Recent Advances in RNA Therapy and Its Carriers to Treat the Single-Gene Neurological Disorders
Abstract
:1. Introduction
2. RNA-Based Therapy
2.1. Therapeutic mRNA for Protein Replacement
2.2. Single-Stranded Oligonucleotide as a Therapeutic Agent
3. Blood Brain Barrier and Molecular Carriers
3.1. Blood Brain Barrier
3.2. Brain Drug Delivery Strategies
3.2.1. Lipid-Based Nanocarriers
3.2.2. Extracellular Vesicles
3.2.3. Viral Carriers
4. RNA Therapies to Treat Single-Gene Neurological Disorders
4.1. Familial Amyloid Polyneuropathy
4.2. Acute Intermittent Porphyria
4.3. Spinal Muscular Atrophy
4.4. Duchenne Muscular Dystrophy
4.5. Huntington’s Disease and Other Neurodegenerative Disorders That Are Caused by Microsatellite Repeat Expansion
4.6. Lysosomal Storage Diseases
5. Limitations and Future Potentials of RNA Therapies
6. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Acknowledgments
Conflicts of Interest
References
- International Human Genome Sequencing, C. Finishing the euchromatic sequence of the human genome. Nature 2004, 431, 931–945. [Google Scholar] [CrossRef]
- Lander, E.S.; Linton, L.M.; Birren, B.; Nusbaum, C.; Zody, M.C.; Baldwin, J.; Devon, K.; Dewar, K.; Doyle, M.; FitzHugh, W.; et al. Initial sequencing and analysis of the human genome. Nature 2001, 409, 860–921. [Google Scholar] [CrossRef] [Green Version]
- Berger, A.H.; Pandolfi, P.P. Haplo-insufficiency: A driving force in cancer. J. Pathol. 2011, 223, 137–146. [Google Scholar] [CrossRef]
- Datta, N.; Chakraborty, S.; Basu, M.; Ghosh, M.K. Tumor Suppressors Having Oncogenic Functions: The Double Agents. Cells 2020, 10, 46. [Google Scholar] [CrossRef] [PubMed]
- Boudewyn, L.C.; Walkley, S.U. Current concepts in the neuropathogenesis of mucolipidosis type IV. J. Neurochem. 2019, 148, 669–689. [Google Scholar] [CrossRef] [Green Version]
- Schoser, B. Pompe disease: What are we missing? Ann. Transl. Med. 2019, 7, 292. [Google Scholar] [CrossRef] [PubMed]
- Snell, R.G.; MacMillan, J.C.; Cheadle, J.P.; Fenton, I.; Lazarou, L.P.; Davies, P.; MacDonald, M.E.; Gusella, J.F.; Harper, P.S.; Shaw, D.J. Relationship between trinucleotide repeat expansion and phenotypic variation in Huntington’s disease. Nat. Genet. 1993, 4, 393–397. [Google Scholar] [CrossRef]
- Kleinjan, D.A.; van Heyningen, V. Long-range control of gene expression: Emerging mechanisms and disruption in disease. Am. J. Hum. Genet. 2005, 76, 8–32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sahin, U.; Kariko, K.; Tureci, O. mRNA-based therapeutics--developing a new class of drugs. Nat. Rev. Drug Discov. 2014, 13, 759–780. [Google Scholar] [CrossRef]
- Gan, L.M.; Lagerstrom-Fermer, M.; Carlsson, L.G.; Arfvidsson, C.; Egnell, A.C.; Rudvik, A.; Kjaer, M.; Collen, A.; Thompson, J.D.; Joyal, J.; et al. Intradermal delivery of modified mRNA encoding VEGF-A in patients with type 2 diabetes. Nat. Commun. 2019, 10, 871. [Google Scholar] [CrossRef]
- Hewitt, S.L.; Bai, A.; Bailey, D.; Ichikawa, K.; Zielinski, J.; Karp, R.; Apte, A.; Arnold, K.; Zacharek, S.J.; Iliou, M.S.; et al. Durable anticancer immunity from intratumoral administration of IL-23, IL-36gamma, and OX40L mRNAs. Sci. Transl. Med. 2019, 11, eaat9143. [Google Scholar] [CrossRef]
- Pastor, F.; Berraondo, P.; Etxeberria, I.; Frederick, J.; Sahin, U.; Gilboa, E.; Melero, I. An RNA toolbox for cancer immunotherapy. Nat. Rev. Drug Discov. 2018, 17, 751–767. [Google Scholar] [CrossRef]
- Baldo, B.A. Enzymes approved for human therapy: Indications, mechanisms and adverse effects. BioDrugs 2015, 29, 31–55. [Google Scholar] [CrossRef]
- Berraondo, P.; Martini, P.G.V.; Avila, M.A.; Fontanellas, A. Messenger RNA therapy for rare genetic metabolic diseases. Gut 2019, 68, 1323–1330. [Google Scholar] [CrossRef] [Green Version]
- Azevedo, O.; Gago, M.F.; Miltenberger-Miltenyi, G.; Sousa, N.; Cunha, D. Fabry Disease Therapy: State-of-the-Art and Current Challenges. Int. J. Mol. Sci. 2020, 22, 206. [Google Scholar] [CrossRef]
- Seker Yilmaz, B.; Davison, J.; Jones, S.A.; Baruteau, J. Novel therapies for mucopolysaccharidosis type III. J. Inherit. Metab. Dis. 2021, 44, 129–147. [Google Scholar] [CrossRef] [PubMed]
- de Moor, C.H.; Richter, J.D. Translational control in vertebrate development. Int. Rev. Cytol. 2001, 203, 567–608. [Google Scholar] [CrossRef] [PubMed]
- Wang, Z.; Day, N.; Trifillis, P.; Kiledjian, M. An mRNA stability complex functions with poly(A)-binding protein to stabilize mRNA in vitro. Mol. Cell Biol. 1999, 19, 4552–4560. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Santos-Reboucas, C.B.; Pimentel, M.M. MicroRNAs: Macro challenges on understanding human biological functions and neurological diseases. Curr. Mol. Med. 2010, 10, 692–704. [Google Scholar] [CrossRef]
- Gaspar, P.; Moura, G.; Santos, M.A.; Oliveira, J.L. mRNA secondary structure optimization using a correlated stem-loop prediction. Nucleic Acids Res. 2013, 41, e73. [Google Scholar] [CrossRef] [Green Version]
- Kariko, 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] [PubMed]
- Scharner, J.; Aznarez, I. Clinical Applications of Single-Stranded Oligonucleotides: Current Landscape of Approved and In-Development Therapeutics. Mol. Ther. 2021, 29, 540–554. [Google Scholar] [CrossRef] [PubMed]
- Zhu, G.; Chen, X. Aptamer-based targeted therapy. Adv. Drug Deliv. Rev. 2018, 134, 65–78. [Google Scholar] [CrossRef]
- Kole, R.; Krainer, A.R.; Altman, S. RNA therapeutics: Beyond RNA interference and antisense oligonucleotides. Nat. Rev. Drug Discov. 2012, 11, 125–140. [Google Scholar] [CrossRef] [Green Version]
- Koller, E.; Vincent, T.M.; Chappell, A.; De, S.; Manoharan, M.; Bennett, C.F. Mechanisms of single-stranded phosphorothioate modified antisense oligonucleotide accumulation in hepatocytes. Nucleic Acids Res. 2011, 39, 4795–4807. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Roehr, B. Fomivirsen approved for CMV retinitis. J. Int. Assoc. Physicians AIDS Care 1998, 4, 14–16. [Google Scholar] [PubMed]
- Geary, R.S.; Baker, B.F.; Crooke, S.T. Clinical and preclinical pharmacokinetics and pharmacodynamics of mipomersen (kynamro®): A second-generation antisense oligonucleotide inhibitor of apolipoprotein B. Clin. Pharmacokinet. 2015, 54, 133–146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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] [PubMed] [Green Version]
- Bennett, C.F.; Swayze, E.E. RNA targeting therapeutics: Molecular mechanisms of antisense oligonucleotides as a therapeutic platform. Annu. Rev. Pharmacol. Toxicol. 2010, 50, 259–293. [Google Scholar] [CrossRef]
- Asami, Y.; Nagata, T.; Yoshioka, K.; Kunieda, T.; Yoshida-Tanaka, K.; Bennett, C.F.; Seth, P.P.; Yokota, T. Efficient Gene Suppression by DNA/DNA Double-Stranded Oligonucleotide In Vivo. Mol. Ther. 2021, 29, 838–847. [Google Scholar] [CrossRef]
- Nishina, K.; Piao, W.; Yoshida-Tanaka, K.; Sujino, Y.; Nishina, T.; Yamamoto, T.; Nitta, K.; Yoshioka, K.; Kuwahara, H.; Yasuhara, H.; et al. DNA/RNA heteroduplex oligonucleotide for highly efficient gene silencing. Nat. Commun. 2015, 6, 7969. [Google Scholar] [CrossRef] [Green Version]
- Lei Mon, S.S.; Yoshioka, K.; Jia, C.; Kunieda, T.; Asami, Y.; Yoshida-Tanaka, K.; Piao, W.; Kuwahara, H.; Nishina, K.; Nagata, T.; et al. Highly efficient gene silencing in mouse brain by overhanging-duplex oligonucleotides via intraventricular route. FEBS Lett. 2020, 594, 1413–1423. [Google Scholar] [CrossRef] [PubMed]
- Smith, C.C.; Aurelian, L.; Reddy, M.P.; Miller, P.S.; Ts’o, P.O. Antiviral effect of an oligo(nucleoside methylphosphonate) complementary to the splice junction of herpes simplex virus type 1 immediate early pre-mRNAs 4 and 5. Proc. Natl. Acad. Sci. USA 1986, 83, 2787–2791. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lundin, K.E.; Gissberg, O.; Smith, C.I. Oligonucleotide Therapies: The Past and the Present. Hum. Gene Ther. 2015, 26, 475–485. [Google Scholar] [CrossRef] [Green Version]
- Hammond, S.M.; Aartsma-Rus, A.; Alves, S.; Borgos, S.E.; Buijsen, R.A.M.; Collin, R.W.J.; Covello, G.; Denti, M.A.; Desviat, L.R.; Echevarria, L.; et al. Delivery of oligonucleotide-based therapeutics: Challenges and opportunities. EMBO Mol. Med. 2021, 13, e13243. [Google Scholar] [CrossRef] [PubMed]
- Scharner, J.; Figeac, N.; Ellis, J.A.; Zammit, P.S. Ameliorating pathogenesis by removing an exon containing a missense mutation: A potential exon-skipping therapy for laminopathies. Gene Ther. 2015, 22, 503–515. [Google Scholar] [CrossRef]
- Pendse, N.D.; Lamas, V.; Pawlyk, B.S.; Maeder, M.L.; Chen, Z.Y.; Pierce, E.A.; Liu, Q. In Vivo Assessment of Potential Therapeutic Approaches for USH2A-Associated Diseases. Adv. Exp. Med. Biol. 2019, 1185, 91–96. [Google Scholar] [CrossRef]
- Lim, K.H.; Han, Z.; Jeon, H.Y.; Kach, J.; Jing, E.; Weyn-Vanhentenryck, S.; Downs, M.; Corrionero, A.; Oh, R.; Scharner, J.; et al. Antisense oligonucleotide modulation of non-productive alternative splicing upregulates gene expression. Nat. Commun. 2020, 11, 3501. [Google Scholar] [CrossRef]
- Miziak, B.; Czuczwar, S. Advances in the design and discovery of novel small molecule drugs for the treatment of Dravet Syndrome. Expert Opin. Drug Discov. 2021, 16, 579–593. [Google Scholar] [CrossRef]
- Begley, D.J. Delivery of therapeutic agents to the central nervous system: The problems and the possibilities. Pharmacol. Ther. 2004, 104, 29–45. [Google Scholar] [CrossRef]
- Cecchelli, R.; Berezowski, V.; Lundquist, S.; Culot, M.; Renftel, M.; Dehouck, M.P.; Fenart, L. Modelling of the blood-brain barrier in drug discovery and development. Nat. Rev. Drug Discov. 2007, 6, 650–661. [Google Scholar] [CrossRef]
- Newton, H.B. Advances in strategies to improve drug delivery to brain tumors. Expert Rev. Neurother. 2006, 6, 1495–1509. [Google Scholar] [CrossRef]
- Zhou, J.; Atsina, K.B.; Himes, B.T.; Strohbehn, G.W.; Saltzman, W.M. Novel delivery strategies for glioblastoma. Cancer J. 2012, 18, 89–99. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zlokovic, B.V. The blood-brain barrier in health and chronic neurodegenerative disorders. Neuron 2008, 57, 178–201. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zlokovic, B.V. Neurovascular pathways to neurodegeneration in Alzheimer’s disease and other disorders. Nat. Rev. Neurosci. 2011, 12, 723–738. [Google Scholar] [CrossRef]
- Pardridge, W.M. Drug transport across the blood-brain barrier. J. Cereb. Blood Flow Metab. 2012, 32, 1959–1972. [Google Scholar] [CrossRef]
- Patel, M.; Souto, E.B.; Singh, K.K. Advances in brain drug targeting and delivery: Limitations and challenges of solid lipid nanoparticles. Expert Opin. Drug Deliv. 2013, 10, 889–905. [Google Scholar] [CrossRef] [PubMed]
- Pardridge, W.M. Molecular biology of the blood-brain barrier. Mol. Biotechnol. 2005, 30, 57–70. [Google Scholar] [CrossRef]
- Satapathy, M.K.; Yen, T.L.; Jan, J.S.; Tang, R.D.; Wang, J.Y.; Taliyan, R.; Yang, C.H. Solid Lipid Nanoparticles (SLNs): An Advanced Drug Delivery System Targeting Brain through BBB. Pharmaceutics 2021, 13, 1183. [Google Scholar] [CrossRef]
- Persidsky, Y.; Ramirez, S.H.; Haorah, J.; Kanmogne, G.D. Blood-brain barrier: Structural components and function under physiologic and pathologic conditions. J. Neuroimmune Pharmacol. 2006, 1, 223–236. [Google Scholar] [CrossRef]
- Van De Waterbeemd, H.; Smith, D.A.; Beaumont, K.; Walker, D.K. Property-based design: Optimization of drug absorption and pharmacokinetics. J. Med. Chem. 2001, 44, 1313–1333. [Google Scholar] [CrossRef]
- Chaichana, K.L.; Pinheiro, L.; Brem, H. Delivery of local therapeutics to the brain: Working toward advancing treatment for malignant gliomas. Ther. Deliv. 2015, 6, 353–369. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Patel, M.M.; Goyal, B.R.; Bhadada, S.V.; Bhatt, J.S.; Amin, A.F. Getting into the brain: Approaches to enhance brain drug delivery. CNS Drugs 2009, 23, 35–58. [Google Scholar] [CrossRef]
- Lochhead, J.J.; Thorne, R.G. Intranasal delivery of biologics to the central nervous system. Adv. Drug Deliv. Rev. 2012, 64, 614–628. [Google Scholar] [CrossRef]
- Posadas, I.; Monteagudo, S.; Cena, V. Nanoparticles for brain-specific drug and genetic material delivery, imaging and diagnosis. Nanomedicine 2016, 11, 833–849. [Google Scholar] [CrossRef] [Green Version]
- Bing, K.F.; Howles, G.P.; Qi, Y.; Palmeri, M.L.; Nightingale, K.R. Blood-brain barrier (BBB) disruption using a diagnostic ultrasound scanner and Definity in Mice. Ultrasound Med. Biol. 2009, 35, 1298–1308. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Etame, A.B.; Diaz, R.J.; O’Reilly, M.A.; Smith, C.A.; Mainprize, T.G.; Hynynen, K.; Rutka, J.T. Enhanced delivery of gold nanoparticles with therapeutic potential into the brain using MRI-guided focused ultrasound. Nanomedicine 2012, 8, 1133–1142. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Blanchette, M.; Fortin, D. Blood-brain barrier disruption in the treatment of brain tumors. Methods Mol. Biol. 2011, 686, 447–463. [Google Scholar] [CrossRef]
- Wilczewska, A.Z.; Niemirowicz, K.; Markiewicz, K.H.; Car, H. Nanoparticles as drug delivery systems. Pharmacol. Rep. 2012, 64, 1020–1037. [Google Scholar] [CrossRef]
- Sung, H.W.; Liu, Z. Advanced drug delivery systems for therapeutic applications. Adv. Healthc. Mater. 2014, 3, 1130–1132. [Google Scholar] [CrossRef]
- Safari, J.; Zarnegar, Z. Advanced drug delivery systems: Nanotechnology of health design A review. J. Saudi Chem. Soc. 2014, 18, 85–99. [Google Scholar] [CrossRef]
- Ragelle, H.; Danhier, F.; Preat, V.; Langer, R.; Anderson, D.G. Nanoparticle-based drug delivery systems: A commercial and regulatory outlook as the field matures. Expert Opin. Drug Deliv. 2017, 14, 851–864. [Google Scholar] [CrossRef] [PubMed]
- Mishra, D.K.; Shandilya, R.; Mishra, P.K. Lipid based nanocarriers: A translational perspective. Nanomedicine 2018, 14, 2023–2050. [Google Scholar] [CrossRef] [PubMed]
- Tapeinos, C.; Battaglini, M.; Ciofani, G. Advances in the design of solid lipid nanoparticles and nanostructured lipid carriers for targeting brain diseases. J. Control Release 2017, 264, 306–332. [Google Scholar] [CrossRef]
- Attama, A.A.; Momoh, M.A.; Builders, P.F. Builders. Lipid Nanoparticulate Drug Delivery Systems: A Revolution in Dosage Form Design and Development. In Recent Advances in Novel Drug Carrier Systems; Sezer, A.D., Ed.; InTech Prepress: Rijeka, Croatia, 2012. [Google Scholar]
- 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]
- 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]
- Sabnis, S.; Kumarasinghe, E.S.; Salerno, T.; Mihai, C.; Ketova, T.; Senn, J.J.; Lynn, A.; Bulychev, A.; McFadyen, I.; Chan, J.; et al. A Novel Amino Lipid Series for mRNA Delivery: Improved Endosomal Escape and Sustained Pharmacology and Safety in Non-human Primates. Mol. Ther. 2018, 26, 1509–1519. [Google Scholar] [CrossRef] [Green Version]
- Kreuter, J. Drug delivery to the central nervous system by polymeric nanoparticles: What do we know? Adv. Drug Deliv. Rev. 2014, 71, 2–14. [Google Scholar] [CrossRef]
- Patel, T.; Zhou, J.; Piepmeier, J.M.; Saltzman, W.M. Polymeric nanoparticles for drug delivery to the central nervous system. Adv. Drug Deliv. Rev. 2012, 64, 701–705. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Saraiva, C.; Praca, C.; Ferreira, R.; Santos, T.; Ferreira, L.; Bernardino, L. Nanoparticle-mediated brain drug delivery: Overcoming blood-brain barrier to treat neurodegenerative diseases. J. Control Release 2016, 235, 34–47. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wohlfart, S.; Gelperina, S.; Kreuter, J. Transport of drugs across the blood-brain barrier by nanoparticles. J. Control Release 2012, 161, 264–273. [Google Scholar] [CrossRef] [PubMed]
- Buse, J.; El-Aneed, A. Properties, engineering and applications of lipid-based nanoparticle drug-delivery systems: Current research and advances. Nanomedicine 2010, 5, 1237–1260. [Google Scholar] [CrossRef] [PubMed]
- Khan, I.; Saeed, K.; Khan, I. Nanoparticles: Properties, applications and toxicities. Arab. J. Chem. 2019, 12, 908–931. [Google Scholar] [CrossRef]
- Shvedova, A.; Pietroiusti, A.; Kagan, V. Nanotoxicology ten years later: Lights and shadows. Toxicol. Appl. Pharmacol. 2016, 299, 1–2. [Google Scholar] [CrossRef] [Green Version]
- Jumaa, M.; Muller, B.W. Lipid emulsions as a novel system to reduce the hemolytic activity of lytic agents: Mechanism of the protective effect. Eur. J. Pharm. Sci. 2000, 9, 285–290. [Google Scholar] [CrossRef]
- Kaur, I.P.; Bhandari, R.; Bhandari, S.; Kakkar, V. Potential of solid lipid nanoparticles in brain targeting. J. Control Release 2008, 127, 97–109. [Google Scholar] [CrossRef]
- Pardeshi, C.; Rajput, P.; Belgamwar, V.; Tekade, A.; Patil, G.; Chaudhary, K.; Sonje, A. Solid lipid based nanocarriers: An overview. Acta Pharm. 2012, 62, 433–472. [Google Scholar] [CrossRef]
- Muller, R.H.; Mader, K.; Gohla, S. Solid lipid nanoparticles (SLN) for controlled drug delivery—A review of the state of the art. Eur. J. Pharm. Biopharm. 2000, 50, 161–177. [Google Scholar] [CrossRef]
- Mukherjee, S.; Ray, S.; Thakur, R.S. Solid lipid nanoparticles: A modern formulation approach in drug delivery system. Indian J. Pharm. Sci. 2009, 71, 349–358. [Google Scholar] [CrossRef] [Green Version]
- Tabatt, K.; Kneuer, C.; Sameti, M.; Olbrich, C.; Muller, R.H.; Lehr, C.M.; Bakowsky, U. Transfection with different colloidal systems: Comparison of solid lipid nanoparticles and liposomes. J. Control Release 2004, 97, 321–332. [Google Scholar] [CrossRef]
- Hamdani, J.; Moes, A.J.; Amighi, K. Physical and thermal characterisation of Precirol and Compritol as lipophilic glycerides used for the preparation of controlled-release matrix pellets. Int. J. Pharm. 2003, 260, 47–57. [Google Scholar] [CrossRef]
- Mosallaei, N.; Jaafari, M.R.; Hanafi-Bojd, M.Y.; Golmohammadzadeh, S.; Malaekeh-Nikouei, B. Docetaxel-loaded solid lipid nanoparticles: Preparation, characterization, in vitro, and in vivo evaluations. J. Pharm. Sci. 2013, 102, 1994–2004. [Google Scholar] [CrossRef] [PubMed]
- Yang, S.C.; Lu, L.F.; Cai, Y.; Zhu, J.B.; Liang, B.W.; Yang, C.Z. Body distribution in mice of intravenously injected camptothecin solid lipid nanoparticles and targeting effect on brain. J. Control Release 1999, 59, 299–307. [Google Scholar] [CrossRef]
- Johnstone, R.M.; Adam, M.; Hammond, J.R.; Orr, L.; Turbide, C. Vesicle formation during reticulocyte maturation. Association of plasma membrane activities with released vesicles (exosomes). J. Biol. Chem. 1987, 262, 9412–9420. [Google Scholar] [CrossRef]
- Abels, E.R.; Breakefield, X.O. Introduction to Extracellular Vesicles: Biogenesis, RNA Cargo Selection, Content, Release, and Uptake. Cell Mol. Neurobiol. 2016, 36, 301–312. [Google Scholar] [CrossRef] [PubMed]
- Xu, R.; Greening, D.W.; Zhu, H.J.; Takahashi, N.; Simpson, R.J. Extracellular vesicle isolation and characterization: Toward clinical application. J. Clin. Investig. 2016, 126, 1152–1162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Akers, J.C.; Gonda, D.; Kim, R.; Carter, B.S.; Chen, C.C. Biogenesis of extracellular vesicles (EV): Exosomes, microvesicles, retrovirus-like vesicles, and apoptotic bodies. J. Neuro-Oncol. 2013, 113, 1–11. [Google Scholar] [CrossRef] [Green Version]
- Lynch, C.; Panagopoulou, M.; Gregory, C.D. Extracellular Vesicles Arising from Apoptotic Cells in Tumors: Roles in Cancer Pathogenesis and Potential Clinical Applications. Front. Immunol. 2017, 8, 1174. [Google Scholar] [CrossRef] [Green Version]
- Chakrabortty, S.K.; Prakash, A.; Nechooshtan, G.; Hearn, S.; Gingeras, T.R. Extracellular vesicle-mediated transfer of processed and functional RNY5 RNA. RNA 2015, 21, 1966–1979. [Google Scholar] [CrossRef] [Green Version]
- Ridder, K.; Sevko, A.; Heide, J.; Dams, M.; Rupp, A.K.; Macas, J.; Starmann, J.; Tjwa, M.; Plate, K.H.; Sultmann, H.; et al. Extracellular vesicle-mediated transfer of functional RNA in the tumor microenvironment. Oncoimmunology 2015, 4, e1008371. [Google Scholar] [CrossRef] [Green Version]
- Waldenstrom, A.; Genneback, N.; Hellman, U.; Ronquist, G. Cardiomyocyte microvesicles contain DNA/RNA and convey biological messages to target cells. PLoS ONE 2012, 7, e34653. [Google Scholar] [CrossRef] [Green Version]
- Zheng, P.; Luo, Q.; Wang, W.; Li, J.; Wang, T.; Wang, P.; Chen, L.; Zhang, P.; Chen, H.; Liu, Y.; et al. Tumor-associated macrophages-derived exosomes promote the migration of gastric cancer cells by transfer of functional Apolipoprotein E. Cell Death Dis. 2018, 9, 434. [Google Scholar] [CrossRef] [PubMed]
- Riazifar, M.; Pone, E.J.; Lotvall, J.; Zhao, W. Stem Cell Extracellular Vesicles: Extended Messages of Regeneration. Annu. Rev. Pharmacol. Toxicol. 2017, 57, 125–154. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Valadi, H.; Ekstrom, K.; Bossios, A.; Sjostrand, M.; Lee, J.J.; Lotvall, 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] [PubMed] [Green Version]
- Cheng, Y.; Zeng, Q.; Han, Q.; Xia, W. Effect of pH, temperature and freezing-thawing on quantity changes and cellular uptake of exosomes. Protein Cell 2019, 10, 295–299. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jayachandran, M.; Miller, V.M.; Heit, J.A.; Owen, W.G. Methodology for isolation, identification and characterization of microvesicles in peripheral blood. J. Immunol. Methods 2012, 375, 207–214. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sokolova, V.; Ludwig, A.K.; Hornung, S.; Rotan, O.; Horn, P.A.; Epple, M.; Giebel, B. Characterisation of exosomes derived from human cells by nanoparticle tracking analysis and scanning electron microscopy. Colloids Surf. B Biointerfaces 2011, 87, 146–150. [Google Scholar] [CrossRef]
- Sun, D.; Zhuang, X.; Xiang, X.; Liu, Y.; Zhang, S.; Liu, C.; Barnes, S.; Grizzle, W.; Miller, D.; Zhang, H.G. A novel nanoparticle drug delivery system: The anti-inflammatory activity of curcumin is enhanced when encapsulated in exosomes. Mol. Ther. 2010, 18, 1606–1614. [Google Scholar] [CrossRef]
- Marcus, M.E.; Leonard, J.N. FedExosomes: Engineering Therapeutic Biological Nanoparticles that Truly Deliver. Pharmaceuticals 2013, 6, 659–680. [Google Scholar] [CrossRef] [Green Version]
- Sanghani, A.; Andriesei, P.; Kafetzis, K.N.; Tagalakis, A.D.; Yu-Wai-Man, C. Advances in exosome therapies in ophthalmology-From bench to clinical trial. Acta Ophthalmol. 2021. [Google Scholar] [CrossRef] [PubMed]
- Sil, S.; Dagur, R.S.; Liao, K.; Peeples, E.S.; Hu, G.; Periyasamy, P.; Buch, S. Strategies for the use of Extracellular Vesicles for the Delivery of Therapeutics. J. Neuroimmune Pharmacol. 2020, 15, 422–442. [Google Scholar] [CrossRef]
- Spellicy, S.E.; Stice, S.L. Tissue and Stem Cell Sourced Extracellular Vesicle Communications with Microglia. Stem Cell Rev. Rep. 2021, 17, 357–368. [Google Scholar] [CrossRef] [PubMed]
- Kim, S.M.; Kim, H.S. Engineering of extracellular vesicles as drug delivery vehicles. Stem Cell Investig. 2017, 4, 74. [Google Scholar] [CrossRef] [Green Version]
- Yang, T.; Martin, P.; Fogarty, B.; Brown, A.; Schurman, K.; Phipps, R.; Yin, V.P.; Lockman, P.; Bai, S. Exosome delivered anticancer drugs across the blood-brain barrier for brain cancer therapy in Danio rerio. Pharm. Res. 2015, 32, 2003–2014. [Google Scholar] [CrossRef] [PubMed]
- Escrevente, C.; Keller, S.; Altevogt, P.; Costa, J. Interaction and uptake of exosomes by ovarian cancer cells. BMC Cancer 2011, 11, 108. [Google Scholar] [CrossRef] [Green Version]
- Montecalvo, A.; Larregina, A.T.; Shufesky, W.J.; Stolz, D.B.; Sullivan, M.L.; Karlsson, J.M.; Baty, C.J.; Gibson, G.A.; Erdos, G.; Wang, Z.; et al. Mechanism of transfer of functional microRNAs between mouse dendritic cells via exosomes. Blood 2012, 119, 756–766. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Morelli, A.E.; Larregina, A.T.; Shufesky, W.J.; Sullivan, M.L.; Stolz, D.B.; Papworth, G.D.; Zahorchak, A.F.; Logar, A.J.; Wang, Z.; Watkins, S.C.; et al. Endocytosis, intracellular sorting, and processing of exosomes by dendritic cells. Blood 2004, 104, 3257–3266. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Aiuti, A.; Roncarolo, M.G.; Naldini, L. Gene therapy for ADA-SCID, the first marketing approval of an ex vivo gene therapy in Europe: Paving the road for the next generation of advanced therapy medicinal products. EMBO Mol. Med. 2017, 9, 737–740. [Google Scholar] [CrossRef] [PubMed]
- Porter, D.L.; Levine, B.L.; Kalos, M.; Bagg, A.; June, C.H. Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N. Engl. J. Med. 2011, 365, 725–733. [Google Scholar] [CrossRef] [Green Version]
- Andtbacka, R.H.; Kaufman, H.L.; Collichio, F.; Amatruda, T.; Senzer, N.; Chesney, J.; Delman, K.A.; Spitler, L.E.; Puzanov, I.; Agarwala, S.S.; et al. Talimogene Laherparepvec Improves Durable Response Rate in Patients With Advanced Melanoma. J. Clin. Oncol. 2015, 33, 2780–2788. [Google Scholar] [CrossRef]
- Maguire, A.M.; Simonelli, F.; Pierce, E.A.; Pugh, E.N., Jr.; Mingozzi, F.; Bennicelli, J.; Banfi, S.; Marshall, K.A.; Testa, F.; Surace, E.M.; et al. Safety and efficacy of gene transfer for Leber’s congenital amaurosis. N. Engl. J. Med. 2008, 358, 2240–2248. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nathwani, A.C.; Tuddenham, E.G.; Rangarajan, S.; Rosales, C.; McIntosh, J.; Linch, D.C.; Chowdary, P.; Riddell, A.; Pie, A.J.; Harrington, C.; et al. Adenovirus-associated virus vector-mediated gene transfer in hemophilia B. N. Engl. J. Med. 2011, 365, 2357–2365. [Google Scholar] [CrossRef]
- Hawkins, P.N.; Ando, Y.; Dispenzeri, A.; Gonzalez-Duarte, A.; Adams, D.; Suhr, O.B. Evolving landscape in the management of transthyretin amyloidosis. Ann. Med. 2015, 47, 625–638. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ruberg, F.L.; Berk, J.L. Transthyretin (TTR) cardiac amyloidosis. Circulation 2012, 126, 1286–1300. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Conceicao, I.; Gonzalez-Duarte, A.; Obici, L.; Schmidt, H.H.; Simoneau, D.; Ong, M.L.; Amass, L. “Red-flag” symptom clusters in transthyretin familial amyloid polyneuropathy. J. Peripher. Nerv. Syst. 2016, 21, 5–9. [Google Scholar] [CrossRef] [PubMed]
- Ando, Y.; Coelho, T.; Berk, J.L.; Cruz, M.W.; Ericzon, B.G.; Ikeda, S.; Lewis, W.D.; Obici, L.; Plante-Bordeneuve, V.; Rapezzi, C.; et al. Guideline of transthyretin-related hereditary amyloidosis for clinicians. Orphanet J. Rare Dis. 2013, 8, 31. [Google Scholar] [CrossRef] [Green Version]
- Chao, H.C.; Liao, Y.C.; Liu, Y.T.; Guo, Y.C.; Chang, F.P.; Lee, Y.C.; Lin, K.P. Clinical and genetic profiles of hereditary transthyretin amyloidosis in Taiwan. Ann. Clin. Transl. Neurol. 2019, 6, 913–922. [Google Scholar] [CrossRef]
- Yang, N.C.; Lee, M.J.; Chao, C.C.; Chuang, Y.T.; Lin, W.M.; Chang, M.F.; Hsieh, P.C.; Kan, H.W.; Lin, Y.H.; Yang, C.C.; et al. Clinical presentations and skin denervation in amyloid neuropathy due to transthyretin Ala97Ser. Neurology 2010, 75, 532–538. [Google Scholar] [CrossRef]
- Dungu, J.N.; Anderson, L.J.; Whelan, C.J.; Hawkins, P.N. Cardiac transthyretin amyloidosis. Heart 2012, 98, 1546–1554. [Google Scholar] [CrossRef]
- Lai, H.J.; Huang, K.C.; Liang, Y.C.; Chien, K.L.; Lee, M.J.; Hsieh, S.T.; Chao, C.C.; Yang, C.C. Cardiac manifestations and prognostic implications of hereditary transthyretin amyloidosis associated with transthyretin Ala97Ser. J. Formos. Med. Assoc. 2020, 119, 693–700. [Google Scholar] [CrossRef]
- Su, Y.; Jono, H.; Misumi, Y.; Senokuchi, T.; Guo, J.; Ueda, M.; Shinriki, S.; Tasaki, M.; Shono, M.; Obayashi, K.; et al. Novel function of transthyretin in pancreatic alpha cells. FEBS Lett. 2012, 586, 4215–4222. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Coelho, T.; Adams, D.; Silva, A.; Lozeron, P.; Hawkins, P.N.; Mant, T.; Perez, J.; Chiesa, J.; Warrington, S.; Tranter, E.; et al. Safety and efficacy of RNAi therapy for transthyretin amyloidosis. N. Engl. J. Med. 2013, 369, 819–829. [Google Scholar] [CrossRef] [PubMed]
- Adams, D.; Polydefkis, M.; Gonzalez-Duarte, A.; Wixner, J.; Kristen, A.V.; Schmidt, H.H.; Berk, J.L.; Losada Lopez, I.A.; Dispenzieri, A.; Quan, D.; et al. Long-term safety and efficacy of patisiran for hereditary transthyretin-mediated amyloidosis with polyneuropathy: 12-month results of an open-label extension study. Lancet Neurol. 2021, 20, 49–59. [Google Scholar] [CrossRef]
- Adams, D.; Suhr, O.B.; Dyck, P.J.; Litchy, W.J.; Leahy, R.G.; Chen, J.; Gollob, J.; Coelho, T. Trial design and rationale for APOLLO, a Phase 3, placebo-controlled study of patisiran in patients with hereditary ATTR amyloidosis with polyneuropathy. BMC Neurol. 2017, 17, 181. [Google Scholar] [CrossRef] [PubMed]
- Dohrn, M.F.; Ihne, S.; Hegenbart, U.; Medina, J.; Zuchner, S.L.; Coelho, T.; Hahn, K. Targeting transthyretin—Mechanism-based treatment approaches and future perspectives in hereditary amyloidosis. J. Neurochem. 2021, 156, 802–818. [Google Scholar] [CrossRef]
- Gillmore, J.D.; Gane, E.; Taubel, J.; Kao, J.; Fontana, M.; Maitland, M.L.; Seitzer, J.; O’Connell, D.; Walsh, K.R.; Wood, K.; et al. CRISPR-Cas9 In Vivo Gene Editing for Transthyretin Amyloidosis. N. Engl. J. Med. 2021, 385, 493–502. [Google Scholar] [CrossRef]
- Anderson, K.E.; Bloomer, J.R.; Bonkovsky, H.L.; Kushner, J.P.; Pierach, C.A.; Pimstone, N.R.; Desnick, R.J. Recommendations for the diagnosis and treatment of the acute porphyrias. Ann. Intern. Med. 2005, 142, 439–450. [Google Scholar] [CrossRef]
- Fratz, E.J.; Stojanovski, B.M.; Ferreira, G.C. Toward Heme: 5-Aminolevulinate Synthase and Initiation of Porphyrin Synthesis; World Scientific Publishing Co. Pte. Ltd.: Toh Tuck Link, Singapore, 2014; Volume 26, p. 596224. [Google Scholar]
- Puy, H.; Gouya, L.; Deybach, J.C. Porphyrias. Lancet 2010, 375, 924–937. [Google Scholar] [CrossRef]
- Marsden, J.T.; Guppy, S.; Stein, P.; Cox, T.M.; Badminton, M.; Gardiner, T.; Barth, J.H.; Stewart, M.F.; Rees, D.C. Audit of the Use of Regular Haem Arginate Infusions in Patients with Acute Porphyria to Prevent Recurrent Symptoms. JIMD Rep. 2015, 22, 57–65. [Google Scholar] [CrossRef] [Green Version]
- Meyer, U.A.; Schuurmans, M.M.; Lindberg, R.L. Acute porphyrias: Pathogenesis of neurological manifestations. Semin. Liver Dis. 1998, 18, 43–52. [Google Scholar] [CrossRef]
- Willandt, B.; Langendonk, J.G.; Biermann, K.; Meersseman, W.; D’Heygere, F.; George, C.; Verslype, C.; Monbaliu, D.; Cassiman, D. Liver Fibrosis Associated with Iron Accumulation Due to Long-Term Heme-Arginate Treatment in Acute Intermittent Porphyria: A Case Series. JIMD Rep. 2016, 25, 77–81. [Google Scholar] [CrossRef] [Green Version]
- Yasuda, M.; Gan, L.; Chen, B.; Kadirvel, S.; Yu, C.; Phillips, J.D.; New, M.I.; Liebow, A.; Fitzgerald, K.; Querbes, W.; et al. RNAi-mediated silencing of hepatic Alas1 effectively prevents and treats the induced acute attacks in acute intermittent porphyria mice. Proc. Natl. Acad. Sci. USA 2014, 111, 7777–7782. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Balwani, M.; Sardh, E.; Ventura, P.; Peiro, P.A.; Rees, D.C.; Stolzel, U.; Bissell, D.M.; Bonkovsky, H.L.; Windyga, J.; Anderson, K.E.; et al. Phase 3 Trial of RNAi Therapeutic Givosiran for Acute Intermittent Porphyria. N. Engl. J. Med. 2020, 382, 2289–2301. [Google Scholar] [CrossRef]
- de Paula Brandao, P.R.; Titze-de-Almeida, S.S.; Titze-de-Almeida, R. Leading RNA Interference Therapeutics Part 2: Silencing Delta-Aminolevulinic Acid Synthase 1, with a Focus on Givosiran. Mol. Diagn. Ther. 2020, 24, 61–68. [Google Scholar] [CrossRef] [PubMed]
- Syed, Y.Y. Givosiran: A Review in Acute Hepatic Porphyria. Drugs 2021, 81, 841–848. [Google Scholar] [CrossRef] [PubMed]
- Du, L.; Pollard, J.M.; Gatti, R.A. Correction of prototypic ATM splicing mutations and aberrant ATM function with antisense morpholino oligonucleotides. Proc. Natl. Acad. Sci. USA 2007, 104, 6007–6012. [Google Scholar] [CrossRef] [Green Version]
- Aartsma-Rus, A.; Krieg, A.M. FDA Approves Eteplirsen for Duchenne Muscular Dystrophy: The Next Chapter in the Eteplirsen Saga. Nucleic Acid Ther. 2017, 27, 1–3. [Google Scholar] [CrossRef] [Green Version]
- Jarver, P.; Zaghloul, E.M.; Arzumanov, A.A.; Saleh, A.F.; McClorey, G.; Hammond, S.M.; Hallbrink, M.; Langel, U.; Smith, C.I.; Wood, M.J.; et al. Peptide nanoparticle delivery of charge-neutral splice-switching morpholino oligonucleotides. Nucleic Acid Ther. 2015, 25, 65–77. [Google Scholar] [CrossRef]
- Lunn, M.R.; Wang, C.H. Spinal muscular atrophy. Lancet 2008, 371, 2120–2133. [Google Scholar] [CrossRef]
- Kolb, S.J.; Kissel, J.T. Spinal muscular atrophy: A timely review. Arch. Neurol. 2011, 68, 979–984. [Google Scholar] [CrossRef] [Green Version]
- Eggermann, K.; Glaser, D.; Abicht, A.; Wirth, B. Spinal muscular atrophy (5qSMA): Best practice of diagnostics, newborn screening and therapy. Med. Gen. 2020, 32, 263–272. [Google Scholar] [CrossRef]
- Tajik-Ahmadabad, B.; Polyzos, A.; Separovic, F.; Shabanpoor, F. Amphiphilic lipopeptide significantly enhances uptake of charge-neutral splice switching morpholino oligonucleotide in spinal muscular atrophy patient-derived fibroblasts. Int. J. Pharm. 2017, 532, 21–28. [Google Scholar] [CrossRef]
- Mercuri, E.; Finkel, R.S.; Muntoni, F.; Wirth, B.; Montes, J.; Main, M.; Mazzone, E.S.; Vitale, M.; Snyder, B.; Quijano-Roy, S.; et al. Diagnosis and management of spinal muscular atrophy: Part 1: Recommendations for diagnosis, rehabilitation, orthopedic and nutritional care. Neuromuscul. Disord. 2018, 28, 103–115. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Day, J.W.; Finkel, R.S.; Chiriboga, C.A.; Connolly, A.M.; Crawford, T.O.; Darras, B.T.; Iannaccone, S.T.; Kuntz, N.L.; Pena, 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]
- Day, J.W.; Mendell, J.R.; Mercuri, E.; Finkel, R.S.; Strauss, K.A.; Kleyn, A.; Tauscher-Wisniewski, S.; Tukov, F.F.; Reyna, S.P.; Chand, D.H. Clinical Trial and Postmarketing Safety of Onasemnogene Abeparvovec Therapy. Drug Saf. 2021, 44, 1109–1119. [Google Scholar] [CrossRef]
- Bischof, M.; Lorenzi, M.; Lee, J.; Druyts, E.; Balijepalli, C.; Dabbous, O. Matching-adjusted indirect treatment comparison of onasemnogene abeparvovec and nusinersen for the treatment of symptomatic patients with spinal muscular atrophy type 1. Curr. Med. Res. Opin. 2021, 37, 1719–1730. [Google Scholar] [CrossRef] [PubMed]
- Emery, A.E. The muscular dystrophies. Lancet 2002, 359, 687–695. [Google Scholar] [CrossRef]
- Kingston, H.M.; Harper, P.S.; Pearson, P.L.; Davies, K.E.; Williamson, R.; Page, D. Localization of Gene for Becker Muscular-Dystrophy. Lancet 1983, 2, 1200. [Google Scholar] [CrossRef]
- Douglas, A.G.; Wood, M.J. Splicing therapy for neuromuscular disease. Mol. Cell Neurosci. 2013, 56, 169–185. [Google Scholar] [CrossRef]
- Buvoli, M.; Buvoli, A.; Leinwand, L.A. Interplay between exonic splicing enhancers, mRNA processing, and mRNA surveillance in the dystrophic Mdx mouse. PLoS ONE 2007, 2, e427. [Google Scholar] [CrossRef] [Green Version]
- Bello, L.; Morgenroth, L.P.; Gordish-Dressman, H.; Hoffman, E.P.; McDonald, C.M.; Cirak, S.; CINRG Investigators. DMD genotypes and loss of ambulation in the CINRG Duchenne Natural History Study. Neurology 2016, 87, 401–409. [Google Scholar] [CrossRef] [Green Version]
- Brogna, C.; Coratti, G.; Pane, M.; Ricotti, V.; Messina, S.; D’Amico, A.; Bruno, C.; Vita, G.; Berardinelli, A.; Mazzone, E.; et al. Long-term natural history data in Duchenne muscular dystrophy ambulant patients with mutations amenable to skip exons 44, 45, 51 and 53. PLoS ONE 2019, 14, e0218683. [Google Scholar] [CrossRef] [Green Version]
- Ricotti, V.; Mandy, W.P.; Scoto, M.; Pane, M.; Deconinck, N.; Messina, S.; Mercuri, E.; Skuse, D.H.; Muntoni, F. Neurodevelopmental, emotional, and behavioural problems in Duchenne muscular dystrophy in relation to underlying dystrophin gene mutations. Dev. Med. Child Neurol. 2016, 58, 77–84. [Google Scholar] [CrossRef]
- Bello, L.; D’Angelo, G.; Villa, M.; Fusto, A.; Vianello, S.; Merlo, B.; Sabbatini, D.; Barp, A.; Gandossini, S.; Magri, F.; et al. Genetic modifiers of respiratory function in Duchenne muscular dystrophy. Ann. Clin. Transl. Neurol. 2020, 7, 786–798. [Google Scholar] [CrossRef]
- Aartsma-Rus, A.; van Ommen, G.J. Antisense-mediated exon skipping: A versatile tool with therapeutic and research applications. RNA 2007, 13, 1609–1624. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- McDonald, C.M.; Shieh, P.B.; Abdel-Hamid, H.Z.; Connolly, A.M.; Ciafaloni, E.; Wagner, K.R.; Goemans, N.; Mercuri, E.; Khan, N.; Koenig, E.; et al. Open-Label Evaluation of Eteplirsen in Patients with Duchenne Muscular Dystrophy Amenable to Exon 51 Skipping: PROMOVI Trial. J. Neuromuscul. Dis. 2021, 8, 989–1001. [Google Scholar] [CrossRef] [PubMed]
- Golodirsen (Vyondys 53) for Duchenne muscular dystrophy. Med. Lett. Drugs Ther. 2020, 62, 119–120.
- Anwar, S.; Yokota, T. Golodirsen for Duchenne muscular dystrophy. Drugs Today 2020, 56, 491–504. [Google Scholar] [CrossRef] [PubMed]
- Schwartz, J.L.; Jones, K.L.; Yeo, G.W. Repeat RNA expansion disorders of the nervous system: Post-transcriptional mechanisms and therapeutic strategies. Crit. Rev. Biochem. Mol. Biol. 2021, 56, 31–53. [Google Scholar] [CrossRef]
- Rohilla, K.J.; Gagnon, K.T. RNA biology of disease-associated microsatellite repeat expansions. Acta Neuropathol. Commun. 2017, 5, 63. [Google Scholar] [CrossRef] [Green Version]
- Ross, C.A.; Aylward, E.H.; Wild, E.J.; Langbehn, D.R.; Long, J.D.; Warner, J.H.; Scahill, R.I.; Leavitt, B.R.; Stout, J.C.; Paulsen, J.S.; et al. Huntington disease: Natural history, biomarkers and prospects for therapeutics. Nat. Rev. Neurol. 2014, 10, 204–216. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Carroll, J.B.; Warby, S.C.; Southwell, A.L.; Doty, C.N.; Greenlee, S.; Skotte, N.; Hung, G.; Bennett, C.F.; Freier, S.M.; Hayden, M.R. Potent and selective antisense oligonucleotides targeting single-nucleotide polymorphisms in the Huntington disease gene / allele-specific silencing of mutant huntingtin. Mol. Ther. 2011, 19, 2178–2185. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Skotte, N.H.; Southwell, A.L.; Ostergaard, M.E.; Carroll, J.B.; Warby, S.C.; Doty, C.N.; Petoukhov, E.; Vaid, K.; Kordasiewicz, H.; Watt, A.T.; et al. Allele-specific suppression of mutant huntingtin using antisense oligonucleotides: Providing a therapeutic option for all Huntington disease patients. PLoS ONE 2014, 9, e107434. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kay, C.; Collins, J.A.; Skotte, N.H.; Southwell, A.L.; Warby, S.C.; Caron, N.S.; Doty, C.N.; Nguyen, B.; Griguoli, A.; Ross, C.J.; et al. Huntingtin Haplotypes Provide Prioritized Target Panels for Allele-specific Silencing in Huntington Disease Patients of European Ancestry. Mol. Ther. 2015, 23, 1759–1771. [Google Scholar] [CrossRef] [PubMed]
- Leavitt, B.R.; Kordasiewicz, H.B.; Schobel, S.A. Huntingtin-Lowering Therapies for Huntington Disease: A Review of the Evidence of Potential Benefits and Risks. JAMA Neurol. 2020, 77, 764–772. [Google Scholar] [CrossRef]
- Tabrizi, S.; Leavitt, B.; Kordasiewicz, H.; Czech, C.; Swayze, E.; Norris, D.A.; Baumann, T.; Gerlach, I.; Schobel, S.; Smith, A.; et al. Effects of IONIS-HTTRx in Patients with Early Huntington’s Disease, Results of the First HTT-Lowering Drug Trial. Neurology 2018, 90, A97–A98. [Google Scholar] [CrossRef]
- Boudreau, R.L.; McBride, J.L.; Martins, I.; Shen, S.; Xing, Y.; Carter, B.J.; Davidson, B.L. Nonallele-specific silencing of mutant and wild-type huntingtin demonstrates therapeutic efficacy in Huntington’s disease mice. Mol. Ther. 2009, 17, 1053–1063. [Google Scholar] [CrossRef]
- Pfister, E.L.; DiNardo, N.; Mondo, E.; Borel, F.; Conroy, F.; Fraser, C.; Gernoux, G.; Han, X.; Hu, D.; Johnson, E.; et al. Artificial miRNAs Reduce Human Mutant Huntingtin Throughout the Striatum in a Transgenic Sheep Model of Huntington’s Disease. Hum. Gene Ther. 2018, 29, 663–673. [Google Scholar] [CrossRef]
- Stanek, L.M.; Sardi, S.P.; Mastis, B.; Richards, A.R.; Treleaven, C.M.; Taksir, T.; Misra, K.; Cheng, S.H.; Shihabuddin, L.S. Silencing mutant huntingtin by adeno-associated virus-mediated RNA interference ameliorates disease manifestations in the YAC128 mouse model of Huntington’s disease. Hum. Gene Ther. 2014, 25, 461–474. [Google Scholar] [CrossRef] [Green Version]
- Wraith, J.E. The clinical presentation of lysosomal storage disorders. Acta Neurol. Taiwan 2004, 13, 101–106. [Google Scholar]
- Kelly, J.M.; Bradbury, A.; Martin, D.R.; Byrne, M.E. Emerging therapies for neuropathic lysosomal storage disorders. Prog. Neurobiol. 2017, 152, 166–180. [Google Scholar] [CrossRef]
- Dhillon, S. Avalglucosidase alfa: First Approval. Drugs 2021, 81, 1803–1809. [Google Scholar] [CrossRef] [PubMed]
- Kishnani, P.S.; Beckemeyer, A.A. New therapeutic approaches for Pompe disease: Enzyme replacement therapy and beyond. Pediatr Endocrinol. Rev. 2014, 12 (Suppl. 1), 114–124. [Google Scholar]
- Lenders, M.; Brand, E. Precision medicine in Fabry disease. Nephrol. Dial. Transpl. 2021, 36, 14–23. [Google Scholar] [CrossRef]
- Shemesh, E.; Deroma, L.; Bembi, B.; Deegan, P.; Hollak, C.; Weinreb, N.J.; Cox, T.M. Enzyme replacement and substrate reduction therapy for Gaucher disease. Cochrane Database Syst. Rev. 2015. [Google Scholar] [CrossRef] [PubMed]
- Diaz-Font, A.; Chabas, A.; Grinberg, D.; Vilageliu, L. RNAi-mediated inhibition of the glucosylceramide synthase (GCS) gene: A preliminary study towards a therapeutic strategy for Gaucher disease and other glycosphingolipid storage diseases. Blood Cells Mol. Dis. 2006, 37, 197–203. [Google Scholar] [CrossRef]
- Abasolo, I.; Seras-Franzoso, J.; Molto-Abad, M.; Diaz-Riascos, V.; Corchero, J.L.; Pintos-Morell, G.; Schwartz, S., Jr. Nanotechnology-based approaches for treating lysosomal storage disorders, a focus on Fabry disease. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 2021, 13, e1684. [Google Scholar] [CrossRef]
- Pawelczak, K.S.; Gavande, N.S.; VanderVere-Carozza, P.S.; Turchi, J.J. Modulating DNA Repair Pathways to Improve Precision Genome Engineering. ACS Chem. Biol. 2018, 13, 389–396. [Google Scholar] [CrossRef] [PubMed]
- Coutinho, M.F.; Santos, J.I.; Mendonça, L.S.; Matos, L.; Prata, M.J.; Jurado, A.S.; De Lima, M.C.P.; Alves, S. Lysosomal Storage Disease-Associated Neuropathy: Targeting Stable Nucleic Acid Lipid Particle (SNALP)-Formulated siRNAs to the Brain as a Therapeutic Approach. Int. J. Mol. Sci. 2020, 21, 5732. [Google Scholar] [CrossRef]
- Kariko, K. In vitro-Transcribed mRNA Therapeutics: Out of the Shadows and Into the Spotlight. Mol. Ther. 2019, 27, 691–692. [Google Scholar] [CrossRef] [Green Version]
- Winkle, M.; El-Daly, S.M.; Fabbri, M.; Calin, G.A. Noncoding RNA therapeutics—Challenges and potential solutions. Nat. Rev. Drug Discov. 2021, 20, 629–651. [Google Scholar] [CrossRef] [PubMed]
- Damase, T.R.; Sukhovershin, R.; Boada, C.; Taraballi, F.; Pettigrew, R.I.; Cooke, J.P. The Limitless Future of RNA Therapeutics. Front. Bioeng. Biotech. 2021, 9. [Google Scholar] [CrossRef] [PubMed]
Nanocarrier Type | Strengths | Limitations |
---|---|---|
Lipid nanoparticles |
|
|
Liposomes |
|
|
Nanoemulsions |
|
|
Drug (Approved Year) | Target Disease | Target Molecule | Mode of Action | Category of RNA Therapy |
---|---|---|---|---|
Nusinersen (2016) | Spinal muscular atrophy | SMN2 mRNA | Modifying alternative splicing of the SMN2 mRNA to increase the SMN protein level | Single-strand antisense |
Eteplirsen (2016) | Duchene muscular dystrophy | Dystrophy mRNA | Induce the exon 51 skipping during the splicing process to produce a functional dystrophin protein | Single-strand antisense |
Inotersen (2018) | Hereditary transthyretin amyloidosis | Transthyretin mRNA | Complementary binding to the Transthyretin mRNA to induce RNase H-based degradation | Single-strand antisense |
Golodirsen (2019) | Duchene muscular dystrophy | Dystrophy mRNA | Inducing the skipping of exon 53 during the splicing process to produce a functional dystrophin protein | Single-strand antisense |
Patisiran (2018) | Hereditary transthyretin amyloidosis | Transthyretin mRNA | RNA interference to inhibit the production of transthyretin protein | Double-strand small interfering |
Givosiran (2019) | Acute hepatic porphyria | ALAS1 mRNA | RNA interference to suppress the hepatic production of ALAS1 enzyme | Double-strand small interfering |
Pegaptanib (2004) | Age-related macular degeneration | VEGF protein | Specific binding to the 165 isoforms of VEGF to inhibit its function | RNA aptamer |
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
Lee, M.-J.; Lee, I.; Wang, K. Recent Advances in RNA Therapy and Its Carriers to Treat the Single-Gene Neurological Disorders. Biomedicines 2022, 10, 158. https://doi.org/10.3390/biomedicines10010158
Lee M-J, Lee I, Wang K. Recent Advances in RNA Therapy and Its Carriers to Treat the Single-Gene Neurological Disorders. Biomedicines. 2022; 10(1):158. https://doi.org/10.3390/biomedicines10010158
Chicago/Turabian StyleLee, Ming-Jen, Inyoul Lee, and Kai Wang. 2022. "Recent Advances in RNA Therapy and Its Carriers to Treat the Single-Gene Neurological Disorders" Biomedicines 10, no. 1: 158. https://doi.org/10.3390/biomedicines10010158
APA StyleLee, M. -J., Lee, I., & Wang, K. (2022). Recent Advances in RNA Therapy and Its Carriers to Treat the Single-Gene Neurological Disorders. Biomedicines, 10(1), 158. https://doi.org/10.3390/biomedicines10010158