Restoring Dystrophin Expression in Duchenne Muscular Dystrophy: Current Status of Therapeutic Approaches
Abstract
:1. Introduction
2. Read-Through Therapy
3. Exon Skipping
4. Vector-Mediated Gene Therapy
5. Cell Therapy
6. Challenges and Limitations in Therapies to Restore Dystrophin
7. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Ervasti, J.M.; Ohlendieck, K.; Kahl, S.D.; Gaver, M.G.; Campbell, K.P. Deficiency of a glycoprotein component of the dystrophin complex in dystrophic muscle. Nature 1990, 345, 315–319. [Google Scholar] [CrossRef] [PubMed]
- Pasternak, C.; Wong, S.; Elson, E.L. Mechanical function of dystrophin in muscle cells. J. Cell Biol. 1995, 128, 355–361. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, S.H.; Lee, J.H.; Lee, K.A.; Choi, Y.C. Clinical and genetic characterization of female Dystrophinopathy. J. Clin. Neurol. 2015, 11, 248–251. [Google Scholar] [CrossRef] [PubMed]
- Ishikawa, Y.; Miura, T.; Ishikawa, Y.; Aoyagi, T.; Ogata, H.; Hamada, S.; Minami, R. Duchenne muscular dystrophy: Survival by cardio-respiratory interventions. Neuromuscul. Disord. 2011, 21, 47–51. [Google Scholar] [CrossRef] [PubMed]
- Bladen, C.L.; Salgado, D.; Monges, S.; Foncuberta, M.E.; Kekou, K.; Kosma, K.; Dawkins, H.; Lamont, L.; Roy, A.J.; Chamova, T.; et al. The TREAT-NMD DMD Global Database: Analysis of more than 7,000 Duchenne muscular dystrophy mutations. Hum. Mutat. 2015, 36, 395–402. [Google Scholar] [CrossRef] [PubMed]
- Koenig, M.; Beggs, A.H.; Moyer, M.; Scherpf, S.; Heindrich, K.; Bettecken, T.; Meng, G.; Müller, C.R.; Lindlöf, M.; Kaariainen, H.; et al. The molecular basis for Duchenne versus Becker muscular dystrophy: Correlation of severity with type of deletion. Am. J. Hum. Genet. 1989, 45, 498–506. [Google Scholar]
- Welch, E.M.; Barton, E.R.; Zhuo, J.; Tomizawa, Y.; Friesen, W.J.; Trifillis, P.; Paushkin, S.; Patel, M.; Trotta, C.R.; Hwang, S.; et al. PTC124 targets genetic disorders caused by nonsense mutations. Nature 2007, 447, 87–91. [Google Scholar] [CrossRef]
- Shimizu-Motohashi, Y.; Miyatake, S.; Komaki, H.; Takeda, S.; Aoki, Y. Recent advances in innovative therapeutic approaches for Duchenne muscular dystrophy: From discovery to clinical trials. Am. J. Transl. Res. 2016, 8, 2471–2489. [Google Scholar]
- McDonald, C.M.; Campbell, C.; Torricelli, R.E.; Finkel, R.S.; Flanigan, K.M.; Goemans, N.; Heydemann, P.; Kaminska, A.; Kirschner, J.; Muntoni, F.; et al. Ataluren in patients with nonsense mutation Duchenne muscular dystrophy (ACT DMD): A multicentre, randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2017, 390, 1489–1498. [Google Scholar] [CrossRef]
- Davies, J.; Gorini, L.; Davis, B.D. Misreading of RNA codewords induced by aminoglycoside antibiotics. Mol. Pharmacol. 1965, 1, 93–106. [Google Scholar]
- Palmer, E.; Wilhelm, J.M.; Sherman, F. Phenotypic suppression of nonsense mutants in yeast by aminoglycoside antibiotics. Nature 1979, 277, 148–150. [Google Scholar] [CrossRef] [PubMed]
- Sicinski, P.; Geng, Y.; Ryder-Cook, A.S.; Barnard, E.A.; Darlison, M.G.; Barnard, P.J. The molecular basis of muscular dystrophy in the mdx mouse: A point mutation. Science 1989, 244, 1578–1580. [Google Scholar] [CrossRef] [PubMed]
- Barton-Davis, E.R.; Cordier, L.; Shoturma, D.I.; Leland, S.E.; Sweeney, H.L. Aminoglycoside antibiotics restore dystrophin function to skeletal muscles of mdx mice. J. Clin. Invest. 1999, 104, 375–381. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wagner, K.R.; Hamed, S.; Hadley, D.W.; Gropman, A.L.; Burstein, A.H.; Escolar, D.M.; Hoffman, E.P.; Fischbeck, K.H. Gentamicin treatment of Duchenne and Becker muscular dystrophy due to nonsense mutations. Ann. Neurol. 2001, 49, 706–711. [Google Scholar] [CrossRef] [PubMed]
- Malik, V.; Rodino-Klapac, L.R.; Viollet, L.; Wall, C.; King, W.; Al-Dahhak, R.; Lewis, S.; Shilling, C.J.; Kota, J.; Serrano-Munuera, C.; et al. Gentamicin-induced readthrough of stop codons in Duchenne muscular dystrophy. Ann. Neurol. 2010, 67, 771–780. [Google Scholar] [CrossRef]
- Finkel, R.S.; Flanigan, K.M.; Wong, B.; Bönnemann, C.; Sampson, J.; Sweeney, H.L.; Reha, A.; Northcutt, V.J.; Elfring, G.; Barth, J.; et al. Phase 2a study of ataluren-mediated dystrophin production in patients with nonsense mutation Duchenne muscular dystrophy. PLoS ONE 2013, 8, e81302. [Google Scholar] [CrossRef] [PubMed]
- Bushby, K.; Finkel, R.; Wong, B.; Barohn, R.; Campbell, C.; Comi, G.P.; Connolly, A.M.; Day, J.W.; Flanigan, K.M.; Goemans, N.; et al. Ataluren treatment of patients with nonsense mutation dystrophinopathy. Muscle Nerve 2014, 50, 477–487. [Google Scholar] [CrossRef] [Green Version]
- Available online: http://ir.ptcbio.com/news-releases/news-release-details/ptc-therapeutics-announces-initial-data-patient-registry (accessed on 11 November 2018).
- Mullard, A. EMA reconsiders 'read-through' drug against Duchenne muscular dystrophy following appeal. Nat. Biotechnol. 2014, 32, 706. [Google Scholar] [CrossRef]
- Available online: https://www.ema.europa.eu/en/medicines/human/EPAR/translarna (accessed on 11 November 2018).
- Available online: http://www. Retrieved from ptcbio.com/en/pipeline/clinical-trials-ataluren/ (accessed on 11 November 2018).
- Aartsma-Rus, A.; Fokkema, I.; Verschuuren, J.; Ginjaar, I.; van Deutekom, J.; van Ommen, G.J.; den Dunnen, J.T. Theoretic applicability of antisense-mediated exon skipping for Duchenne muscular dystrophy mutations. Hum. Mutat. 2009, 30, 293–299. [Google Scholar] [CrossRef]
- Nakamura, A.; Si, T. Exon-skipping therapy for Duchenne muscular dystrophy. Lancet 2011, 378, 546–547. [Google Scholar] [CrossRef]
- Shimizu-Motohashi, Y.; Murakami, T.; Kimura, E.; Komaki, H.; Watanabe, N. Exon skipping for Duchenne muscular dystrophy: A systematic review and meta-analysis. Orphanet. J. Rare Dis. 2018, 13, 93. [Google Scholar] [CrossRef] [PubMed]
- Mendell, J.R.; Rodino-Klapac, L.R.; Sahenk, Z.; Roush, K.; Bird, L.; Lowes, L.P.; Alfano, L.; Gomez, A.M.; Lewis, S.; Kota, J.; et al. Eteplirsen for the treatment of Duchenne muscular dystrophy. Ann. Neurol. 2013, 74, 637–647. [Google Scholar] [CrossRef] [Green Version]
- Kesselheim, A.S.; Avorn, J. Approving a problematic muscular dystrophy drug: Implications for FDA Policy. JAMA 2016, 316, 2357–2358. [Google Scholar] [CrossRef] [PubMed]
- Available online: https://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm521263.htm (accessed on 11 November 2018).
- Mendell, J.R.; Goemans, N.; Lowes, L.P.; Alfano, L.N.; Berry, K.; Shao, J.; Kaye, E.M.; Mercuri, E.; Eteplirsen Study Group and Telethon Foundation DMD Italian Network. Longitudinal effect of eteplirsen versus historical control on ambulation in Duchenne muscular dystrophy. Ann. Neurol. 2016, 79, 257–271. [Google Scholar] [CrossRef] [PubMed]
- Muntoni, F.; Frank, D.; Sardone, V.; Morgan, J.; Schnell, F.; Charleston, J.; Desjardins, C.; Phadke, R.; Sewry, C.; Popplewell, L.; et al. Golodirsen induces exon skipping leading to sarcolemmal dystrophin expression in Duchenne muscular dystrophy patients With mutations amenable to Exon 53 skipping. Neurology 2018, 90, S22.001. [Google Scholar]
- Komaki, H.; Nagata, T.; Saito, T.; Masuda, S.; Takeshita, E.; Sasaki, M.; Tachimori, H.; Nakamura, H.; Aoki, Y.; Takeda, S. Systemic administration of the antisense oligonucleotide NS-065/NCNP-01 for skipping of exon 53 in patients with Duchenne muscular dystrophy. Sci. Transl. Med. 2018, 10. [Google Scholar] [CrossRef] [PubMed]
- Available online: https://www.sarepta.com/pipeline/exon-skipping-duchenne (accessed on 11 November 2018).
- Moulton, H.M.; Moulton, J.D. Morpholinos and their peptide conjugates: Therapeutic promise and challenge for Duchenne muscular dystrophy. Biochim. Biophys. Acta 2010, 1798, 2296–2303. [Google Scholar] [CrossRef] [Green Version]
- Wu, R.P.; Youngblood, D.S.; Hassinger, J.N.; Lovejoy, C.E.; Nelson, M.H.; Iversen, P.L.; Moulton, H.M. Cell-penetrating peptides as transporters for morpholino oligomers: Effects of amino acid composition on intracellular delivery and cytotoxicity. Nucleic Acids Res. 2007, 35, 5182–5191. [Google Scholar] [CrossRef]
- Amantana, A.; Moulton, H.M.; Cate, M.L.; Reddy, M.T.; Whitehead, T.; Hassinger, J.N.; Youngblood, D.S.; Iversen, P.L. Pharmacokinetics, biodistribution, stability and toxicity of a cell-penetrating peptide-morpholino oligomer conjugate. Bioconjug. Chem. 2007, 18, 1325–1331. [Google Scholar] [CrossRef]
- Available online: https://ir.wavelifesciences.com/static-files/6285cdfd-2559-474b-9997-8b20eccbcad4 (accessed on 11 November 2018).
- 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]
- Available online: https://www.daiichisankyo.com/media_investors/media_relations/press_releases/detail/006840.html (accessed on 11 November 2018).
- England, S.B.; Nicholson, L.V.; Johnson, M.A.; Forrest, S.M.; Love, D.R.; Zubrzycka-Gaarn, E.E.; Bulman, D.E.; Harris, J.B.; Davies, K.E. Very mild muscular dystrophy associated with the deletion of 46% of dystrophin. Nature 1990, 343, 180–182. [Google Scholar] [CrossRef] [PubMed]
- Benedetti, S.; Hoshiya, H.; Tedesco, F.S. Repair or replace? Exploiting novel gene and cell therapy strategies for muscular dystrophies. FEBS J. 2013, 280, 4263–4280. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chamberlain, J.S. Gene therapy of muscular dystrophy. Hum. Mol. Genet. 2002, 11, 2355–2362. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cox, G.A.; Sunada, Y.; Campbell, K.P.; Chamberlain, J.S. Dp71 can restore the dystrophin-associated glycoprotein complex in muscle but fails to prevent dystrophy. Nat. Genet. 1994, 8, 333–339. [Google Scholar] [CrossRef]
- Rafael, J.A.; Cox, G.A.; Corrado, K.; Jung, D.; Campbell, K.P.; Chamberlain, J.S. Forced expression of dystrophin deletion constructs reveals structure-function correlations. J. Cell Biol. 1996, 134, 93–102. [Google Scholar] [CrossRef] [PubMed]
- Wang, B.; Li, J.; Xiao, X. Adeno-associated virus vector carrying human minidystrophin genes effectively ameliorates muscular dystrophy in mdx mouse model. Proc. Natl. Acad. Sci. USA 2000, 97, 13714–13719. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Duan, D. Systemic AAV micro-dystrophin gene therapy for Duchenne muscular dystrophy. Mol. Ther. 2018, 26, 2337–2356. [Google Scholar] [CrossRef] [PubMed]
- Harper, S.Q.; Hauser, M.A.; DelloRusso, C.; Duan, D.; Crawford, R.W.; Phelps, S.F.; Harper, H.A.; Robinson, A.S.; Engelhardt, J.F.; Brooks, S.V.; et al. Modular flexibility of dystrophin: Implications for gene therapy of Duchenne muscular dystrophy. Nat. Med. 2002, 8, 253–261. [Google Scholar] [CrossRef] [PubMed]
- Wang, Z.; Zhu, T.; Qiao, C.; Zhou, L.; Wang, B.; Zhang, J.; Chen, C.; Li, J.; Xiao, X. Adeno-associated virus serotype 8 efficiently delivers genes to muscle and heart. Nat. Biotechnol. 2005, 23, 321–328. [Google Scholar] [CrossRef]
- Gregorevic, P.; Allen, J.M.; Minami, E.; Blankinship, M.J.; Haraguchi, M.; Meuse, L.; Finn, E.; Adams, M.E.; Froehner, S.C.; Murry, C.E.; et al. rAAV6-microdystrophin preserves muscle function and extends lifespan in severely dystrophic mice. Nat. Med. 2006, 12, 787–789. [Google Scholar] [CrossRef] [Green Version]
- Yue, Y.; Pan, X.; Hakim, C.H.; Kodippili, K.; Zhang, K.; Shin, J.H.; Yang, H.T.; McDonald, T.; Duan, D. Safe and bodywide muscle transduction in young adult Duchenne muscular dystrophy dogs with adeno-associated virus. Hum. Mol. Genet. 2015, 24, 5880–5890. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mendell, J.R.; Campbell, K.; Rodino-Klapac, L.; Sahenk, Z.; Shilling, C.; Lewis, S.; Bowles, D.; Gray, S.; Li, C.; Galloway, G.; et al. Dystrophin immunity in Duchenne's muscular dystrophy. N. Engl. J. Med. 2010, 363, 1429–1437. [Google Scholar] [CrossRef] [PubMed]
- Bowles, D.E.; McPhee, S.W.; Li, C.; Gray, S.J.; Samulski, J.J.; Camp, A.S.; Li, J.; Wang, B.; Monahan, P.E.; Rabinowitz, J.E.; et al. Phase 1 gene therapy for Duchenne muscular dystrophy using a translational optimized AAV vector. Mol. Ther. 2012, 20, 443–455. [Google Scholar] [CrossRef] [PubMed]
- Ho, P.P.; Lahey, L.J.; Mourkioti, F.; Kraft, P.E.; Filareto, A.; Brandt, M.; Magnusson, K.E.G.; Finn, E.E.; Chamberlain, J.S.; Robinson, W.H.; et al. Engineered DNA plasmid reduces immunity to dystrophin while improving muscle force in a model of gene therapy of Duchenne dystrophy. Proc. Natl. Acad. Sci. USA 2018, 115, E9182–E9191. [Google Scholar] [CrossRef] [PubMed]
- Barthélémy, F.; Wein, N. Personalized gene and cell therapy for Duchenne muscular dystrophy. Neuromuscul. Disord. 2018, 28, 803–824. [Google Scholar] [CrossRef] [PubMed]
- Mendell, J.R.; Kissel, J.T.; Amato, A.A.; King, W.; Signore, L.; Prior, T.W.; Sahenk, Z.; Benson, S.; McAndrew, P.E.; Rice, R. Myoblast transfer in the treatment of Duchenne's muscular dystrophy. N. Engl. J. Med. 1995, 333, 832–838. [Google Scholar] [CrossRef] [PubMed]
- Skuk, D.; Goulet, M.; Roy, B.; Chapdelaine, P.; Bouchard, J.P.; Roy, R.; Dugré, F.J.; Sylvain, M.; Lachance, J.G.; Deschênes, L.; et al. Dystrophin expression in muscles of duchenne muscular dystrophy patients after high-density injections of normal myogenic cells. J. Neuropathol. Exp. Neurol. 2006, 65, 371–386. [Google Scholar] [CrossRef]
- Skuk, D.; Roy, B.; Goulet, M.; Chapdelaine, P.; Bouchard, J.P.; Roy, R.; Dugré, F.J.; Lachance, J.G.; Deschênes, L.; Hélène, S.; et al. Dystrophin expression in myofibers of Duchenne muscular dystrophy patients following intramuscular injections of normal myogenic cells. Mol. Ther. 2004, 9, 475–482. [Google Scholar] [CrossRef]
- Torrente, Y.; Belicchi, M.; Marchesi, C.; D'Antona, G.; Cogiamanian, F.; Pisati, F.; Gavina, M.; Giordano, R.; Tonlorenzi, R.; Fagiolari, G.; et al. Autologous transplantation of muscle-derived CD133+ stem cells in Duchenne muscle patients. Cell Transplant. 2007, 16, 563–577. [Google Scholar] [CrossRef]
- Torrente, Y.; Belicchi, M.; Sampaolesi, M.; Pisati, F.; Meregalli, M.; D'Antona, G.; Tonlorenzi, R.; Porretti, L.; Gavina, M.; Mamchaoui, K.; et al. Human circulating AC133(+) stem cells restore dystrophin expression and ameliorate function in dystrophic skeletal muscle. J. Clin. Invest. 2004, 114, 182–195. [Google Scholar] [CrossRef]
- Minasi, M.G.; Riminucci, M.; De Angelis, L.; Borello, U.; Berarducci, B.; Innocenzi, A.; Caprioli, A.; Sirabella, D.; Baiocchi, M.; De Maria, R.; et al. The meso-angioblast: A multipotent, self-renewing cell that originates from the dorsal aorta and differentiates into most mesodermal tissues. Development 2002, 129, 2773–2783. [Google Scholar]
- Sampaolesi, M.; Blot, S.; D'Antona, G.; Granger, N.; Tonlorenzi, R.; Innocenzi, A.; Mognol, P.; Thibaud, J.L.; Galvez, B.G.; Barthélémy, I.; et al. Mesoangioblast stem cells ameliorate muscle function in dystrophic dogs. Nature 2006, 444, 574–579. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cossu, G.; Previtali, S.C.; Napolitano, S.; Cicalese, M.P.; Tedesco, F.S.; Nicastro, F.; Noviello, M.; Roostalu, U.; Natali Sora, M.G.; Scarlato, M.; et al. Intra-arterial transplantation of HLA-matched donor mesoangioblasts in Duchenne muscular dystrophy. EMBO Mol. Med. 2015, 7, 1513–1528. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dellavalle, A.; Sampaolesi, M.; Tonlorenzi, R.; Tagliafico, E.; Sacchetti, B.; Perani, L.; Innocenzi, A.; Galvez, B.G.; Messina, G.; Morosetti, R.; et al. Pericytes of human skeletal muscle are myogenic precursors distinct from satellite cells. Nat. Cell Biol. 2007, 9, 255–267. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, P.; Cui, K.; Zhang, B.; Wang, Z.; Shen, Y.; Wang, X.; Zhang, J.; Tong, F.; Li, S. Transplantation of human umbilical cord-derived mesenchymal stems cells for the treatment of Becker muscular dystrophy in affected pedigree members. Int. J. Mol. Med. 2015, 35, 1051–1057. [Google Scholar] [CrossRef] [Green Version]
- Darabi, R.; Arpke, R.W.; Irion, S.; Dimos, J.T.; Grskovic, M.; Kyba, M.; Perlingeiro, R.C. Human ES- and iPS-derived myogenic progenitors restore dystrophin and improve contractility upon transplantation in dystrophic mice. Cell Stem Cell 2012, 10, 610–619. [Google Scholar] [CrossRef] [PubMed]
- Young, C.S.; Hicks, M.R.; Ermolova, N.V.; Nakano, H.; Jan, M.; Younesi, S.; Karumbayaram, S.; Kumagai-Cresse, C.; Wang, D.; Zack, J.A.; et al. A single CRISPR-Cas9 deletion strategy that targets the majority of DMD patients restores dystrophin function in hiPSC-derived muscle cells. Cell Stem Cell 2016, 18, 533–540. [Google Scholar] [CrossRef]
- Benchaouir, R.; Meregalli, M.; Farini, A.; D'Antona, G.; Belicchi, M.; Goyenvalle, A.; Battistelli, M.; Bresolin, N.; Bottinelli, R.; Garcia, L.; et al. Restoration of human dystrophin following transplantation of exon-skipping-engineered DMD patient stem cells into dystrophic mice. Cell Stem Cell 2007, 1, 646–657. [Google Scholar] [CrossRef]
- Meng, J.; Counsell, J.R.; Reza, M.; Laval, S.H.; Danos, O.; Thrasher, A.; Lochmüller, H.; Muntoni, F.; Morgan, J.E. Autologous skeletal muscle derived cells expressing a novel functional dystrophin provide a potential therapy for Duchenne Muscular Dystrophy. Sci. Rep. 2016, 6, 19750. [Google Scholar] [CrossRef] [Green Version]
- Tedesco, F.S.; Hoshiya, H.; D'Antona, G.; Gerli, M.F.; Messina, G.; Antonini, S.; Tonlorenzi, R.; Benedetti, S.; Berghella, L.; Torrente, Y.; et al. Stem cell-mediated transfer of a human artificial chromosome ameliorates muscular dystrophy. Sci. Transl. Med. 2011, 3, 96ra78. [Google Scholar] [CrossRef]
- Mauro, A. Satellite cell of skeletal muscle fibers. J. Biophys. Biochem. Cytol. 1961, 9, 493–495. [Google Scholar] [CrossRef] [PubMed]
- Tedesco, F.S.; Dellavalle, A.; Diaz-Manera, J.; Messina, G.; Cossu, G. Repairing skeletal muscle: Regenerative potential of skeletal muscle stem cells. J. Clin. Invest. 2010, 120, 11–19. [Google Scholar] [CrossRef] [PubMed]
- Usas, A.; Mačiulaitis, J.; Mačiulaitis, R.; Jakubonienė, N.; Milašius, A.; Huard, J. Skeletal muscle-derived stem cells: Implications for cell-mediated therapies. Medicina 2011, 47, 469–479. [Google Scholar] [CrossRef] [PubMed]
- Meng, J.; Chun, S.; Asfahani, R.; Lochmüller, H.; Muntoni, F.; Morgan, J. Human skeletal muscle-derived CD133(+) cells form functional satellite cells after intramuscular transplantation in immunodeficient host mice. Mol. Ther. 2014, 22, 1008–1017. [Google Scholar] [CrossRef] [PubMed]
- Farrington-Rock, C.; Crofts, N.J.; Doherty, M.J.; Ashton, B.A.; Griffin-Jones, C.; Canfield, A.E. Chondrogenic and adipogenic potential of microvascular pericytes. Circulation 2004, 110, 2226–2232. [Google Scholar] [CrossRef]
- Li, H.L.; Fujimoto, N.; Sasakawa, N.; Shirai, S.; Ohkame, T.; Sakuma, T.; Tanaka, M.; Amano, N.; Watanabe, A.; Sakurai, H., etc. Precise correction of the dystrophin gene in duchenne muscular dystrophy patient induced pluripotent stem cells by TALEN and CRISPR-Cas9. Stem Cell Rep 2015, 4, 143–154. [Google Scholar] [CrossRef]
- Available online: http://www.ema.europa.eu/docs/en_GB/document_library/Scientific_guideline/2015/12/WC500199239.pdf (accessed on 11 November 2018).
- Available online: https://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/UCM450229.pdf (accessed on 11 November 2018).
- Gagne, J.J.; Thompson, L.; O'Keefe, K.; Kesselheim, A.S. Innovative research methods for studying treatments for rare diseases: Methodological review. BMJ 2014, 349, g6802. [Google Scholar] [CrossRef]
- Ezzat, K.; Aoki, Y.; Koo, T.; McClorey, G.; Benner, L.; Coenen-Stass, A.; O'Donovan, L.; Lehto, T.; Garcia-Guerra, A.; Nordin, J.; et al. Self-assembly into nanoparticles is essential for receptor mediated uptake of therapeutic antisense oligonucleotides. Nano Lett. 2015, 15, 4364–4373. [Google Scholar] [CrossRef]
- Lim, K.R.; Maruyama, R.; Yokota, T. Eteplirsen in the treatment of Duchenne muscular dystrophy. Drug Des. Dev. Ther. 2017, 11, 533–545. [Google Scholar] [CrossRef] [Green Version]
- Fanin, M.; Freda, M.P.; Vitiello, L.; Danieli, G.A.; Pegoraro, E.; Angelini, C. Duchenne phenotype with in-frame deletion removing major portion of dystrophin rod: Threshold effect for deletion size? Muscle Nerve 1996, 19, 1154–1160. [Google Scholar] [CrossRef]
- Benedetti, S.; Uno, N.; Hoshiya, H.; Ragazzi, M.; Ferrari, G.; Kazuki, Y.; Moyle, L.A.; Tonlorenzi, R.; Lombardo, A.; Chaouch, S.; et al. Reversible immortalisation enables genetic correction of human muscle progenitors and engineering of next-generation human artificial chromosomes for Duchenne muscular dystrophy. EMBO Mol. Med. 2018, 10, 254–275. [Google Scholar] [CrossRef] [PubMed]
- Counsell, J.R.; Asgarian, Z.; Meng, J.; Ferrer, V.; Vink, C.A.; Howe, S.J.; Waddington, S.N.; Thrasher, A.J.; Muntoni, F.; Morgan, J.E.; et al. Lentiviral vectors can be used for full-length dystrophin gene therapy. Sci. Rep. 2017, 7, 44775. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sweeney, N.P.; Meng, J.; Patterson, H.; Morgan, J.E.; McClure, M. Delivery of large transgene cassettes by foamy virus vector. Sci. Rep. 2017, 7, 8085. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Le Guiner, C.; Servais, L.; Montus, M.; Larcher, T.; Fraysse, B.; Moullec, S.; Allais, M.; François, V.; Dutilleul, M.; Malerba, A.; et al. Long-term microdystrophin gene therapy is effective in a canine model of Duchenne muscular dystrophy. Nat. Commun. 2017, 8, 16105. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Drug/Compound | Description | Company/Institute | Clinical Trial Number | Status |
---|---|---|---|---|
Ataluren | Read-through | PTC Therapeutics | Conditional approval in Europe | |
NCT01247207 | Phase 3 | |||
NCT01557400 | Phase 3 | |||
NCT02090959 | Phase 3 | |||
NCT03179631 | Phase 3 | |||
Eteplirsen | Exon skip PMO targeting exon 51 | Sarepta Therapeutics | Conditional approval in the USA | |
NCT02255552 | Phase 3 | |||
NCT02420379 | Phase 2 | |||
NCT03218995 | Phase 2 | |||
SRP-4053 (Golodirsen) | Exon skip PMO targeting exon 53 | Sarepta Therapeutics | NCT02310906 | Phase 1/2 |
NCT02500381 | Phase 3 | |||
NCT03532542 | Phase 3 | |||
SRP-4045 (Casimersen) | Exon skip PMO targeting exon 45 | Sarepta Therapeutics | NCT02500381 | Phase 3 |
NCT02530905 | Phase 1 | |||
NCT03532542 | Phase 3 | |||
SRP-5051 | Exon skip PPMO targeting exon 51 | Sarepta Therapeutics | NCT03375255 | Phase 1 |
NCT03675126 | Phase 1/2 | |||
NS-065/NCNP-01 | Exon skip PMO targeting exon 53 | NS Pharma, Inc., Nippon Shinyaku Co., Ltd., Cooperative International Neuromuscular Research Group, Therapeutic Research in Neuromuscular Disorders Solutions (TRiNDS) | NCT03167255 | Phase 2 |
DS-5141b | ENA Exon skip targeting exon 45 | Daiichi Sankyo Co., Ltd., Orphan Disease Treatment Institute Co., Ltd., Daiichi Sankyo, Inc. | NCT02667483 | Phase 1/2 |
WVE-210201 | Steropure ASO Exon skip targeting exon 51 | Wave Life Sciences Ltd. | NCT03508947 | Phase 1 |
rAAVrh74.MCK.GALGT2 | Micro-dystrophin Gene Transfer | Nationwide Children’s Hospital (USA) | NCT03333590 | Phase 1/2 |
PF-06939926 | Mini-dystrophin gene transfer | Pfizer | NCT03362502 | Phase 1 |
SGT-001 | Micro-dystrophin gene transfer | Solid Biosciences, LLC | NCT03368742 | Phase 1/2 |
rAAVrh74.MHCK7.micro-dystrophin | Micro-dystrophin gene transfer | Nationwide Children’s Hospital, Washington University School of Medicine (USA) | NCT03375164 | Phase 1/2 |
Myoblasts | Cell therapy | CHU de Quebec-Universite Laval (Canada) | NCT02196467 | Phase 1/2 |
Umbilical cord mesenchymal stem cells | Cell therapy | Allergy and Asthma Consultants (USA), Aidan Foundation | NCT02235844 | Phase 1 |
Bone marrow-derived autologous stem cells | Cell therapy | Stem Cells Arabia | NCT03067831 | Phase 1/2 |
© 2019 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 (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Shimizu-Motohashi, Y.; Komaki, H.; Motohashi, N.; Takeda, S.; Yokota, T.; Aoki, Y. Restoring Dystrophin Expression in Duchenne Muscular Dystrophy: Current Status of Therapeutic Approaches. J. Pers. Med. 2019, 9, 1. https://doi.org/10.3390/jpm9010001
Shimizu-Motohashi Y, Komaki H, Motohashi N, Takeda S, Yokota T, Aoki Y. Restoring Dystrophin Expression in Duchenne Muscular Dystrophy: Current Status of Therapeutic Approaches. Journal of Personalized Medicine. 2019; 9(1):1. https://doi.org/10.3390/jpm9010001
Chicago/Turabian StyleShimizu-Motohashi, Yuko, Hirofumi Komaki, Norio Motohashi, Shin’ichi Takeda, Toshifumi Yokota, and Yoshitsugu Aoki. 2019. "Restoring Dystrophin Expression in Duchenne Muscular Dystrophy: Current Status of Therapeutic Approaches" Journal of Personalized Medicine 9, no. 1: 1. https://doi.org/10.3390/jpm9010001
APA StyleShimizu-Motohashi, Y., Komaki, H., Motohashi, N., Takeda, S., Yokota, T., & Aoki, Y. (2019). Restoring Dystrophin Expression in Duchenne Muscular Dystrophy: Current Status of Therapeutic Approaches. Journal of Personalized Medicine, 9(1), 1. https://doi.org/10.3390/jpm9010001