9,197
Views
88
CrossRef citations to date
0
Altmetric
Review

Exon skipping: a first in class strategy for Duchenne muscular dystrophy

&
Pages 225-236 | Received 24 Aug 2016, Accepted 09 Dec 2016, Published online: 23 Dec 2016

References

  • Mendell JR, Shilling C, Leslie ND, et al. Evidence-based path to newborn screening for Duchenne muscular dystrophy.. Ann Neurol. 2012;71:304–313.
  • Pane M, Mazzone ES, Sivo S, et al. Long term natural history data in ambulant boys with Duchenne muscular dystrophy: 36-month changes. PLoS One. 2014;9:e108205.
  • Goemans N, Vanden Hauwe M, Signorovitch J, et al. Individualized prediction of changes in 6-minute walk distance for patients with Duchenne muscular dystrophy. PLoS One. 2016;11:e0164684.
  • Van Den Bergen JC, Hiller M, Bohringer S, et al. Validation of genetic modifiers for Duchenne muscular dystrophy: a multicentre study assessing SPP1 and LTBP4 variants. J Neurol Neurosurg Psychiatry. 2014.
  • Bello L, Flanigan KM, Weiss RB, et al. Association study of exon variants in the NF-kappaB and TGFbeta pathways identifies CD40 as a modifier of Duchenne muscular dystrophy. Am J Hum Genet. 2016. DOI:10.1016/j.ajhg.2016.08.023
  • Ricotti V, Ridout DA, Pane M, et al. The NorthStar ambulatory assessment in Duchenne muscular dystrophy: considerations for the design of clinical trials. J Neurol Neurosurg Psychiatry. 2016;87:149–155.
  • Bushby K, Finkel R, Birnkrant DJ, et al. Diagnosis and management of Duchenne muscular dystrophy, part 1: diagnosis, and pharmacological and psychosocial management. Lancet Neurol. 2010;9:77–93.
  • Bello L, Morgenroth LP, Gordish-Dressman H, et al. DMD genotypes and loss of ambulation in the CINRG Duchenne Natural History Study. Neurology. 2016;87:401–409.
  • Monaco AP, Bertelson CJ, Liechti-Gallati S, et al. An explanation for the phenotypic differences between patients bearing partial deletions of the DMD locus. Genomics. 1988;2:90–95.
  • Koenig M, Monaco AP, Kunkel LM. The complete sequence of dystrophin predicts a rod-shaped cytoskeletal protein. Cell. 1988;53:219–228.
  • Hoffman EP, Brown RH, Kunkel LM. Dystrophin: the protein product of the Duchene muscular dystrophy locus.1987. Biotechnology. 1992;24:457–466.
  • Aartsma-Rus A, Fokkema I, Verschuuren J, et al. Theoretic applicability of antisense-mediated exon skipping for Duchenne muscular dystrophy mutations. Hum Mutat. 2009;30:293–299.
  • Aartsma-Rus A. Antisense-mediated modulation of splicing: therapeutic implications for Duchenne muscular dystrophy. RNA Biol. 2010;7:453–461.
  • Aartsma-Rus A, Ginjaar IB, Bushby K. The importance of genetic diagnosis for Duchenne muscular dystrophy. J Med Genet. 2016;53:145–151.
  • Bladen CL, Salgado D, Monges S, et al. The TREAT-NMD DMD global database: analysis of more than 7,000 Duchenne muscular dystrophy mutations. Hum Mutat. 2015; DOI:10.1002/humu.22758
  • Aartsma-Rus A, Janson AA, Kaman WE, et al. Antisense-induced multiexon skipping for Duchenne muscular dystrophy makes more sense. Am J Hum Genet. 2004;74:83–92.
  • Mitrpant C, Fletcher S, Iversen PL, et al. By-passing the nonsense mutation in the 4 CV mouse model of muscular dystrophy by induced exon skipping. J Gene Med. 2009;11:46–56.
  • Yokota T, Lu QL, Partridge T, et al. Efficacy of systemic morpholino exon-skipping in Duchenne dystrophy dogs. Ann Neurol. 2009;65:667–676.
  • Aartsma-Rus A, Ferlini A, Goemans N, et al. Translational and regulatory challenges for exon skipping therapies. Hum Gene Ther. 2014;25:885–892.
  • Greer KL, Lochmüller H, Flanigan K, et al. Targeted exon skipping to correct exon duplications in the dystrophin gene. Mol Ther Nucleic Acids. 2014;3:e155.
  • Aartsma-Rus A, Janson AA, Van Ommen GJ, et al. Antisense-induced exon skipping for duplications in Duchenne muscular dystrophy. BMC Med Genet. 2007;8:43.
  • Vulin A, Wein N, Simmons TR, et al. The first exon duplication mouse model of Duchenne muscular dystrophy: a tool for therapeutic development. Neuromuscul Disord. 2015;25:827–834.
  • White SJ, Aartsma-Rus A, Flanigan KM, et al. Duplications in the DMD gene. Hum Mutat. 2006;27:938–945.
  • Gurvich OL, Tuohy TM, Howard MT, et al. DMD pseudoexon mutations: splicing efficiency, phenotype, and potential therapy. Ann Neurol. 2008;63:81–89.
  • Beroud C, Tuffery-Giraud S, Matsuo M, et al. Multiexon skipping leading to an artificial DMD protein lacking amino acids from exons 45 through 55 could rescue up to 63% of patients with Duchenne muscular dystrophy. Hum Mutat. 2007;28:196–202.
  • Van V, De Winter CL, Van Deutekom JC, et al. Assessment of the feasibility of exon 45-55 multiexon skipping for duchenne muscular dystrophy. BMC Med Genet. 2008;9:105.
  • Echigoya Y, Aoki Y, Miskew B, et al. Long-term efficacy of systemic multiexon skipping targeting dystrophin exons 45-55 with a cocktail of vivo-morpholinos in mdx52 mice. Mol Ther Nucleic Acids. 2015;4:e225.
  • Aartsma-Rus A. Dystrophin analysis in clinical trials. J Neuromuscular Dis. 2014;1:41–53.
  • Aartsma-Rus A, Van Deutekom JCT, Fokkema IF, et al. Entries in the leiden Duchenne muscular dystrophy mutation database: an overview of mutation types and paradoxical cases that confirm the reading-frame rule. Muscle Nerve. 2006;34:135–144.
  • Goemans NM, Tulinius M, Van Den Akker JT, et al. Systemic administration of PRO051 in Duchenne’s muscular dystrophy. N Engl J Med. 2011;364:1513–1522.
  • Mendell JR, Rodino-Klapac LR, Sahenk Z, et al. Eteplirsen for the treatment of Duchenne muscular dystrophy. Ann Neurol. 2013;74:637–647.
  • Van Deutekom JC, Janson AA, Ginjaar IB, et al. Local dystrophin restoration with antisense oligonucleotide PRO051. N Engl J Med. 2007;357:2677–2686.
  • Beekman C, Sipkens JA, Testerink J, et al. A sensitive, reproducible and objective immunofluorescence analysis method of dystrophin in individual fibers in samples from patients with Duchenne muscular dystrophy. PLoS One. 2014;9:e107494. DOI:10.1371/journal.pone.0107494
  • Anthony K, Arechavala-Gomeza V, Taylor LE, et al. Dystrophin quantification: Biological and translational research implications. Neurology. 2014;83:2062–2069.
  • Goemans NM, Tulinius M, Van Den Hauwe M, et al. Long-term efficacy, safety, and pharmacokinetics of drisapersen in Duchenne muscular dystrophy: results from an Open-Label extension study. PLoS One. 2016;11:e0161955. DOI:10.1371/journal.pone.0161955
  • Voit T, Topaloglu H, Straub V, et al. Safety and efficacy of drisapersen for the treatment of Duchenne muscular dystrophy (DEMAND II): an exploratory, randomised, placebo-controlled phase 2 study. Lancet Neurol. 2014;13:987–996.
  • Mazzone ES, Pane M, Sormani MP, et al. 24 month longitudinal data in ambulant boys with Duchenne muscular dystrophy. PLoS One. 2013;8:e52512.
  • McDonald CM, Henricson EK, Abresch RT, et al. The 6-minute walk test and other endpoints in Duchenne muscular dystrophy: longitudinal natural history observations over 48 weeks from a multicenter study. Muscle Nerve. 2013;48:343–356.
  • Straub V, Balabanov P, Bushby K, et al. Stakeholder cooperation to overcome challenges in orphan medicine development: the example of Duchenne muscular dystrophy. Lancet Neurol. 2016;15:882–890.
  • Van ML, Moerland M, Gallagher J, et al. Injection site reactions after subcutaneous oligonucleotide therapy. Br J Clin Pharmacol. 2016;82(2):340–351.
  • Kinali M, Arechavala-Gomeza V, Feng L, et al. Local restoration of dystrophin expression with the morpholino oligomer AVI-4658 in Duchenne muscular dystrophy: a single-blind, placebo-controlled, dose-escalation, proof-of-concept study. Lancet Neurol. 2009;8:918–928.
  • Cirak S, Arechavala-Gomeza V, Guglieri M, et al. Exon skipping and dystrophin restoration in patients with Duchenne muscular dystrophy after systemic phosphorodiamidate morpholino oligomer treatment: an open-label, phase 2, dose-escalation study. Lancet. 2011;378:595–605.
  • Mendell JR, Goemans N, Lowes LP, et al. Longitudinal effect of eteplirsen versus historical control on ambulation in Duchenne muscular dystrophy. Ann Neurol. 2016;79:257–271.
  • http://www.fda.gov/downloads/advisorycommittees/committeesmeetingmaterials/drugs/peripheralandcentralnervoussystemdrugsadvisorycommittee/ucm497064.pdf. 2016.
  • http://investorrelations.sarepta.com/phoenix.zhtml?c=64231&p=irol-newsArticle&ID=2175522. 2016.
  • http://www.accessdata.fda.gov/drugsatfda_docs/nda/2016/206488_summary%20review_Redacted.pdf. 2016.
  • http://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm521263.htm. 2016.
  • Aartsma-Rus A, Krieg AM. FDA approves eteplirsen for Duchenne muscular dystrophy - the next chapter in the eteplirsen saga. Nucleic Acid Ther. 2016. DOI:10.1089/nat.2016.0657
  • Järver P, O’Donovan L, Gait MJ. A chemical view of oligonucleotides for exon skipping and related drug applications. Nucleic Acid Ther. 2014;24:37–47.
  • Goyenvalle A, Griffith G, Babbs A, et al. Functional correction in mouse models of muscular dystrophy using exon-skipping tricyclo-DNA oligomers. Nat Med. 2015;21:270–275.
  • Rigo F, Hua Y, Chun SJ, et al. Synthetic oligonucleotides recruit ILF2/3 to RNA transcripts to modulate splicing. Nat Chem Biol. 2012;8:555–561.
  • Jirka SM, Tanganyika-De Winter CL, Boertje-Van Der Meulen JW, et al. Evaluation of 2ʹ-deoxy-2ʹ-fluoro antisense oligonucleotides for exon skipping in Duchenne muscular dystrophy. Mol Ther Nucleic Acids. 2015;4:e265.
  • Betts CA, Saleh AF, Carr CA, et al. Prevention of exercised induced cardiomyopathy following Pip-PMO treatment in dystrophic mdx mice. Sci Rep. 2015;5:8986.
  • Shabanpoor F, McClorey G, Saleh AF, et al. Bi-specific splice-switching PMO oligonucleotides conjugated via a single peptide active in a mouse model of Duchenne muscular dystrophy. Nucleic Acids Res. 2015;43:29–39.
  • Betts C, Saleh AF, Arzumanov AA, et al. 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.
  • Yin H, Saleh AF, Betts C, et al. Pip5 transduction peptides direct high efficiency oligonucleotide-mediated dystrophin exon skipping in heart and phenotypic correction in mdx mice.. Mol Ther. 2011;19:1295–1303.
  • Yin H, Moulton HM, Betts C, et al. Functional rescue of dystrophin-deficient mdx mice by a chimeric peptide-PMO. Mol Ther. 2010;18:1822–1829.
  • Yin H, Moulton HM, Seow Y, et al. Cell-penetrating peptide-conjugated antisense oligonucleotides restore systemic muscle and cardiac dystrophin expression and function. Hum Mol Genet. 2008;17:3909–3918.
  • Wu B, Moulton HM, Iversen PL, et al. Effective rescue of dystrophin improves cardiac function in dystrophin-deficient mice by a modified morpholino oligomer. Proc Natl Acad Sci U S A. 2008;105:14814–14819.
  • Moulton HM, Moulton JD. Morpholinos and their peptide conjugates: therapeutic promise and challenge for Duchenne muscular dystrophy. Biochim Biophys Acta. 2010;1798:2296–2303.
  • Jirka SM, Heemskerk H, Tanganyika-De Winter CL, et al. Peptide conjugation of 2ʹ-O-methyl phosphorothioate antisense oligonucleotides enhances cardiac uptake and exon skipping in mdx mice. Nucleic Acid Ther. 2014;24:25–36.
  • Cao L, Han G, Lin C, et al. Fructose promotes uptake and activity of oligonucleotides with different chemistries in a context-dependent manner in mdx mice. Mol Ther Nucleic Acids. 2016;5:e329.
  • Han G, Gu B, Cao L, et al. Hexose enhances oligonucleotide delivery and exon skipping in dystrophin-deficient mdx mice. Nat Commun. 2016;7:10981.
  • Coley WD, Bogdanik L, Vila MC, et al. Effect of genetic background on the dystrophic phenotype in mdx mice. Hum Mol Genet. 2016;25:130–145.
  • Young CS, Hicks MR, Ermolova NV, 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.
  • Iyombe-Engembe J-P, Ouellet DL, Barbeau X, et al. Efficient restoration of the dystrophin gene reading frame and protein structure in DMD myoblasts using the cindel method. Mol Ther Nucleic Acids. 2016;5:e283.
  • Maggio I, Stefanucci L, Janssen JM, et al. Selection-free gene repair after adenoviral vector transduction of designer nucleases: rescue of dystrophin synthesis in DMD muscle cell populations. Nucleic Acids Res. 2016;44:1449–1470.
  • Tabebordbar M, Zhu K, Cheng JKW, et al. In vivo gene editing in dystrophic mouse muscle and muscle stem cells. Science. 2016;351:407–411.
  • Nelson CE, Hakim CH, Ousterout DG, et al. In vivo genome editing improves muscle function in a mouse model of Duchenne muscular dystrophy. Science. 2016;351:403–407.
  • Long C, Amoasii L, Mireault AA, et al. Postnatal genome editing partially restores dystrophin expression in a mouse model of muscular dystrophy. Science. 2016;351:400–403.
  • Xu L, Park KH, Zhao L, et al. CRISPR-mediated genome editing restores dystrophin expression and function in mdx mice. Mol Ther. 2016;24:564–569.
  • Ousterout DG, Kabadi AM, Thakore PI, et al. Multiplex CRISPR/Cas9-based genome editing for correction of dystrophin mutations that cause Duchenne muscular dystrophy. Nat Communications. 2015;6:6244.
  • Li HL, Fujimoto N, Sasakawa N, et al. Precise correction of the dystrophin gene in Duchenne muscular dystrophy patient induced pluripotent stem cells by TALEN and CRISPR-Cas9. Stem Rep. 2015;4:143–154.
  • Long C, McAnally JR, Shelton JM, et al. Prevention of muscular dystrophy in mice by CRISPR/Cas9-mediated editing of germline DNA.. Science. 2014;345:1184–1188.
  • Béroud C, Tuffery-Giraud S, Matsuo M, et al. Multiexon skipping leading to an artificial DMD protein lacking amino acids from exons 45 through 55 could rescue up to 63% of patients with Duchenne muscular dystrophy. Hum Mutat. 2007;28:196–202.
  • Butland RJ, Pang J, Gross ER, et al. Two-, six-, and 12-minute walking tests in respiratory disease. Br Med J (Clin Res Ed). 1982;284:1607–1608.
  • Bushby K, Finkel R, Wong B, et al. Ataluren treatment of patients with nonsense mutation dystrophinopathy. Muscle Nerve. 2014;50:477–487.
  • Mayhew A, Mazzone ES, Eagle M, et al. Development of the performance of the upper limb module for Duchenne muscular dystrophy. Dev Med Child Neurol. 2013;55:1038–1045.
  • Pane M, Mazzone ES, Fanelli L, et al. Reliability of the performance of upper limb assessment in Duchenne muscular dystrophy.. Neuromuscul Disord. 2014;24:201–206.
  • Jirka S, Aartsma-Rus A. An update on RNA-targeting therapies for neuromuscular disorders. Curr Opin Neurol. 2015;28:515–521.