376
Views
2
CrossRef citations to date
0
Altmetric
Review

Pharmacological Profile of Viltolarsen for the Treatment of Duchenne Muscular Dystrophy: A Japanese Experience

ORCID Icon & ORCID Icon
Pages 235-242 | Published online: 16 Dec 2021

References

  • Mendell JR, Shilling C, Leslie ND, et al. Evidence-based path to newborn screening for Duchenne muscular dystrophy. Ann Neurol. 2012;71(3):304–313. doi:10.1002/ana.2352822451200
  • Hoffman EP, Brown RH, Kunkel LM. Dystrophin: the protein product of the Duchenne muscular dystrophy locus. Cell. 1987;51(6):919–928. doi:10.1016/0092-8674(87)90579-43319190
  • Zubrzycka-Gaarn EE, Bulman DE, Karpati G, et al. The Duchenne muscular dystrophy gene product is localized in sarcolemma of human skeletal muscle. Nature. 1988;333(6172):466–469. doi:10.1038/333466a03287171
  • Manzur AY, Kinali M, Muntoni F. Update on the management of Duchenne muscular dystrophy. Arch Dis Child. 2008;93(11):986–990. doi:10.1136/adc.2007.11814118667451
  • Mah J. Current and emerging treatment strategies for Duchenne muscular dystrophy. Neuropsychiatr Dis Treat. 2016;12:1795–1807. doi:10.2147/NDT.S9387327524897
  • Koenig M, Hoffman EP, Bertelson CJ, Monaco AP, Feener C, Kunkel LM. Complete cloning of the Duchenne muscular dystrophy (DMD) cDNA and preliminary genomic organization of the DMD gene in normal and affected individuals. Cell. 1987;50(3):509–517. doi:10.1016/0092-8674(87)90504-63607877
  • van Deutekom JCT, van Ommen G-JB. Advances in Duchenne muscular dystrophy gene therapy. Nat Rev Genet. 2003;4(10):774–783. doi:10.1038/nrg118014526374
  • Juan-Mateu J, Gonzalez-Quereda L, Rodriguez MJ, et al. DMD mutations in 576 dystrophinopathy families: a step forward in genotype-phenotype correlations. Ervasti JM, ed. PLoS One. 2015;10(8):e0135189. doi:10.1371/journal.pone.013518926284620
  • Oudet C, Hanauer A, Clemens P, Caskey T, Mandel J-L. Two hot spots of recombination in the DMD gene correlate with the deletion prone regions. Hum Mol Genet. 1992;1(8):599–603. doi:10.1093/hmg/1.8.5991363782
  • Muntoni F, Torelli S, Ferlini A. Dystrophin and mutations: one gene, several proteins, multiple phenotypes. Lancet Neurol. 2003;2(12):731–740. doi:10.1016/S1474-4422(03)00585-414636778
  • Werneck LC, Lorenzoni PJ, Ducci RD-P, Fustes OH, Kay CSK, Scola RH. Duchenne muscular dystrophy: an historical treatment review. Arq Neuropsiquiatr. 2019;77(8):579–589. doi:10.1590/0004-282x2019008831508685
  • Rutter MM, Collins J, Rose SR, et al. Growth hormone treatment in boys with Duchenne muscular dystrophy and glucocorticoid-induced growth failure. Neuromuscul Disord. 2012;22(12):1046–1056. doi:10.1016/j.nmd.2012.07.00922967789
  • Drachman DB, Toyka KV, Myer E. Prednisone in Duchenne muscular dystrophy. Lancet. 1974;304(7894):1409–1412. doi:10.1016/S0140-6736(74)90071-3
  • Wood CL, Straub V, Guglieri M, Bushby K, Cheetham T. Short stature and pubertal delay in Duchenne muscular dystrophy. Arch Dis Child. 2016;101(1):101–106. doi:10.1136/archdischild-2015-30865426141541
  • Echevarría L, Aupy P, Goyenvalle A. Exon-skipping advances for Duchenne muscular dystrophy. Hum Mol Genet. 2018;27(R2):R163–R172. doi:10.1093/hmg/ddy17129771317
  • Sheikh O, Yokota T. Developing DMD therapeutics: a review of the effectiveness of small molecules, stop-codon readthrough, dystrophin gene replacement, and exon-skipping therapies. Expert Opin Investig Drugs. 2021;30(2):167–176. doi:10.1080/13543784.2021.1868434
  • Verhaart IEC, Aartsma-Rus A. Therapeutic developments for Duchenne muscular dystrophy. Nat Rev Neurol. 2019;15(7):373–386. doi:10.1038/s41582-019-0203-331147635
  • Aartsma-Rus A, Corey DR. The 10th oligonucleotide therapy approved: golodirsen for Duchenne muscular dystrophy. Nucleic Acid Ther. 2020;30(2):67–70. doi:10.1089/nat.2020.084532043902
  • Casimersen (Amondys 45) for Duchenne muscular dystrophy. Med Lett Drugs Ther. 2021;63(1627):e104–e105. Available from: http://www.ncbi.nlm.nih.gov/pubmed/34181634. Accessed November 15, 2021.34181634
  • Shirley M. Casimersen: first approval. Drugs. 2021;81(7):875–879. doi:10.1007/s40265-021-01512-233861387
  • Roshmi RR, Yokota T. Viltolarsen for the treatment of Duchenne muscular dystrophy. Drugs Today. 2019;55(10):627. doi:10.1358/dot.2019.55.10.3045038
  • Dhillon S. Viltolarsen: first approval. Drugs. 2020;80(10):1027–1031. doi:10.1007/s40265-020-01339-332519222
  • Lim KR, Maruyama R, Yokota T. Eteplirsen in the treatment of Duchenne muscular dystrophy. Drug Des Devel Ther. 2017;11:533–545. doi:10.2147/DDDT.S97635
  • Nan Y, Zhang Y-J. Antisense phosphorodiamidate morpholino oligomers as novel antiviral compounds. Front Microbiol. 2018;9. doi:10.3389/fmicb.2018.00750
  • Gao QQ, McNally EM. The dystrophin complex: structure, function, and implications for therapy. In: Comprehensive Physiology. Wiley; 2015:1223–1239. doi:10.1002/cphy.c140048
  • Le Rumeur E. Dystrophin and the two related genetic diseases, Duchenne and Becker muscular dystrophies. Bosn J Basic Med Sci. 2015;15(3). doi:10.17305/bjbms.2015.636
  • Yokota T, Lu Q, Partridge T, et al. Efficacy of systemic morpholino exon-skipping in Duchenne dystrophy dogs. Ann Neurol. 2009;65(6):667–676. doi:10.1002/ana.2162719288467
  • Watanabe N, Nagata T, Satou Y, et al. NS-065/NCNP-01: an antisense oligonucleotide for potential treatment of exon 53 skipping in Duchenne muscular dystrophy. Mol Ther Nucleic Acids. 2018;13:442–449. doi:10.1016/j.omtn.2018.09.01730388618
  • Heo Y-A. Golodirsen: first approval. Drugs. 2020;80(3):329–333. doi:10.1007/s40265-020-01267-232026421
  • Viltolarsen. Am J Health Pharm. 2020;77(24):2032–2033. doi:10.1093/ajhp/zxaa324
  • Komaki H, Takeshima Y, Matsumura T, et al. Viltolarsen in Japanese Duchenne muscular dystrophy patients: a Phase 1/2 study. Ann Clin Transl Neurol. 2020;7(12):2393–2408. doi:10.1002/acn3.5123533285037
  • Clemens PR, Rao VK, Connolly AM, et al. Safety, tolerability, and efficacy of viltolarsen in boys with Duchenne muscular dystrophy amenable to exon 53 skipping. JAMA Neurol. 2020;77(8):982. doi:10.1001/jamaneurol.2020.126432453377
  • Komaki H, Nagata T, Saito T, et al. 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(437). doi:10.1126/scitranslmed.aan0713
  • The Cooperative International Neuromuscular Research Group (CINRG). Available from: https://cinrgresearch.org/duchenne-natural-history/. Accessed November 15, 2021.
  • NS PHARMA’S VILTEPSOTM (viltolarsen) injection now FDA-approved in the U.S. for the treatment of Duchenne muscular dystrophy in patients amenable to exon 53 skipping therapy. NS Pharma News Release; 2020. Available from: https://www.nspharma.com/pdfs/Viltepso_Approval_Press_Release.pdf. Accessed November 15, 2021.
  • ClinicalTrials.gov Exploratory Study of NS-065/NCNP-01 in DMD. Available from: https://clinicaltrials.gov/ct2/show/NCT02081625. Accessed November 15, 2021.
  • ClinicalTrials.gov safety and dose finding study of NS-065/NCNP-01 in boys with Duchenne Muscular Dystrophy (DMD). Available from: https://clinicaltrials.gov/ct2/show/NCT02740972. Accessed November 15, 2021.
  • ClinicalTrials.gov Study to assess the efficacy and safety of viltolarsen in ambulant boys with DMD (RACER53). Available from: https://clinicaltrials.gov/ct2/show/NCT04060199. Accessed November 15, 2021.
  • ClinicalTrials.gov Extension Study of NS-065/NCNP-01 in boys with Duchenne Muscular Dystrophy (DMD). Available from: https://clinicaltrials.gov/ct2/show/NCT03167255. Accessed November 15, 2021.
  • Matsuo M, Takeshima Y, Nishio H. Contributions of Japanese patients to development of antisense therapy for DMD. Brain Dev. 2016;38(1):4–9. doi:10.1016/j.braindev.2015.05.01426094594
  • Takeshima Y, Nishio H, Sakamoto H, Nakamura H, Matsuo M. Modulation of in vitro splicing of the upstream intron by modifying an intra-exon sequence which is deleted from the dystrophin gene in dystrophin Kobe. J Clin Invest. 1995;95(2):515–520. doi:10.1172/JCI1176937860733
  • Shiga N, Takeshima Y, Sakamoto H, et al. Disruption of the splicing enhancer sequence within exon 27 of the dystrophin gene by a nonsense mutation induces partial skipping of the exon and is responsible for Becker muscular dystrophy. J Clin Invest. 1997;100(9):2204–2210. doi:10.1172/JCI1197579410897
  • Nakamura H, Kimura E, Mori-Yoshimura M, et al. Characteristics of Japanese Duchenne and Becker muscular dystrophy patients in a novel Japanese national registry of muscular dystrophy (Remudy). Orphanet J Rare Dis. 2013;8(1):60. doi:10.1186/1750-1172-8-6023601510
  • Remudy. Available from: http://www.remudy.jp/news/2021/09/005992.html. Accessed November 15, 2021.
  • Viltolarsen (viltepso) for Duchenne muscular dystrophy. Med Lett Drugs Ther. 2020;62(1609):167.33429411
  • Meyers TA, Townsend D. Cardiac pathophysiology and the future of cardiac therapies in Duchenne muscular dystrophy. Int J Mol Sci. 2019;20(17):4098. doi:10.3390/ijms20174098