Small Drugs, Huge Impact: The Extraordinary Impact of Antisense Oligonucleotides in Research and Drug Development
Abstract
:1. Introduction
2. Evolution of Chemistry and Properties of ASOs
3. First-Generation ASOs
4. Second-Generation ASOs
5. Third-Generation ASOs
6. ASO Design
7. ASO-Based Therapeutics
8. FDA Approved ASO-Based Pharmaceuticals
8.1. Formirsen (Vitraven®)
8.2. Mipomersen (Kinamro®)
8.3. Patisiran (Onpattro®)
8.4. Inotersen (Tegsedi®)
8.5. Givosiran (Givlaari®)
9. New Drugs Currently under Investigation
9.1. Vutrisiran
9.2. Tominersen
9.3. Volanesorsen (Waylivra®)
9.4. Miravirsen
10. ASO Application in a Pandemic World
11. Discussion
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- 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]
- Liang, X.-H.; Sun, H.; Shen, W.; Wang, S.; Yao, J.; Migawa, M.T.; Bui, H.-H.; Damle, S.S.; Riney, S.; Graham, M.J.; et al. Antisense oligonucleotides targeting translation inhibitory elements in 5′ UTRs can selectively increase protein levels. Nucleic Acids Res. 2017, 45, 9528–9546. [Google Scholar] [CrossRef] [Green Version]
- Crooke, S.T. Molecular Mechanisms of Antisense Oligonucleotides. Nucleic Acid Ther. 2017, 27, 70–77. [Google Scholar] [CrossRef]
- Hagedorn, P.H.; Persson, R.; Funder, E.D.; Albæk, N.; Diemer, S.L.; Hansen, D.J.; Møller, M.R.; Papargyri, N.; Christiansen, H.; Hansen, B.R.; et al. Locked nucleic acid: Modality, diversity, and drug discovery. Drug Discov. Today 2018, 23, 101–114. [Google Scholar] [CrossRef] [Green Version]
- Roberts, T.C.; Langer, R.; Wood, M.J.A. Advances in oligonucleotide drug delivery. Nat. Rev. Drug Discov. 2020, 19, 673–694. [Google Scholar] [CrossRef]
- Ward, A.J.; Norrbom, M.; Chun, S.; Bennett, C.F.; Rigo, F. Nonsense-mediated decay as a terminating mechanism for antisense oligonucleotides. Nucleic Acids Res. 2014, 42, 5871–5879. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Aoki, Y.; Wood, M.J.A. Emerging Oligonucleotide Therapeutics for Rare Neuromuscular Diseases. J. Neuromuscul. Dis. 2021, 8, 869–884. [Google Scholar] [CrossRef] [PubMed]
- Aartsma-Rus, A.; Straub, V.; Hemmings, R.; Haas, M.; Schlosser-Weber, G.; Stoyanova-Beninska, V.; Mercuri, E.; Muntoni, F.; Sepodes, B.; Vroom, E.; et al. Development of Exon Skipping Therapies for Duchenne Muscular Dystrophy: A Critical Review and a Perspective on the Outstanding Issues. Nucleic Acid Ther. 2017, 27, 251–259. [Google Scholar] [CrossRef] [PubMed]
- Aartsma-Rus, A.; Corey, D.R. The 10th Oligonucleotide Therapy Approved: Golodirsen for Duchenne Muscular Dystrophy. Nucleic Acid Ther. 2020, 30, 67–70. [Google Scholar] [CrossRef] [Green Version]
- Charleston, J.S.; Schnell, F.J.; Dworzak, J.; Donoghue, C.; Lewis, S.; Chen, L.; Young, G.D.; Milici, A.J.; Voss, J.; Dealwis, U.; et al. Eteplirsen treatment for Duchenne muscular dystrophy: Exon skipping and dystrophin production. Neurology 2018, 90, e2146–e2154. [Google Scholar] [CrossRef] [PubMed]
- Meng, L.; Ward, A.J.; Chun, S.; Bennett, C.F.; Beaudet, A.L.; Rigo, F. Towards a therapy for Angelman syndrome by targeting a long non-coding RNA. Nature 2015, 518, 409–412. [Google Scholar] [CrossRef] [PubMed]
- Naveed, A.; Cooper, J.A.; Li, R.; Hubbard, A.; Chen, J.; Liu, T.; Wilton, S.D.; Fletcher, S.; Fox, A.H. NEAT1 polyA-modulating antisense oligonucleotides reveal opposing functions for both long non-coding RNA isoforms in neuroblastoma. Cell. Mol. Life Sci. 2021, 78, 2213–2230. [Google Scholar] [CrossRef] [PubMed]
- Mansoor, M.; Melendez, A.J. Advances in Antisense Oligonucleotide Development for Target Identification, Validation, and as Novel Therapeutics. Gene Regul. Syst. Biol. 2008, 2, 275–295. [Google Scholar] [CrossRef] [PubMed]
- Paterson, B.M.; Roberts, B.E.; Kuff, E.L. Structural gene identification and mapping by DNA-mRNA hybrid-arrested cell-free translation. Proc. Natl. Acad. Sci. USA 1977, 74, 4370–4374. [Google Scholar] [CrossRef] [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] [Green Version]
- Quemener, A.M.; Bachelot, L.; Forestier, A.; Donnou-Fournet, E.; Gilot, D.; Galibert, M. The powerful world of antisense oligonucleotides: From bench to bedside. Wiley Interdiscip. Rev. RNA 2020, 11, e1594. [Google Scholar] [CrossRef]
- Dias, N.; Stein, C.A. Antisense oligonucleotides: Basic concepts and mechanisms. Mol. Cancer Ther. 2002, 1, 347–355. [Google Scholar]
- Furdon, P.J.; Dominski, Z.; Kole, R. RNase H cleavage of RNA hybridized to oligonucleotides containing methylphosphonate, phosphorothioate and phosphodiester bonds. Nucleic Acids Res. 1989, 17, 9193–9204. [Google Scholar] [CrossRef] [Green Version]
- Koziołkiewicz, M.; Gendaszewska, E.; Maszewska, M.; Stein, C.A.; Stec, W.J.; Drbal, K.; Angelisová, P.; Hilgert, I.; Černý, J.; Novák, P.; et al. The mononucleotide-dependent, nonantisense mechanism of action of phosphodiester and phosphorothioate oligonucleotides depends upon the activity of an ecto-5′-nucleotidase. Blood 2001, 98, 995–1002. [Google Scholar] [CrossRef]
- Miller, P.S.; McParland, K.B.; Jayaraman, K.; Tso, P.O.P. Biochemical and biological effects of nonionic nucleic acid methylphosphonates. Biochemistry 1981, 20, 1874–1880. [Google Scholar] [CrossRef]
- Miller, P.S.; Yano, J.; Yano, E.; Carroll, C.; Jayaraman, K.; Ts’O, P.O.P. Nonionic nucleic acid analogs. Synthesis and characterization of dideoxyribonucleoside methylphosphonates. Biochemistry 1979, 18, 5134–5143. [Google Scholar] [CrossRef]
- Shoji, Y.; Akhtar, S.; Periasamy, A.; Herman, B.; Juliano, R.L. Mechansism of cellular uptake of modified oligodeoxynucleotides containing methylphosphonate linkages. Nucleic Acids Res. 1991, 19, 5543–5550. [Google Scholar] [CrossRef] [Green Version]
- Tonkinson, J.L.; Stein, C.A. Patterns of intracellular compartmentalization, trafficking and acidification of 5×-fluorescein labeled phosphodiester and phosphorothioate oligodeoxynucleotides in HL60 cells. Nucleic Acids Res. 1994, 22, 4268–4275. [Google Scholar] [CrossRef] [Green Version]
- Peyrottes, S.; Vasseur, J.-J.; Imbach, J.-L.; Rayner, B. Oligodeoxynucleoside phosphoramidates (P-NH2): Synthesis and thermal stability of duplexes with DNA and RNA targets. Nucleic Acids Res. 1996, 24, 1841–1848. [Google Scholar] [CrossRef] [Green Version]
- Heidenreich, O.; Gryaznov, S.; Nerenberg, M. RNase H-independent antisense activity of oligonucleotide N3’->P5’ phosphoramidates. Nucleic Acids Res. 1997, 25, 776–780. [Google Scholar] [CrossRef] [Green Version]
- Faria, M.; Spiller, D.G.; Dubertret, C.; Nelson, J.S.; White, M.R.; Scherman, D.; Hélène, C.; Giovannangeli, C. Phosphoramidate oligonucleotides as potent antisense molecules in cells and in vivo. Nat. Biotechnol. 2001, 19, 40–44. [Google Scholar] [CrossRef]
- Agrawal, S.; Goodchild, J.; Civeira, M.P.; Thornton, A.H.; Sarin, P.S.; Zamecnik, P.C. Oligodeoxynucleoside phosphoramidates and phosphorothioates as inhibitors of human immunodeficiency virus. Proc. Natl. Acad. Sci. USA 1988, 85, 7079–7083. [Google Scholar] [CrossRef] [Green Version]
- Asai, A.; Oshima, Y.; Yamamoto, Y.; Uochi, T.-A.; Kusaka, H.; Akinaga, S.; Yamashita, Y.; Pongracz, K.; Pruzan, R.; Wunder, E.; et al. A novel telomerase template antagonist (GRN163) as a potential anticancer agent. Cancer Res. 2003, 63, 3931–3939. [Google Scholar] [PubMed]
- Sproat, B.S.; Lamond, A.I.; Beijer, B.; Neuner, P.; Ryder, U. Highly efficient chemical synthesis of 2′-O-methyloligoribonucleotides and tetrabiotinylated derivatives; novel probes that are resistant to degradation by RNA or DNA specific nucleases. Nucleic Acids Res. 1989, 17, 3373–3386. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Khvorova, A.; Watts, J.K. The chemical evolution of oligonucleotide therapies of clinical utility. Nat. Biotechnol. 2017, 35, 238–248. [Google Scholar] [CrossRef] [PubMed]
- Krefft, S.D.; Rose, C.S.; Nawaz, S.; Miller, Y.E. Deployment-Related Lung Disorders. Fed. Pract. 2015, 32, 32–38. [Google Scholar] [PubMed]
- Burel, S.A.; Hart, C.E.; Cauntay, P.; Hsiao, J.; Machemer, T.; Katz, M.; Watt, A.T.; Bui, H.-H.; Younis, H.; Sabripour, M.; et al. Hepatotoxicity of high affinity gapmer antisense oligonucleotides is mediated by RNase H1 dependent promiscuous reduction of very long pre-mRNA transcripts. Nucleic Acids Res. 2016, 44, 2093–2109. [Google Scholar] [CrossRef] [Green Version]
- Swayze, E.E.; Siwkowski, A.M.; Wancewicz, E.V.; Migawa, M.T.; Wyrzykiewicz, T.K.; Hung, G.; Monia, B.P.; Bennett, A.C.F.; Bennett, C.F. Antisense oligonucleotides containing locked nucleic acid improve potency but cause significant hepatotoxicity in animals. Nucleic Acids Res. 2007, 35, 687–700. [Google Scholar] [CrossRef]
- Straarup, E.M.; Fisker, N.; Hedtjärn, M.; Lindholm, M.W.; Rosenbohm, C.; Aarup, V.; Hansen, H.F.; Ørum, H.; Hansen, J.B.R.; Koch, T. Short locked nucleic acid antisense oligonucleotides potently reduce apolipoprotein B mRNA and serum cholesterol in mice and non-human primates. Nucleic Acids Res. 2010, 38, 7100–7111. [Google Scholar] [CrossRef] [Green Version]
- Geary, R.S.; Henry, S.P.; Grillone, L.R. Fomivirsen: Clinical pharmacology and potential drug interactions. Clin. Pharmacokinet. 2002, 41, 255–260. [Google Scholar] [CrossRef] [PubMed]
- Roehr, B. Fomivirsen approved for CMV retinitis. J. Int. Assoc. Physicians AIDS Care 1998, 4, 14–16. [Google Scholar]
- Vitravene Study Group. A randomized controlled clinical trial of intravitreous fomivirsen for treatment of newly diagnosed peripheral cytomegalovirus retinitis in patients with aids. Am. J. Ophthalmol. 2002, 133, 467–474. [Google Scholar] [CrossRef]
- Moulton, J.D. Using Morpholinos to Control Gene Expression. Curr. Protoc. Nucleic Acid Chem. 2017, 68, 4.30.1–4.30.29. [Google Scholar] [CrossRef]
- Larsen, H.J.; Bentin, T.; Nielsen, P.E. Antisense properties of peptide nucleic acid. Biochim. Biophys. Acta 1999, 1489, 159–166. [Google Scholar] [CrossRef]
- Goyenvalle, A.; Griffith, G.; Babbs, A.; El Andaloussi, S.; Ezzat, K.; Avril, A.; Dugovic, B.; Chaussenot, R.; Ferry, A.; Voit, T.; et al. Functional correction in mouse models of muscular dystrophy using exon-skipping tricyclo-DNA oligomers. Nat. Med. 2015, 21, 270–275. [Google Scholar] [CrossRef]
- Imbert, M.; Blandel, F.; Leumann, C.; Garcia, L.; Goyenvalle, A. Lowering Mutant Huntingtin Using Tricyclo-DNA Antisense Oligonucleotides as a Therapeutic Approach for Huntington’s Disease. Nucleic Acid Ther. 2019, 29, 256–265. [Google Scholar] [CrossRef] [PubMed]
- Shen, W.; De Hoyos, C.L.; Migawa, M.T.; Vickers, T.A.; Sun, H.; Low, A.; Bell, T.A.; Rahdar, M.; Mukhopadhyay, S.; Hart, C.E.; et al. Chemical modification of PS-ASO therapeutics reduces cellular protein-binding and improves the therapeutic index. Nat. Biotechnol. 2019, 37, 640–650. [Google Scholar] [CrossRef]
- Amantana, A.; Iversen, P.L. Pharmacokinetics and biodistribution of phosphorodiamidate morpholino antisense oligomers. Curr. Opin. Pharmacol. 2005, 5, 550–555. [Google Scholar] [CrossRef]
- Ostergaard, M.E.; Southwell, A.L.; Kordasiewicz, H.; Watt, A.T.; Skotte, N.H.; Doty, C.N.; Vaid, K.; Villanueva, E.; Swayze, E.; Bennett, C.F.; et al. Rational design of antisense oligonucleotides targeting single nucleotide polymorphisms for potent and allele selective suppression of mutant Huntingtin in the CNS. Nucleic Acids Res. 2013, 41, 9634–9650. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Southwell, A.L.; Skotte, N.H.; Kordasiewicz, H.B.; Østergaard, M.E.; Watt, A.T.; Carroll, J.B.; Doty, C.N.; Villanueva, E.; Petoukhov, E.; Vaid, K.; et al. In Vivo Evaluation of Candidate Allele-specific Mutant Huntingtin Gene Silencing Antisense Oligonucleotides. Mol. Ther. 2014, 22, 2093–2106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pedersen, L.; Hagedorn, P.H.; Lindholm, M.W.; Lindow, M. A Kinetic Model Explains Why Shorter and Less Affine Enzyme-recruiting Oligonucleotides Can Be More Potent. Mol. Ther. Nucleic Acids 2014, 3, e149. [Google Scholar] [CrossRef] [PubMed]
- Vickers, T.A.; Wyatt, J.R.; Freier, S.M. Effects of RNA secondary structure on cellular antisense activity. Nucleic Acids Res. 2000, 28, 1340–1347. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Andronescu, M.; Zhang, Z.C.; Condon, A. Secondary Structure Prediction of Interacting RNA Molecules. J. Mol. Biol. 2005, 345, 987–1001. [Google Scholar] [CrossRef]
- Ding, Y.; Lawrence, C.E. Statistical prediction of single-stranded regions in RNA secondary structure and application to predicting effective antisense target sites and beyond. Nucleic Acids Res. 2001, 29, 1034–1046. [Google Scholar] [CrossRef] [Green Version]
- Zhang, Y.C.; Taylor, M.M.; Samson, W.K.; Phillips, M.I. Antisense inhibition: Oligonucleotides, ribozymes, and siRNAs. Methods Mol. Med. 2005, 106, 11–34. [Google Scholar] [CrossRef]
- Zuker, M. Mfold web server for nucleic acid folding and hybridization prediction. Nucleic Acids Res. 2003, 31, 3406–3415. [Google Scholar] [CrossRef]
- Ding, Y.; Chan, C.Y.; Lawrence, C.E. Sfold web server for statistical folding and rational design of nucleic acids. Nucleic Acids Res. 2004, 32, W135–W141. [Google Scholar] [CrossRef]
- Yang, S.-P.; Song, S.-T.; Tang, Z.-M.; Song, H.-F. Optimization of antisense drug design against conservative local motif in simulant secondary structures of HER-2 mRNA and QSAR analysis. Acta Pharmacol. Sin. 2003, 24, 897–902. [Google Scholar]
- Ho, S.P.; Bao, Y.; Lesher, T.; Malhotra, R.; Ma, L.Y.; Fluharty, S.J.; Sakai, R.R. Mapping of RNA accessible sites for antisense experiments with oligonucleotide libraries. Nat. Biotechnol. 1998, 16, 59–63. [Google Scholar] [CrossRef]
- Bo, X.; Wang, S. TargetFinder: A software for antisense oligonucleotide target site selection based on MAST and secondary structures of target mRNA. Bioinformatics 2005, 21, 1401–1402. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Matveeva, O.V.; Tsodikov, A.D.; Giddings, M.; Freier, S.M.; Wyatt, J.R.; Spiridonov, A.N.; Shabalina, S.A.; Gesteland, R.F.; Atkins, J.F. Identification of sequence motifs in oligonucleotides whose presence is correlated with antisense activity. Nucleic Acids Res. 2000, 28, 2862–2865. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ho, S.P.; Britton, D.H.O.; Stone, B.A.; Behrens, D.L.; Leffet, L.M.; Hobbs, F.W.; Miller, J.A.; Trainor, G.L. Potent antisense oligonucleotides to the human multidrug resistance-1 mRNA are rationally selected by mapping RNA-accessible sites with oligonucleotide libraries. Nucleic Acids Res. 1996, 24, 1901–1907. [Google Scholar] [CrossRef] [Green Version]
- Matveeva, O.; Mathews, D.H.; Tsodikov, A.D.; Shabalina, S.A.; Gesteland, R.F.; Atkins, J.F.; Freier, S.M. Thermodynamic criteria for high hit rate antisense oligonucleotide design. Nucleic Acids Res. 2003, 31, 4989–4994. [Google Scholar] [CrossRef] [Green Version]
- Yamamoto, T.; Mukai, Y.; Wada, F.; Terada, C.; Kayaba, Y.; Oh, K.; Yamayoshi, A.; Obika, S.; Harada–Shiba, M. Highly Potent GalNAc-Conjugated Tiny LNA Anti-miRNA-122 Antisense Oligonucleotides. Pharmaceutics 2021, 13, 817. [Google Scholar] [CrossRef]
- Catuogno, S.; Di Martino, M.T.; Nuzzo, S.; Esposito, C.L.; Tassone, P.; de Franciscis, V. An Anti-BCMA RNA Aptamer for miRNA Intracellular Delivery. Mol. Ther. Nucleic Acids 2019, 18, 981–990. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Affinito, A.; Quintavalle, C.; Esposito, C.L.; Roscigno, G.; Giordano, C.; Nuzzo, S.; Ricci-Vitiani, L.; Scognamiglio, I.; Minic, Z.; Pallini, R.; et al. Targeting Ephrin Receptor Tyrosine Kinase A2 with a Selective Aptamer for Glioblastoma Stem Cells. Mol. Ther. Nucleic Acids 2020, 20, 176–185. [Google Scholar] [CrossRef]
- Esposito, C.L.; Autiero, I.; Basal, M.A.; Sandomenico, A.; Ummarino, S.; Borchiellini, M.; Ruvo, M.; Catuogno, S.; Ebralidze, A.K.; de Franciscis, V.; et al. Targeted systematic evolution of an RNA platform neutralizing DNMT1 function and controlling DNA methylation. bioRxiv 2020, bioRxiv:2020.2007.2029.226803. [Google Scholar] [CrossRef]
- Liang, X.-H.; Sun, H.; Nichols, J.G.; Crooke, S.T. RNase H1-Dependent Antisense Oligonucleotides Are Robustly Active in Directing RNA Cleavage in Both the Cytoplasm and the Nucleus. Mol. Ther. 2017, 25, 2075–2092. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Taiana, E.; Favasuli, V.; Ronchetti, D.; Todoerti, K.; Pelizzoni, F.; Manzoni, M.; Barbieri, M.; Fabris, S.; Silvestris, I.; Cantafio, M.E.G.; et al. Long non-coding RNA NEAT1 targeting impairs the DNA repair machinery and triggers anti-tumor activity in multiple myeloma. Leukemia 2020, 34, 234–244. [Google Scholar] [CrossRef] [PubMed]
- Maruyama, R.; Yokota, T. Knocking Down Long Noncoding RNAs Using Antisense Oligonucleotide Gapmers. Methods Mol. Biol. 2020, 2176, 49–56. [Google Scholar] [CrossRef] [PubMed]
- Amodio, N.; Stamato, M.A.; Juli, G.; Morelli, E.; Fulciniti, M.; Manzoni, M.; Taiana, E.; Agnelli, L.; Cantafio, M.E.G.; Romeo, E.; et al. Drugging the lncRNA MALAT1 via LNA gapmeR ASO inhibits gene expression of proteasome subunits and triggers anti-multiple myeloma activity. Leukemia 2018, 32, 1948–1957. [Google Scholar] [CrossRef] [Green Version]
- Obad, S.; Dos Santos, C.O.; Petri, A.; Heidenblad, M.; Broom, O.; Ruse, C.; Fu, C.; Lindow, M.; Stenvang, J.; Straarup, E.M.; et al. Silencing of microRNA families by seed-targeting tiny LNAs. Nat. Genet. 2011, 43, 371–378. [Google Scholar] [CrossRef]
- Jearawiriyapaisarn, N.; Moulton, H.M.; Sazani, P.; Kole, R.; Willis, M.S. Long-term improvement in mdx cardiomyopathy after therapy with peptide-conjugated morpholino oligomers. Cardiovasc. Res. 2010, 85, 444–453. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Iwamoto, N.; Butler, D.C.D.; Svrzikapa, N.; Mohapatra, S.; Zlatev, I.; Sah, D.W.Y.; Meena; Standley, S.M.; Lu, G.; Apponi, L.H.; et al. Control of phosphorothioate stereochemistry substantially increases the efficacy of antisense oligonucleotides. Nat. Biotechnol. 2017, 35, 845–851. [Google Scholar] [CrossRef]
- Dhuri, K.; Bechtold, C.; Quijano, E.; Pham, H.; Gupta, A.; Vikram, A.; Bahal, R. Antisense Oligonucleotides: An Emerging Area in Drug Discovery and Development. J. Clin. Med. 2020, 9, 2004. [Google Scholar] [CrossRef]
- Wang, Y.; Yu, R.Z.; Henry, S.; Geary, R.S. Pharmacokinetics and Clinical Pharmacology Considerations of GalNAc3-Conjugated Antisense Oligonucleotides. Expert Opin. Drug Metab. Toxicol. 2019, 15, 475–485. [Google Scholar] [CrossRef]
- Prakash, T.P.; Mullick, A.E.; Lee, R.G.; Yu, J.; Yeh, S.T.; Low, A.; Chappell, A.E.; Østergaard, M.E.; Murray, S.; Gaus, H.J.; et al. Fatty acid conjugation enhances potency of antisense oligonucleotides in muscle. Nucleic Acids Res. 2019, 47, 6029–6044. [Google Scholar] [CrossRef]
- Betts, C.; Saleh, A.F.; Arzumanov, A.A.; Hammond, S.; Godfrey, C.; Coursindel, T.; Gait, M.J.; Wood, M.J. Pip6-PMO, A New Generation of Peptide-oligonucleotide Conjugates with Improved Cardiac Exon Skipping Activity for DMD Treatment. Mol. Ther. Nucleic Acids 2012, 1, e38. [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]
- Perry, C.M.; Balfour, J.A.B. Fomivirsen. Drugs 1999, 57, 375–380. [Google Scholar] [CrossRef]
- Phillips, J.; Craig, S.; Bayley, D.; Christian, R.; Geary, R.; Nicklin, P. Pharmacokinetics, metabolism, and elimination of a 20-mer phosphorothioate oligodeoxynucleotide (cgp 69846a) after intravenous and subcutaneous administration. Biochem. Pharmacol. 1997, 54, 657–668. [Google Scholar] [CrossRef]
- Rinaldi, C.; Wood, M.J.A. Antisense oligonucleotides: The next frontier for treatment of neurological disorders. Nat. Rev. Neurol. 2018, 14, 9–21. [Google Scholar] [CrossRef] [PubMed]
- Sermet-Gaudelus, I.; Clancy, J.P.; Nichols, D.P.; Nick, J.A.; De Boeck, K.; Solomon, G.M.; Mall, M.A.; Bolognese, J.; Bouisset, F.; Hollander, W.D.; et al. Antisense oligonucleotide eluforsen improves CFTR function in F508del cystic fibrosis. J. Cyst. Fibros. 2019, 18, 536–542. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Drevinek, P.; Pressler, T.; Cipolli, M.; De Boeck, K.; Schwarz, C.; Bouisset, F.; Boff, M.; Henig, N.; Paquette-Lamontagne, N.; Montgomery, S.; et al. Antisense oligonucleotide eluforsen is safe and improves respiratory symptoms in F508DEL cystic fibrosis. J. Cyst. Fibros. 2020, 19, 99–107. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Debacker, A.J.; Voutila, J.; Catley, M.; Blakey, D.; Habib, N. Delivery of Oligonucleotides to the Liver with GalNAc: From Research to Registered Therapeutic Drug. Mol. Ther. 2020, 28, 1759–1771. [Google Scholar] [CrossRef]
- 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] [PubMed] [Green Version]
- Cui, H.; Zhu, X.; Li, S.; Wang, P.; Fang, J. Liver-Targeted Delivery of Oligonucleotides with N-Acetylgalactosamine Conjugation. ACS Omega 2021, 6, 16259–16265. [Google Scholar] [CrossRef] [PubMed]
- Tanowitz, M.; Hettrick, L.; Revenko, A.; Kinberger, G.A.; Prakash, T.P.; Seth, P.P. Asialoglycoprotein receptor 1 mediates productive uptake of N-acetylgalactosamine-conjugated and unconjugated phosphorothioate antisense oligonucleotides into liver hepatocytes. Nucleic Acids Res. 2017, 45, 12388–12400. [Google Scholar] [CrossRef] [PubMed]
- Huang, Y. Preclinical and Clinical Advances of GalNAc-Decorated Nucleic Acid Therapeutics. Mol. Ther. Nucleic Acids 2017, 6, 116–132. [Google Scholar] [CrossRef] [Green Version]
- Viney, N.J.; van Capelleveen, J.C.; Geary, R.S.; Xia, S.; Tami, J.A.; Yu, R.Z.; Marcovina, S.M.; Hughes, S.G.; Graham, M.J.; Crooke, R.M.; et al. Antisense oligonucleotides targeting apolipoprotein(a) in people with raised lipoprotein(a): Two randomised, double-blind, placebo-controlled, dose-ranging trials. Lancet 2016, 388, 2239–2253. [Google Scholar] [CrossRef]
- Arnold, A.E.; Malek-Adamian, E.; Le, P.U.; Meng, A.; Martínez-Montero, S.; Petrecca, K.; Damha, M.J.; Shoichet, M.S. Antibody-Antisense Oligonucleotide Conjugate Downregulates a Key Gene in Glioblastoma Stem Cells. Mol. Ther. Nucleic Acids 2018, 11, 518–527. [Google Scholar] [CrossRef] [Green Version]
- Nuzzo, S.; Roscigno, G.; Affinito, A.; Ingenito, F.; Quintavalle, C.; Condorelli, G. Potential and Challenges of Aptamers as Specific Carriers of Therapeutic Oligonucleotides for Precision Medicine in Cancer. Cancers 2019, 11, 1521. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- White, R.R.; Sullenger, B.A.; Rusconi, C.P. Developing aptamers into therapeutics. J. Clin. Investig. 2000, 106, 929–934. [Google Scholar] [CrossRef] [Green Version]
- McClorey, G.; Banerjee, S. Cell-Penetrating Peptides to Enhance Delivery of Oligonucleotide-Based Therapeutics. Biomedicines 2018, 6, 51. [Google Scholar] [CrossRef] [Green Version]
- Nikan, M.; Tanowitz, M.; Dwyer, C.A.; Jackson, M.; Gaus, H.J.; Swayze, E.E.; Rigo, F.; Seth, P.P.; Prakash, T.P. Targeted Delivery of Antisense Oligonucleotides Using Neurotensin Peptides. J. Med. Chem. 2020, 63, 8471–8484. [Google Scholar] [CrossRef]
- Ämmälä, C.; Drury, W.J.; Knerr, L.; Ahlstedt, I.; Stillemark-Billton, P.; Wennberg-Huldt, C.; Andersson, E.-M.; Valeur, E.; Jansson-Löfmark, R.; Janzén, D.; et al. Targeted delivery of antisense oligonucleotides to pancreatic β-cells. Sci. Adv. 2018, 4, eaat3386. [Google Scholar] [CrossRef] [Green Version]
- Blain, A.M.; Greally, E.; McClorey, G.; Manzano, R.; Betts, C.A.; Godfrey, C.; O’Donovan, L.; Coursindel, T.; Gait, M.J.; Wood, M.J.; et al. Peptide-conjugated phosphodiamidate oligomer-mediated exon skipping has benefits for cardiac function in mdx and Cmah-/-mdx mouse models of Duchenne muscular dystrophy. PLoS ONE 2018, 13, e0198897. [Google Scholar] [CrossRef] [PubMed]
- Tsoumpra, M.K.; Fukumoto, S.; Matsumoto, T.; Takeda, S.; Wood, M.J.; Aoki, Y. Peptide-conjugate antisense based splice-correction for Duchenne muscular dystrophy and other neuromuscular diseases. EBioMedicine 2019, 45, 630–645. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Miller, C.M.; Tanowitz, M.; Donner, A.J.; Prakash, T.P.; Swayze, E.E.; Harris, E.N.; Seth, P.P. Receptor-Mediated Uptake of Phosphorothioate Antisense Oligonucleotides in Different Cell Types of the Liver. Nucleic Acid Ther. 2018, 28, 119–127. [Google Scholar] [CrossRef]
- 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]
- 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.J.; et al. MTL-CEBPA, a Small Activating RNA Therapeutic Upregulating C/EBP-α, 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] [PubMed]
- Reebye, V.; Huang, K.-W.; Lin, V.; Jarvis, S.; Cutilas, P.; Dorman, S.; Ciriello, S.; Andrikakou, P.; Voutila, J.; Sætrom, 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]
- Stein, C.A.; Castanotto, D. FDA-Approved Oligonucleotide Therapies in 2017. Mol. Ther. 2017, 25, 1069–1075. [Google Scholar] [CrossRef] [Green Version]
- Goldberg, A.C.; Hopkins, P.N.; Toth, P.P.; Ballantyne, C.M.; Rader, D.J.; Robinson, J.G.; Daniels, S.R.; Gidding, S.S.; de Ferranti, S.D.; Ito, M.K.; et al. Familial Hypercholesterolemia: Screening, diagnosis and management of pediatric and adult patients: Clinical guidance from the National Lipid Association Expert Panel on Familial Hypercholesterolemia. J. Clin. Lipidol. 2011, 5, S1–S8. [Google Scholar] [CrossRef]
- Hair, P.; Cameron, F.; McKeage, K. Mipomersen Sodium: First Global Approval. Drugs 2013, 73, 487–493. [Google Scholar] [CrossRef]
- Raal, F.J.; Santos, R.D.; Blom, D.; Marais, A.D.; Charng, M.-J.; Cromwell, W.; Lachmann, R.; Gaudet, D.; Tan, J.L.; Chasan-Taber, S.; et al. Mipomersen, an apolipoprotein B synthesis inhibitor, for lowering of LDL cholesterol concentrations in patients with homozygous familial hypercholesterolaemia: A randomised, double-blind, placebo-controlled trial. Lancet 2010, 375, 998–1006. [Google Scholar] [CrossRef]
- Akinc, A.; Maier, M.A.; Manoharan, M.; Fitzgerald, K.; Jayaraman, M.; Barros, S.; Ansell, S.; Du, X.; Hope, M.J.; Madden, T.D.; et al. The Onpattro story and the clinical translation of nanomedicines containing nucleic acid-based drugs. Nat. Nanotechnol. 2019, 14, 1084–1087. [Google Scholar] [CrossRef] [PubMed]
- Bissell, D.M.; Anderson, K.E.; Bonkovsky, H.L. Porphyria. N. Engl. J. Med. 2017, 377, 862–872. [Google Scholar] [CrossRef] [PubMed]
- Scott, L.J. Givosiran: First Approval. Drugs 2020, 80, 335–339. [Google Scholar] [CrossRef]
- Shilling, R.; Karsten, V.; Silliman, N.; Chen, J.; Li, W.; Vest, J. Study Design and Rationale of Helios-B: A Phase 3 Study to Evaluate the Clinical Efficacy and Safety of Vutrisiran in Patients with Attr Amyloidosis with Cardiomyopathy. J. Am. Coll. Cardiol. 2020, 75, 3579. [Google Scholar] [CrossRef]
- Finkbeiner, S. Huntington’s Disease. Cold Spring Harb. Perspect. Biol. 2011, 3, a007476. [Google Scholar] [CrossRef] [Green Version]
- O’Donovan, M.C. A novel gene containing a trinucleotide repeat that is expanded and unstable on Huntington’s disease chromosomes. The Huntington’s Disease Collaborative Research Group. Cell 1993, 72, 971–983. [Google Scholar] [CrossRef]
- Witztum, J.L.; Gaudet, D.; Freedman, S.D.; Alexander, V.J.; Digenio, A.; Williams, K.R.; Yang, Q.; Hughes, S.G.; Geary, R.S.; Arca, M.; et al. Volanesorsen and Triglyceride Levels in Familial Chylomicronemia Syndrome. N. Engl. J. Med. 2019, 381, 531–542. [Google Scholar] [CrossRef] [PubMed]
- Bartel, D.P. Metazoan MicroRNAs. Cell 2018, 173, 20–51. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ottosen, S.; Parsley, T.B.; Yang, L.; Zeh, K.; Van Doorn, L.-J.; Van Der Veer, E.; Raney, A.K.; Hodges, M.R.; Patick, A.K. In VitroAntiviral Activity and Preclinical and Clinical Resistance Profile of Miravirsen, a Novel Anti-Hepatitis C Virus Therapeutic Targeting the Human Factor miR-122. Antimicrob. Agents Chemother. 2015, 59, 599–608. [Google Scholar] [CrossRef] [Green Version]
- Janssen, H.L.A.; Reesink, H.W.; Lawitz, E.J.; Zeuzem, S.; Rodriguez-Torres, M.; Patel, K.; Van Der Meer, A.J.; Patick, A.K.; Chen, A.; Zhou, Y.; et al. Treatment of HCV Infection by Targeting MicroRNA. N. Engl. J. Med. 2013, 368, 1685–1694. [Google Scholar] [CrossRef] [Green Version]
- Chakraborty, C.; Sharma, A.R.; Sharma, G.; Doss, C.G.P.; Lee, S.-S. Therapeutic miRNA and siRNA: Moving from Bench to Clinic as Next Generation Medicine. Mol. Ther. Nucleic Acids 2017, 8, 132–143. [Google Scholar] [CrossRef] [Green Version]
- Holshue, M.L.; DeBolt, C.; Lindquist, S.; Lofy, K.H.; Wiesman, J.; Bruce, H.; Spitters, C.; Ericson, K.; Wilkerson, S.; Tural, A.; et al. First Case of 2019 Novel Coronavirus in the United States. N. Engl. J. Med. 2020, 382, 929–936. [Google Scholar] [CrossRef]
- Chung, Y.H.; Beiss, V.; Fiering, S.N.; Steinmetz, N.F. COVID-19 Vaccine Frontrunners and Their Nanotechnology Design. ACS Nano 2020, 14, 12522–12537. [Google Scholar] [CrossRef]
- Beigel, J.H.; Tomashek, K.M.; Dodd, L.E.; Mehta, A.K.; Zingman, B.S.; Kalil, A.C.; Hohmann, E.; Chu, H.Y.; Luetkemeyer, A.; Kline, S.; et al. Remdesivir for the Treatment of COVID-19—Final report. N. Engl. J. Med. 2020, 383, 1813–1826. [Google Scholar] [CrossRef]
- Crooke, S.T.; Baker, B.F.; Crooke, R.M.; Liang, X.-H. Antisense technology: An overview and prospectus. Nat. Rev. Drug Discov. 2021, 20, 427–453. [Google Scholar] [CrossRef] [PubMed]
- Iversen, P.L.; Warren, T.K.; Wells, J.B.; Garza, N.L.; Mourich, D.V.; Welch, L.S.; Panchal, R.G.; Bavari, S. Discovery and Early Development of AVI-7537 and AVI-7288 for the Treatment of Ebola Virus and Marburg Virus Infections. Viruses 2012, 4, 2806–2830. [Google Scholar] [CrossRef] [PubMed]
- Orr, R.M. Technology evaluation: Fomivirsen, Isis Pharmaceuticals Inc/CIBA vision. Curr. Opin. Mol. Ther. 2001, 3, 288–294. [Google Scholar] [PubMed]
- Chery, J.; Petri, A.; Wagschal, A.; Lim, S.-Y.; Cunningham, J.; Vasudevan, S.; Kauppinen, S.; Näär, A.M. Development of Locked Nucleic Acid Antisense Oligonucleotides Targeting Ebola Viral Proteins and Host Factor Niemann-Pick C1. Nucleic Acid Ther. 2018, 28, 273–284. [Google Scholar] [CrossRef]
- Lenartowicz, E.; Nogales, A.; Kierzek, E.; Kierzek, R.; Martínez-Sobrido, L.; Turner, D.H. Antisense Oligonucleotides Targeting Influenza A Segment 8 Genomic RNA Inhibit Viral Replication. Nucleic Acid Ther. 2016, 26, 277–285. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Javanbakht, H.; Mueller, H.; Walther, J.; Zhou, X.; Lopez, A.; Pattupara, T.; Blaising, J.; Pedersen, L.; Albæk, N.; Jackerott, M.; et al. Liver-Targeted Anti-HBV Single-Stranded Oligonucleotides with Locked Nucleic Acid Potently Reduce HBV Gene Expression In Vivo. Mol. Ther. Nucleic Acids 2018, 11, 441–454. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- De Jong, Y.P.; Jacobson, I.M. Antisense therapy for hepatitis C virus infection. J. Hepatol. 2014, 60, 227–228. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, L.; Li, Q.; Ding, X.; Zhang, B.; Zhang, Q.; Qu, X.; Huo, Y.; Yang, J.; Wang, S. Antisense Oligonucleotides Targeting Raf-1 Block Japanese Encephalitis Virus In Vitro and In Vivo. Nucleic Acid Ther. 2017, 27, 78–86. [Google Scholar] [CrossRef]
- Crooke, S.T.E.; Ecker, D.J.; Sampath, R.; Freier, S.M.; Massire, C.; Hofstadler, S.A.; Lowery, K.S.; Swayze, E.E.; Baker, B.F.; Bennett, F.C. Compositions and Methods for the Treatment of Severe Acute Respiratory Syndrome (Sars). International Patent Application No. WO2005023083A3, 4 March 2004. [Google Scholar]
- Smith Alvin, W.; Stein David, A.; Skilling Douglas, E. Antisense antiviral agent and method for treating ssRNA Viral Infection. 2004. Available online: https://www.surechembl.org/document/CA-2522508-A1 (accessed on 17 December 2021).
- Kim, D.; Lee, J.-Y.; Yang, J.-S.; Kim, J.W.; Kim, V.N.; Chang, H. The Architecture of SARS-CoV-2 Transcriptome. Cell 2020, 181, 914–921.e10. [Google Scholar] [CrossRef]
- Barrey, E.B.; Dhorne-Pollet, S.; Eléouët, J.; Delmas, B. Think Different with RNA Therapy: Can Antisense Oligonucleotides Be Used to Inhibit Replication and Transcription of SARS-Cov-2? Preprints 2020. [Google Scholar] [CrossRef] [Green Version]
- Kirby, T. New variant of SARS-CoV-2 in UK causes surge of COVID-19. Lancet Respir. Med. 2021, 9, e20–e21. [Google Scholar] [CrossRef]
- Callaway, E. Fast-spreading COVID variant can elude immune responses. Nature 2021, 589, 500–501. [Google Scholar] [CrossRef]
- Kuzmina, A.; Khalaila, Y.; Voloshin, O.; Keren-Naus, A.; Boehm-Cohen, L.; Raviv, Y.; Shemer-Avni, Y.; Rosenberg, E.; Taube, R. SARS-CoV-2 spike variants exhibit differential infectivity and neutralization resistance to convalescent or post-vaccination sera. Cell Host Microbe 2021, 29, 522–528.e2. [Google Scholar] [CrossRef]
- Coutinho, R.M.; Marquitti, F.M.D.; Ferreira, L.S.; Borges, M.E.; da Silva, R.L.P.; Canton, O.; Portella, T.P.; Lyra, S.P.; Franco, C.; da Silva, A.A.M. Model-based evaluation of transmissibility and reinfection for the P. 1 variant of the SARS-CoV-2. medRxiv 2021, medRxiv: 2021.2003.2003.21252706. [Google Scholar] [CrossRef]
- Davies, N.G.; Jarvis, C.I.; Edmunds, W.J.; Jewell, N.P.; Diaz-Ordaz, K.; Keogh, R.H. Increased mortality in community-tested cases of SARS-CoV-2 lineage B.1.1.7. Nature 2021, 593, 270–274. [Google Scholar] [CrossRef] [PubMed]
- Korber, B.; Fischer, W.M.; Gnanakaran, S.; Yoon, H.; Theiler, J.; Abfalterer, W.; Hengartner, N.; Giorgi, E.E.; Bhattacharya, T.; Foley, B.; et al. Tracking Changes in SARS-CoV-2 Spike: Evidence that D614G Increases Infectivity of the COVID-19 Virus. Cell 2020, 182, 812–827.e19. [Google Scholar] [CrossRef] [PubMed]
- Wang, L.; Wang, L.; Zhuang, H. Profiling and characterization of SARS-CoV-2 mutants’ infectivity and antigenicity. Signal Transduct. Target. Ther. 2020, 5, 185. [Google Scholar] [CrossRef]
- Neuman, B.W.; Stein, D.A.; Kroeker, A.D.; Churchill, M.J.; Kim, A.M.; Kuhn, P.; Dawson, P.; Moulton, H.M.; Bestwick, R.K.; Iversen, P.; et al. Inhibition, Escape, and Attenuated Growth of Severe Acute Respiratory Syndrome Coronavirus Treated with Antisense Morpholino Oligomers. J. Virol. 2005, 79, 9665–9676. [Google Scholar] [CrossRef] [Green Version]
- Rangan, R.; Zheludev, I.N.; Hagey, R.J.; Pham, E.A.; Wayment-Steele, H.K.; Glenn, J.S.; Das, R. RNA genome conservation and secondary structure in SARS-CoV-2 and SARS-related viruses: A first look. RNA 2020, 26, 937–959. [Google Scholar] [CrossRef]
- Tengs, T.; Jonassen, C.M. Distribution and Evolutionary History of the Mobile Genetic Element s2m in Coronaviruses. Diseases 2016, 4, 27. [Google Scholar] [CrossRef]
- Yeh, T.Y.; Contreras, G.P. Emerging viral mutants in Australia suggest RNA recombination event in the SARS -CoV-2 genome. Med. J. Aust. 2020, 213, 44.e1. [Google Scholar] [CrossRef]
- Vahed, M.; Vahed, M.; Sweeney, A.; Shirazi, F.H.; Mirsaeidi, M. Mutation in position of 32 (G>U) of S2M differentiate human SARS-CoV2 from Bat Coronavirus. bioRxiv 2020, bioRxiv:2020.2009.2002.280529. [Google Scholar] [CrossRef]
- Lulla, V.; Wandel, M.P.; Bandyra, K.J.; Ulferts, R.; Wu, M.; Dendooven, T.; Yang, X.; Doyle, N.; Oerum, S.; Beale, R.; et al. Targeting the Conserved Stem Loop 2 Motif in the SARS-CoV-2 Genome. J. Virol. 2021, 95, e0066321. [Google Scholar] [CrossRef] [PubMed]
- Manfredonia, I.; Nithin, C.; Ponce-Salvatierra, A.; Ghosh, P.; Wirecki, T.K.; Marinus, T.; Ogando, N.S.; Snijder, E.J.; van Hemert, M.J.; Bujnicki, J.M.; et al. Genome-wide mapping of SARS-CoV-2 RNA structures identifies therapeutically-relevant elements. Nucleic Acids Res. 2020, 48, 12436–12452. [Google Scholar] [CrossRef] [PubMed]
- Lan, T.C.T. Insights into the secondary structural ensembles of the full SARS-CoV-2 RNA genome in infected cells. bioRxiv 2021, bioRxiv: 2020.2006.2029.178343. [Google Scholar] [CrossRef]
- Huston, N.C.; Wan, H.; Strine, M.S.; Tavares, R.D.C.A.; Wilen, C.B.; Pyle, A.M. Comprehensive in vivo secondary structure of the SARS-CoV-2 genome reveals novel regulatory motifs and mechanisms. Mol. Cell 2021, 81, 584–598.e5. [Google Scholar] [CrossRef]
- Sun, L.; Li, P.; Ju, X.; Rao, J.; Huang, W.; Ren, L.; Zhang, S.; Xiong, T.; Xu, K.; Zhou, X.; et al. In vivo structural characterization of the SARS-CoV-2 RNA genome identifies host proteins vulnerable to repurposed drugs. Cell 2021, 184, 1865–1883.e20. [Google Scholar] [CrossRef]
- Beumer, W.; Swildens, J.; Leal, T.; Noel, S.; Anthonijsz, H.; Van Der Horst, G.; Kuiperij-Boersma, H.; Potman, M.; Van Putten, C.; Biasutto, P.; et al. Evaluation of eluforsen, a novel RNA oligonucleotide for restoration of CFTR function in in vitro and murine models of p.Phe508del cystic fibrosis. PLoS ONE 2019, 14, e0219182. [Google Scholar] [CrossRef] [PubMed]
- Fey, R.A.; Templin, M.V.; McDonald, J.D.; Yu, R.Z.; Hutt, J.A.; Gigliotti, A.P.; Henry, S.P.; Reed, M.D. Local and systemic tolerability of a 2′O-methoxyethyl antisense oligonucleotide targeting interleukin-4 receptor-α delivery by inhalation in mouse and monkey. Inhal. Toxicol. 2014, 26, 452–463. [Google Scholar] [CrossRef]
- Crosby, J.R.; Zhao, C.; Jiang, C.; Bai, D.; Katz, M.; Greenlee, S.; Kawabe, H.; McCaleb, M.; Rotin, D.; Guo, S.; et al. Inhaled ENaC antisense oligonucleotide ameliorates cystic fibrosis-like lung disease in mice. J. Cyst. Fibros. 2017, 16, 671–680. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Allakhverdi, Z.; Allam, M.; Guimond, A.; Ferrari, N.; Zemzoumi, K.; Seguin, R.; Paquet, L.; Renzi, P.M. Multitargeted Approach Using Antisense Oligonucleotides for the Treatment of Asthma. Ann. N. Y. Acad. Sci. 2006, 1082, 62–73. [Google Scholar] [CrossRef] [PubMed]
- Zhu, C.; Lee, J.Y.; Woo, J.Z.; Xu, L.; Nguyenla, X.; Yamashiro, L.H.; Ji, F.; Biering, S.B.; Van Dis, E.; Gonzalez, F.; et al. An intranasal ASO therapeutic targeting SARS-CoV-2. bioRxiv 2021, bioRxiv:2021.2005.2017.444397. [Google Scholar] [CrossRef]
- Zhou, P.; Yang, X.-L.; Wang, X.-G.; Hu, B.; Zhang, L.; Zhang, W.; Si, H.-R.; Zhu, Y.; Li, B.; Huang, C.-L.; et al. A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature 2020, 579, 270–273. [Google Scholar] [CrossRef] [Green Version]
- Tai, W.; He, L.; Zhang, X.; Pu, J.; Voronin, D.; Jiang, S.; Zhou, Y.; Du, L. Characterization of the receptor-binding domain (RBD) of 2019 novel coronavirus: Implication for development of RBD protein as a viral attachment inhibitor and vaccine. Cell. Mol. Immunol. 2020, 17, 613–620. [Google Scholar] [CrossRef] [Green Version]
- Rehman, S.U.; Tabish, M. Alternative splicing of ACE2 possibly generates variants that may limit the entry of SARS-CoV-2: A potential therapeutic approach using SSOs. Clin. Sci. 2020, 134, 1143–1150. [Google Scholar] [CrossRef]
- Su, X.; Ma, W.; Feng, D.; Cheng, B.; Wang, Q.; Guo, Z.; Zhou, D.; Tang, X. Efficient Inhibition of SARS-CoV-2 Using Chimeric Antisense Oligonucleotides through RNase L Activation. Angew. Chem. Int. Ed. Engl. 2021, 60, 21662–21667. [Google Scholar] [CrossRef]
- Li, Y.; Garcia, G.; Arumugaswami, V.; Guo, F. Structure-based design of antisense oligonucleotides that inhibit SARS-CoV-2 replication. bioRxiv 2021. [Google Scholar] [CrossRef]
- Zhang, K.; Zheludev, I.N.; Hagey, R.J.; Wu, M.T.P.; Haslecker, R.; Hou, Y.J.; Kretsch, R.; Pintilie, G.D.; Rangan, R.; Kladwang, W.; et al. Cryo-electron Microscopy and Exploratory Antisense Targeting of the 28-kDa Frameshift Stimulation Element from the SARS-CoV-2 RNA Genome. bioRxiv 2020. [Google Scholar] [CrossRef]
- Messner, C.B.; Demichev, V.; Wendisch, D.; Michalick, L.; White, M.; Freiwald, A.; Textoris-Taube, K.; Vernardis, S.I.; Egger, A.-S.; Kreidl, M.; et al. Ultra-High-Throughput Clinical Proteomics Reveals Classifiers of COVID-19 Infection. Cell Syst. 2020, 11, 11–24.e4. [Google Scholar] [CrossRef] [PubMed]
- Moitra, P.; Alafeef, M.; Dighe, K.; Frieman, M.B.; Pan, D. Selective Naked-Eye Detection of SARS-CoV-2 Mediated by N Gene Targeted Antisense Oligonucleotide Capped Plasmonic Nanoparticles. ACS Nano 2020, 14, 7617–7627. [Google Scholar] [CrossRef]
- Wheeler, T.M.; Leger, A.J.; Pandey, S.K.; MacLeod, A.R.; Nakamori, M.; Cheng, S.H.; Wentworth, B.M.; Bennett, C.F.; Thornton, C.A. Targeting nuclear RNA for in vivo correction of myotonic dystrophy. Nature 2012, 488, 111–115. [Google Scholar] [CrossRef] [Green Version]
- Messina, A.; Langlet, F.; Chachlaki, K.; Roa, J.; Rasika, S.; Jouy, N.; Gallet, S.; Gaytan, F.; Parkash, J.; Tena-Sempere, M.; et al. A microRNA switch regulates the rise in hypothalamic GnRH production before puberty. Nat. Neurosci. 2016, 19, 835–844. [Google Scholar] [CrossRef]
- Frischmeyer, P.A.; Dietz, H.C. Nonsense-mediated mRNA decay in health and disease. Hum. Mol. Genet. 1999, 8, 1893–1900. [Google Scholar] [CrossRef] [Green Version]
- Huang, L.; Low, A.; Damle, S.S.; Keenan, M.M.; Kuntz, S.; Murray, S.F.; Monia, B.P.; Guo, S. Antisense suppression of the nonsense mediated decay factor Upf3b as a potential treatment for diseases caused by nonsense mutations. Genome Biol. 2018, 19, 4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Harigaya, Y.; Parker, R. No-go decay: A quality control mechanism for RNA in translation. Wiley Interdiscip. Rev. RNA 2010, 1, 132–141. [Google Scholar] [CrossRef] [PubMed]
- Liang, X.-H.; Nichols, J.G.; Hsu, C.-W.; Vickers, T.A.; Crooke, S.T. mRNA levels can be reduced by antisense oligonucleotides via no-go decay pathway. Nucleic Acids Res. 2019, 47, 6900–6916. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gilot, D.; Migault, M.; Bachelot, L.; Journé, F.; Rogiers, A.; Donnou-Fournet, E.; Mogha, A.; Mouchet, N.; Pinel-Marie, M.-L.; Mari, B.; et al. A non-coding function of TYRP1 mRNA promotes melanoma growth. Nat. Cell Biol. 2017, 19, 1348–1357. [Google Scholar] [CrossRef] [PubMed]
Drug Name (Company) | Company | Chemistry | Design (Mix-Gap) | Disease | Date FDA Approval Stage of Trial | Administration |
---|---|---|---|---|---|---|
Formirsen (Vitraven®) | Ionis Pharmaceuticals & Novartis Ophthalmics | PS & 2′O-MOE | First generation | Cytomegalovirus (CMV) retinitis | 1999 (FDA approval)—withdrawn (2006 in USA) | Intravitreal |
Mipomersen (Kinamro®) | Grenzyme | PS | 2nd generation—Gapmer | Homozygous familial hypercholesterolemia | 2013 | Intravenous |
Patisiran (Onpattro®) | Alnylam | siRNA | Double-stranded small interfering RNA encapsulated in a lipid nanoparticle | Hereditary transtherthyretin- mediated amyloidosis | 2018 | intravenous |
Inotersen (Tegsedi®) | Ionis Pharmaceuticals | PS & 2′O-MOE | 2nd generation—Gapmer | Hereditary transtherthyretin-mediated amyloidosis | 2018 | intravenous |
Givosiran (Givlaari®) | Alnylam Pharmaceuticals | siRNA | siRNA—conjugated to Gal-Nac | Acute hepatic porphyria | 2019 | intravenous |
Vutrisiran | Alnylam Pharmaceutical | siRNA | siRNA—conjugated to Gal-Nac | Hereditary transtherthyretin-mediated amyloidosis | Phase 3 | subcutaneous |
Volanesorsen (Waylivra®) | Ionis Pharmaceuticals | 2’-MOE | 2nd generation | familial chylomicronemia syndrome | Phase 3/EMA approved 2019 | sub cutaneous |
Miravirsen | Roche/Santaris | LNA & PS | 3rd-generation anti-miRNA | Hepatitis C virus infection | Phase 3 | ND |
RG-101 | Regulus Therapeutics | PS coupled to an N-acetylgalactosamine group | Hepatitis C virus infection | Phase 3 | ND | |
Pegaptanib (Macugen®) | OSI Pharmaceuticals | Aptamer | Aptamer | Neovascular age-related macular degeneration | 2004 | intravitreal |
Eteplirsen (Exondys 51®) | Sarepta Therapeutics | Phosphorodiamidate morpholino oligomer | 3rd generation | Duchenne muscular dystrophy | 2016 | intravenous |
Nusinersen (Spinraza®) | Ionis Pharmaceuticals, Biogen | PS & 2’-MOE | 3rd generation | Spinal muscular atrophy | 2016 | intrathecal |
Defibrotide | Jazz Pharmaceuticals | Mixture of single-stranded and double-stranded phosphodiester oligonucleotides | Aptamer | Veno-occlusive disease in liver | 2016 | intravenous |
Inotersen (Tegsedi®) | Akcea Therapeutics | 2’-MOE | 2nd generation | Polyneuropathy caused by hereditary transthyretin-mediated (hATTR) amyloidosis | 2018 | subcutaneous |
Milasen | Boston Children’s Hospital | PS & 2’-MOE | 2nd generation | Mila Makovec’s CLN7 gene associated with Batten disease | 2018 | intravenous |
Patisiran (Onpattro®) | Alnylam | PS & 2’-MOE | Gapmer | Polyneuropathy caused by hATTR amyloidosis | 2018 | intravenous |
Golodirsen (Vyondys 53®) | Sarepta Therapeutics | PMO | 3rd generation | Duchenne muscular dystrophy | 2019 | intravenous |
Givosiran (Givlaari®) | Alnylam | Gal-Nac-2’OMe | Acute hepatic porphyria (AHP) | 2019 | intravenous | |
Viltolarsen (Viltepso®) | NS Pharma | PMO | 3rd generation | Duchenne muscular dystrophy | 2020 | intravenous |
Casimersen (Amondys 45®) | Sarepta Therapeutics | PMO | 3rd generation | Duchenne muscular dystrophy | 2021 | intravenous |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 by the authors. 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
Quemener, A.M.; Centomo, M.L.; Sax, S.L.; Panella, R. Small Drugs, Huge Impact: The Extraordinary Impact of Antisense Oligonucleotides in Research and Drug Development. Molecules 2022, 27, 536. https://doi.org/10.3390/molecules27020536
Quemener AM, Centomo ML, Sax SL, Panella R. Small Drugs, Huge Impact: The Extraordinary Impact of Antisense Oligonucleotides in Research and Drug Development. Molecules. 2022; 27(2):536. https://doi.org/10.3390/molecules27020536
Chicago/Turabian StyleQuemener, Anais M., Maria Laura Centomo, Scott L. Sax, and Riccardo Panella. 2022. "Small Drugs, Huge Impact: The Extraordinary Impact of Antisense Oligonucleotides in Research and Drug Development" Molecules 27, no. 2: 536. https://doi.org/10.3390/molecules27020536
APA StyleQuemener, A. M., Centomo, M. L., Sax, S. L., & Panella, R. (2022). Small Drugs, Huge Impact: The Extraordinary Impact of Antisense Oligonucleotides in Research and Drug Development. Molecules, 27(2), 536. https://doi.org/10.3390/molecules27020536