C.; Phadke, R.; Sewry, C.; Popplewell, L.; et al. Golodirsen induces exon skipping leading to sarcolemmal dystrophin expression in Duchenne muscular dystrophy patients with mutations amenable to exon 53 skipping (S22.001). Neurology 2018, 90. [Google Scholar]
- Lamb, Y.N. Inclisiran: First approval. Drugs 2021, 81, 389–395. [Google Scholar] [CrossRef]
- Sabatine, M.S. PCSK9 inhibitors: Clinical evidence and implementation. Nat. Rev. Cardiol. 2019, 16, 155–165. [Google Scholar] [CrossRef]
- Ray, K.K.; Landmesser, U.; Leiter, L.A.; Kallend, D.; Dufour, R.; Karakas, M.; Hall, T.; Troquay, R.P.; Turner, T.; Visseren, F.L.; et al. Inclisiran in patients at high cardiovascular risk with elevated LDL cholesterol. N. Engl. J. Med. 2017, 376, 1430–1440. [Google Scholar] [CrossRef] [Green Version]
- European Medicines Agency. Leqvio 284 mg Solution for Injection in Pre-Filled Syringe: Summary of Product Characteristics. 2021. Available online: http://www.ema.europa.eu/ (accessed on 19 July 2021).
- Kelly, R.J.; Hill, A.; Arnold, L.M.; Brooksbank, G.L.; Richards, S.J.; Cullen, M.; Mitchell, L.D.; Cohen, D.R.; Gregory, W.M.; Hillmen, P. Long-term treatment with eculizumab in paroxysmal nocturnal hemoglobinuria: Sustained efficacy and improved survival. Blood 2011, 117, 6786–6792. [Google Scholar] [CrossRef] [Green Version]
- Zuber, J.; Fakhouri, F.; Roumenina, L.T.; Loirat, C.; Frémeaux-Bacchi, V.; French Study Group for aHUS/C3G. Use of eculizumab for atypical haemolytic uraemic syndrome and C3 glomerulopathies. Nat. Rev. Nephrol. 2012, 8, 643–657. [Google Scholar] [CrossRef]
- Hill, A.; Valls, A.G.; Griffin, M.; Munir, T.; Borodovsky, A.; Kawahata, N.; Mclean, H.; Shi, K.; Partisano, A.M.; Kim, J.; et al. A subcutaneously administered investigational RNAi therapeutic (ALN-CC5) targeting complement C5 for treatment of PNH and complement-mediated diseases: Preliminary phase 1/2 study results in patients with PNH. Blood 2016, 128, 3891. [Google Scholar] [CrossRef]
- Badri, P.; Jiang, X.; Borodovsky, A.; Najafian, N.; Kim, J.; Clausen, V.A.; Goel, V.; Habtemariam, B.; Robbie, G.J. Pharmacokinetic and pharmacodynamic properties of cemdisiran, an RNAi therapeutic targeting complement component 5, in healthy subjects and patients with paroxysmal nocturnal hemoglobinuria. Clin. Pharmacokinet. 2021, 60, 365–378. [Google Scholar] [CrossRef] [PubMed]
- Hoy, S.M. Onasemnogene abeparvovec: First global approval. Drugs 2019, 79, 1255–1262. [Google Scholar] [CrossRef] [PubMed]
- Day, J.W.; Finkel, R.S.; Chiriboga, C.A.; Connolly, A.M.; Crawford, T.O.; Darras, B.T.; Iannaccone, S.T.; Kuntz, N.L.; Peña, L.D.M.; Shieh, P.B.; et al. Onasemnogene abeparvovec gene therapy for symptomatic infantile-onset spinal muscular atrophy in patients with two copies of SMN2 (STR1VE): An open-label, single-arm, multicentre, phase 3 trial. Lancet Neurol. 2021, 20, 284–293. [Google Scholar] [CrossRef]
- Foust, K.D.; Wang, X.; McGovern, V.L.; Braun, L.; Bevan, A.K.; Haidet, A.M.; Le, T.T.; Morales, P.R.; Rich, M.M.; Burghes, A.H.; et al. Rescue of the spinal muscular atrophy phenotype in a mouse model by early postnatal delivery of SMN. Nat. Biotechnol. 2010, 28, 271–274. [Google Scholar] [CrossRef] [Green Version]
- Kaspar, B.; Thomsen, G.; Hsieh, C.; Do, J.; Solano, S.; Chu, B.; Barkho, B.; Fugere, M.; Kaufmann, P.; Foust, K.; et al. Biodistribution of onasemnogene abeparvovec (AVXS-101) DNA, mRNA, and SMN protein in human tissue. Neuromuscul. Disord. 2019, 29, S122–S123. [Google Scholar] [CrossRef]
- Saraiva, J.; Nobre, R.J.; de Pereira, A.L. Gene therapy for the CNS using AAVs: The impact of systemic delivery by AAV9. J. Control Release 2016, 241, 94–109. [Google Scholar] [CrossRef]
- Harada, Y.; Rao, V.K.; Arya, K.; Kuntz, N.L.; DiDonato, C.J.; Napchan-Pomerantz, G.; Agarwal, A.; Stefans, V.; Katsuno, M.; Veerapandiyan, A. Combination molecular therapies for type 1 spinal muscular atrophy. Muscle Nerve 2020, 62, 550–554. [Google Scholar] [CrossRef]
- Geary, R.S.; Norris, D.; Yu, R.; Bennett, C.F. Pharmacokinetics, biodistribution and cell uptake of antisense oligonucleotides. Adv. Drug Deliv. Rev. 2015, 87, 46–51. [Google Scholar] [CrossRef] [Green Version]
- Yu, R.Z.; Grundy, J.S.; Geary, R.S. Clinical pharmacokinetics of second generation antisense oligonucleotides. Expert Opin. Drug Metab. Toxicol. 2013, 9, 169–182. [Google Scholar] [CrossRef]
- Crooke, S.T.; Baker, B.F.; Xia, S.; Yu, R.Z.; Viney, N.J.; Wang, Y.; Tsimikas, S.; Geary, R.S. Integrated assessment of the clinical performance of GalNAc (3)-conjugated 2′-O-methoxyethyl chimeric antisense oligonucleotides: I. Human volunteer experience. Nucleic Acid Ther. 2019, 29, 16–32. [Google Scholar] [CrossRef] [Green Version]
- Dowdy, S.F. Overcoming cellular barriers for RNA therapeutics. Nat. Biotechnol. 2017, 35, 222–229. [Google Scholar] [CrossRef]
- Varkouhi, A.K.; Scholte, M.; Storm, G.; Haisma, H.J. Endosomal escape pathways for delivery of biologicals. J. Control Release 2011, 151, 220–228. [Google Scholar] [CrossRef]
- Lönn, P.; Kacsinta, A.D.; Cui, X.S.; Hamil, A.S.; Kaulich, M.; Gogoi, K.; Dowdy, S.F. Enhancing endosomal escape for intracellular delivery of macromolecular biologic therapeutics. Sci. Rep. 2016, 6, 32301. [Google Scholar] [CrossRef]
- Smith, S.A.; Selby, L.I.; Johnston, A.P.R.; Such, G.K. The endosomal escape of nanoparticles: Toward more efficient cellular delivery. Bioconjug Chem. 2019, 30, 263–272. [Google Scholar] [CrossRef]
- Judge, A.D.; Sood, V.; Shaw, J.R.; Fang, D.; McClintock, K.; MacLachlan, I. Sequence-dependent stimulation of the mammalian innate immune response by synthetic siRNA. Nat. Biotechnol. 2005, 23, 457–462. [Google Scholar] [CrossRef]
- Advani, R.; Peethambaram, P.; Lum, B.L.; Fisher, G.A.; Hartmann, L.; Long, H.J.; Halsey, J.; Holmlund, J.T.; Dorr, A.; Sikic, B.I. A phase II trial of aprinocarsen, an antisense oligonucleotide inhibitor of protein kinase C alpha, administered as a 21-day infusion to patients with advanced ovarian carcinoma. Cancer 2004, 100, 321–326. [Google Scholar] [CrossRef]
- Chi, K.N.; Eisenhauer, E.; Fazli, L.; Jones, E.C.; Goldenberg, S.L.; Powers, J.; Tu, D.; Gleave, M.E. A phase I pharmacokinetic and pharmacodynamic study of OGX-011, a 2′-methoxyethyl antisense oligonucleotide to clusterin, in patients with localized prostate cancer. J. Natl. Cancer Inst. 2005, 97, 1287–1296. [Google Scholar] [CrossRef]
- Chi, K.N.; Zoubeidi, A.; Gleave, M.E. Custirsen (OGX-011): A second-generation antisense inhibitor of clusterin for the treatment of cancer. Expert Opin. Investig. Drugs 2008, 17, 1955–1962. [Google Scholar] [CrossRef]
- O′Brien, S.; Moore, J.O.; Boyd, T.E.; Larratt, L.M.; Skotnicki, A.; Koziner, B.; Chanan-Khan, A.A.; Seymour, J.F.; Bociek, R.G.; Pavletic, S.; et al. Randomized phase III trial of fludarabine plus cyclophosphamide with or without oblimersen sodium (Bcl-2 antisense) in patients with relapsed or refractory chronic lymphocytic leukemia. J. Clin. Oncol. 2007, 25, 1114–1120. [Google Scholar] [CrossRef]
- Beer, T.M.; Hotte, S.J.; Saad, F.; Alekseev, B.; Matveev, V.; Fléchon, A.; Gravis, G.; Joly, F.; Chi, K.N.; Malik, Z.; et al. Custirsen (OGX-011) combined with cabazitaxel and prednisone versus cabazitaxel and prednisone alone in patients with metastatic castration-resistant prostate cancer previously treated with docetaxel (AFFINITY): A randomised, open-label, international, phase 3 trial. Lancet Oncol. 2017, 18, 1532–1542. [Google Scholar] [PubMed]
- Bogdahn, U.; Hau, P.; Stockhammer, G.; Venkataramana, N.K.; Mahapatra, A.K.; Suri, A.; Balasubramaniam, A.; Nair, S.; Oliushine, V.; Parfenov, V.; et al. Targeted therapy for high-grade glioma with the TGF-β2 inhibitor trabedersen: Results of a randomized and controlled phase IIb study. Neuro. Oncol. 2011, 13, 132–142. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zuckerman, J.E.; Gritli, I.; Tolcher, A.; Heidel, J.D.; Lim, D.; Morgan, R.; Chmielowski, B.; Ribas, A.; Davis, M.E.; Yen, Y. Correlating animal and human phase Ia/Ib clinical data with CALAA-01, a targeted, polymer-based nanoparticle containing siRNA. Proc. Natl. Acad. Sci. USA 2014, 111, 11449–11454. [Google Scholar] [CrossRef] [Green Version]
- Demeure, M.J.; Armaghany, T.; Ejadi, S.; Ramanathan, R.K.; ElFiky, A.; Strosberg, J.R.; Smith, D.C.; Whitsett, T.; Liang, W.S.; Sekar, S.; et al. A phase I/II study of TKM-080301, a PLK1-targeted RNAi in patients with adrenocortical cancer (ACC). J. Clin. Oncol. 2016, 34, 2547. [Google Scholar] [CrossRef]
- Aleku, M.; Schulz, P.; Keil, O.; Santel, A.; Schaeper, U.; Dieckhoff, B.; Janke, O.; Endruschat, J.; Durieux, B.; Röder, N.; et al. Atu027, a liposomal small interfering RNA formulation targeting protein kinase N3, inhibits cancer progression. Cancer Res. 2008, 68, 9788–9798. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tabernero, J.; Shapiro, G.I.; LoRusso, P.M.; Cervantes, A.; Schwartz, G.K.; Weiss, G.J.; Paz-Ares, L.; Cho, D.C.; Infante, J.R.; Alsina, M.; et al. First-in-humans trial of an RNA interference therapeutic targeting VEGF and KSP in cancer patients with liver involvement. Cancer Discov. 2013, 3, 406–417. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Khvalevsky, E.Z.; Gabai, R.; Rachmut, I.H.; Horwitz, E.; Brunschwig, Z.; Orbach, A.; Shemi, A.; Golan, T.; Domb, A.J.; Yavin, E.; et al. Mutant KRAS is a druggable target for pancreatic cancer. Proc. Natl. Acad. Sci. USA. 2013, 110, 20723–20728. [Google Scholar] [CrossRef] [Green Version]
- Hay, M.; Thomas, D.W.; Craighead, J.L.; Economides, C.; Rosenthal, J. Clinical development success rates for investigational drugs. Nat. Biotechnol. 2014, 32, 40–51. [Google Scholar] [CrossRef]
- Davis, M.E.; Zuckerman, J.E.; Choi, C.H.J.; Seligson, D.; Tolcher, A.; Alabi, C.A.; Yen, Y.; Heidel, J.D.; Ribas, A. Evidence of RNAi in humans from systemically administered siRNA via targeted nanoparticles. Nat. Cell Biol. 2010, 464, 1067–1070. [Google Scholar] [CrossRef]
- Zhang, P.; An, K.; Duan, X.; Xu, H.; Li, F.; Xu, F. Recent advances in siRNA delivery for cancer therapy using smart nanocarriers. Drug Discov. Today. 2018, 23, 900–911. [Google Scholar] [CrossRef]
- Valadi, H.; Ekström, K.; Bossios, A.; Sjöstrand, M.; Lee, J.J.; Lötvall, J.O. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat. Cell Biol. 2007, 9, 654–659. [Google Scholar] [CrossRef] [Green Version]
- Jeppesen, D.K.; Fenix, A.M.; Franklin, J.L.; Higginbotham, J.N.; Zhang, Q.; Zimmerman, L.J.; Liebler, D.C.; Ping, J.; Liu, Q.; Evans, R.; et al. Reassessment of exosome composition. Cell 2019, 177, 428–445. [Google Scholar] [CrossRef] [Green Version]
- Hoshino, A.; Costa-Silva, B.; Shen, T.L.; Rodrigues, G.; Hashimoto, A.; Tesic Mark, M.; Molina, H.; Kohsaka, S.; Di Giannatale, A.; Ceder, S.; et al. Tumour exosome integrins determine organotropic metastasis. Nature 2015, 527, 329–335. [Google Scholar] [CrossRef] [Green Version]
- Cully, M. Exosome-based candidates move into the clinic. Nat. Rev. Drug Discov. 2021, 20, 6–7. [Google Scholar] [CrossRef]
- Didiot, M.C.; Hall, L.M.; Coles, A.H.; Haraszti, R.A.; Godinho, B.M.; Chase, K.; Sapp, E.; Ly, S.; Alterman, J.F.; Hassler, M.R.; et al. Exosome-mediated delivery of hydrophobically modified siRNA for huntingtin mRNA silencing. Mol. Ther. 2016, 24, 1836–1847. [Google Scholar] [CrossRef] [Green Version]
- Lamichhane, T.N.; Jeyaram, A.; Patel, D.B.; Parajuli, B.; Livingston, N.K.; Arumugasaamy, N.; Schardt, J.S.; Jay, S.M. Oncogene knockdown via active loading of small RNAs into extracellular vesicles by sonication. Cell Mol. Bioeng. 2016, 9, 315–324. [Google Scholar] [CrossRef]
- Alvarez-Erviti, L.; Seow, Y.; Yin, H.; Betts, C.; Lakhal, S.; Wood, M.J. Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nat. Biotechnol. 2011, 29, 341–345. [Google Scholar] [CrossRef]
- Sato, Y.T.; Umezaki, K.; Sawada, S.; Mukai, S.A.; Sasaki, Y.; Harada, N.; Shiku, H.; Akiyoshi, K. Engineering hybrid exosomes by membrane fusion with liposomes. Sci. Rep. 2016, 6, 21933. [Google Scholar] [CrossRef] [Green Version]
- Kusuma, G.D.; Barabadi, M.; Tan, J.L.; Morton, D.A.V.; Frith, J.E.; Lim, R. To protect and to preserve: Novel preservation strategies for extracellular vesicles. Front. Pharmacol. 2018, 9, 1199. [Google Scholar] [CrossRef] [Green Version]
- Zimmermann, G.R.; Lehár, J.; Keith, C.T. Multi-target therapeutics: When the whole is greater than the sum of the parts. Drug Discov. Today 2007, 12, 34–42. [Google Scholar] [CrossRef]
- He, B.; Lu, C.; Zheng, G.; He, X.; Wang, M.; Chen, G.; Zhang, G.; Lu, A. Combination therapeutics in complex diseases. J. Cell Mol. Med. 2016, 20, 2231–2240. [Google Scholar] [CrossRef] [PubMed]
- Sehgal, A.; Chen, Q.; Gibbings, D.; Sah, D.W.; Bumcrot, D. Tissue-specific gene silencing monitored in circulating RNA. RNA 2014, 20, 143–149. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chan, A.; Liebow, A.; Yasuda, M.; Gan, L.; Racie, T.; Maier, M.; Kuchimanchi, S.; Foster, D.; Milstein, S.; Charisse, K.; et al. Preclinical development of a subcutaneous ALAS1 RNAi therapeutic for treatment of hepatic porphyrias using circulating RNA quantification. Mol. Ther. Nucleic Acids 2015, 4, e263. [Google Scholar] [CrossRef] [PubMed]
- Sardh, E.; Harper, P.; Balwani, M.; Stein, P.; Rees, D.; Bissell, D.M.; Desnick, R.; Parker, C.; Phillips, J.; Bonkovsky, H.L.; et al. Phase 1 trial of an RNA interference therapy for acute intermittent porphyria. N. Engl. J. Med. 2019, 380, 549–558. [Google Scholar] [CrossRef]
Regulation Type | Name | Structure | Length (nt) | Representative Companies |
---|---|---|---|---|
Inhibition | ASO | ssDNA | 13–30 | Ionis, Nippon Shinyaku |
siRNA | dsRNA | 20–30 | Alnylam, Dicerna, Quark | |
Splice switching | SSO | ssDNA | 20–30 | Sarepta, Ionis |
Editing | EON | ssRNA | 20–40 | ProQR |
Augmentation | saRNA | dsRNA | 20–30 | MiNA |
Replacement | miRNA mimic | dsRNA | 20–30 | Mirna, miReven |
mRNA | ssRNA | hundreds to thousands | BioNTech, Moderna |
Drug Name | Year of Approval | Type | Target | Indication | DDS | Company |
---|---|---|---|---|---|---|
Fomivirsen | 1998 | ASO | CMV IE2 | Cytomegalovirus retinitis | Naked | Ionis |
Mipomersen | 2013 | ASO | ApoB-100 | Homozygous familial hypercholesterolemia | Naked | Ionis |
Eteplirsen | 2016 | SSO | Exon 51 of DMD | Duchenne muscular dystrophy | Naked | Sarepta |
Nusinersen | 2016 | SSO | Exon 7 of SMN2 | Spinal muscular atrophy | Naked | Ionis |
Inotersen | 2018 | ASO | TTR | Hereditary transthyretin mediated amyloidosis | Naked | Ionis |
Patisiran | 2018 | siRNA | TTR | Hereditary transthyretin mediated amyloidosis | LNP | Alnylam |
Golodirsen | 2019 | SSO | Exon 53 of DMD | Duchenne muscular dystrophy | Naked | Sarepta |
Volanesorsen | 2019 | ASO | ApoC3 | Familial chylomicronemia syndrome | Naked | Ionis |
Givosiran | 2019 | siRNA | ALAS1 | Acute hepatic porphyria | GalNAc | Alnylam |
Viltolarsen | 2020 | SSO | Exon 53 of DMD | Duchenne muscular dystrophy | Naked | Nippon Shinyaku |
Lumasiran | 2020 | siRNA | hydroxyacid oxidase 1 | Primary hyperoxaluria type 1 | GalNAc | Alnylam |
Inclisiran | 2020 | siRNA | PCSK9 | Familial hypercholesterolemia | GalNAc | Alnylam, Novartis |
Tozinameran | 2020 | mRNA | SARS-CoV-2 | COVID-19 Vaccine | LNP | BioNTech, Pfizer |
Elasomeran | 2020 | mRNA | SARS-CoV-2 | COVID-19 Vaccine | LNP | Moderna |
Casimersen | 2021 | SSO | Exon 45 of DMD | Duchenne muscular dystrophy | Naked | Sarepta |
Drug Name | Type | Target | Indication | DDS | Company |
---|---|---|---|---|---|
Pelacarsen | ASO | Apolipoprotein A | Cardiovascular disease | GalNAc | Ionis, Novartis |
Eplontersen | ASO | TTR | TTR amyloidosis | GalNAc | Ionis |
APOCIII-LRx | ASO | ApoC3 | Familial chylomicronemia syndrome | GalNAc | Ionis |
Tofersen | ASO | SOD1 | Amyotrophic lateral sclerosis | Naked | Ionis, Biogen |
Tominersen | ASO | HTT | Huntington’s disease | Naked | Ionis, Roche |
ION-363 | ASO | FUS | Amyotrophic lateral sclerosis | Naked | Ionis |
Nexagon | ASO | Connexin 43 | Corneal injury | Naked | Ocunexus |
Fitusiran | siRNA | Antithrombin III | Hemophilia | GalNAc | Alnylam, Sanofi |
Vutrisiran | siRNA | TTR | TTR amyloidosis | GalNAc | Alnylam |
Nedosiran | siRNA | LDHA | Primary hyperoxaluria | GalNAc | Dicerna |
Teprasiran | siRNA | P53 | Acute kidney injury | Naked | Quark |
Cosdosiran | siRNA | Caspase 2 | Non-arteritic anterior ischemic optic neuropathy | Naked | Quark |
Tivanisiran | siRNA | TRPV1 | Dry eye | Naked | Sylentis |
Sepofarsen | EON | CEP290 | Leber congenital amaurosis | Naked | ProQR |
Zorecimeran | mRNA | SARS-CoV-2 | COVID-19 vaccine | LNP | CureVac |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the author. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Yamada, Y. Nucleic Acid Drugs—Current Status, Issues, and Expectations for Exosomes. Cancers 2021, 13, 5002. https://doi.org/10.3390/cancers13195002
Yamada Y. Nucleic Acid Drugs—Current Status, Issues, and Expectations for Exosomes. Cancers. 2021; 13(19):5002. https://doi.org/10.3390/cancers13195002
Chicago/Turabian StyleYamada, Yoji. 2021. "Nucleic Acid Drugs—Current Status, Issues, and Expectations for Exosomes" Cancers 13, no. 19: 5002. https://doi.org/10.3390/cancers13195002
APA StyleYamada, Y. (2021). Nucleic Acid Drugs—Current Status, Issues, and Expectations for Exosomes. Cancers, 13(19), 5002. https://doi.org/10.3390/cancers13195002