Skip to main content

Advertisement

Log in

Update on the Treatment of Duchenne Muscular Dystrophy

  • Nerve and Muscle (M Hirano and LH Weimer, Section Editors)
  • Published:
Current Neurology and Neuroscience Reports Aims and scope Submit manuscript

Abstract

Duchenne muscular dystrophy is the most severe childhood form of muscular dystrophy caused by mutations in the gene responsible for dystrophin production. There is no cure, and treatment is limited to glucocorticoids that prolong ambulation and drugs to treat the cardiomyopathy. Multiple treatment strategies are under investigation and have shown promise for Duchenne muscular dystrophy. Use of molecular-based therapies that replace or correct the missing or nonfunctional dystrophin protein has gained momentum. These strategies include gene replacement with adeno-associated virus, exon skipping with antisense oligonucleotides, and mutation suppression with compounds that “read through” stop codon mutations. Other strategies include cell therapy and surrogate gene products to compensate for the loss of dystrophin. All of these approaches are discussed in this review, with particular emphasis on the most recent advances made in each therapeutic discipline. The advantages of each approach and challenges in translation are outlined in detail. Individually or in combination, all of these therapeutic strategies hold great promise for treatment of this devastating childhood disease.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1

Similar content being viewed by others

References

Papers of particular interest, published recently, have been highlighted as: • Of importance •• Of major importance

  1. •• Mendell JR, et al. Evidence-based path to newborn screening for Duchenne muscular dystrophy. Ann Neurol. 2012;71:304–13. This was the first report of a two-tier system using dried blood spots obtained at birth to identify cases of DMD. Creatine kinase levels are first determined and if they reach a level predictive of DMD, DNA sequencing follows.

    Article  PubMed  CAS  Google Scholar 

  2. Koenig M, et al. Complete cloning of the Duchenne muscular dystrophy (DMD) cDNA and preliminary genomic organization of the DMD gene in normal and affected individuals. Cell. 1987;50:509–17.

    Article  PubMed  CAS  Google Scholar 

  3. Brooke MH, et al. Clinical investigation in Duchenne dystrophy: 2. Determination of the “power” of therapeutic trials based on the natural history. Muscle Nerve. 1983;6:91–103.

    Article  PubMed  CAS  Google Scholar 

  4. England SB, et al. Very mild muscular dystrophy associated with the deletion of 46 % of dystrophin. Nature. 1990;343:180–2.

    Article  PubMed  CAS  Google Scholar 

  5. Harper SQ, Hauser MA, DelloRusso C, et al. Modular flexibility of dystrophin: Implications for gene therapy of Duchenne muscular dystrophy. Nat Med. 2002;8:253–61.

    Article  PubMed  CAS  Google Scholar 

  6. 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;97:13714–9.

    Article  PubMed  CAS  Google Scholar 

  7. Gregorevic P, et al. rAAV6-microdystrophin preserves muscle function and extends lifespan in severely dystrophic mice. Nat Med. 2006;12:787–9.

    Article  PubMed  CAS  Google Scholar 

  8. Watchko J, et al. Adeno-associated virus vector-mediated minidystrophin gene therapy improves dystrophic muscle contractile function in mdx mice. Hum Gene Ther. 2002;13:1451–60.

    Article  PubMed  CAS  Google Scholar 

  9. •• Mendell JR, et al. Dystrophin immunity in Duchenne’s muscular dystrophy. N Engl J Med. 2010;363:1429–37. This work demonstrated immune response to mini-dystrophin following AAV-mediated gene therapy for DMD in a subset of patients.

    Article  PubMed  CAS  Google Scholar 

  10. • Odom GL, et al. Gene therapy of mdx mice with large truncated dystrophins generated by recombination using rAAV6. Mol Ther. 2011;19:36–45. A dual vector strategy delivering a mini-dystrophin cassette containing the dystrophin C-terminus demonstrated increased functional benefit in mdx mice over microdystrophin.

    Article  PubMed  CAS  Google Scholar 

  11. Zhang Y, Duan D. Novel mini-dystrophin gene dual adeno-associated virus vectors restore neuronal nitric oxide synthase expression at the sarcolemma. Hum Gene Ther. 2012;23:98–103.

    Article  PubMed  Google Scholar 

  12. Li D, et al. Nitrosative stress elicited by nNOSmicro delocalization inhibits muscle force in dystrophin-null mice. J Pathol. 2011;223:88–98.

    Article  PubMed  CAS  Google Scholar 

  13. Krahn M, et al. A naturally occurring human minidysferlin protein repairs sarcolemmal lesions in a mouse model of dysferlinopathy. Sci Transl Med. 2010;2:50ra69.

    Article  PubMed  Google Scholar 

  14. Lostal W, et al. Efficient recovery of dysferlin deficiency by dual adeno-associated vector-mediated gene transfer. Hum Mol Genet 19:1897–907.

  15. Grose WE, et al. Homologous recombination mediates functional recovery of dysferlin deficiency following AAV5 gene transfer. PLoS One. 2012;7:e39233.

    Article  PubMed  CAS  Google Scholar 

  16. Mann CJ, et al. Antisense-induced exon skipping and synthesis of dystrophin in the mdx mouse. Proc Natl Acad Sci USA. 2001;98:42–7.

    Article  PubMed  CAS  Google Scholar 

  17. Goyenvalle A, et al. Prevention of dystrophic pathology in severely affected dystrophin/utrophin-deficient mice by morpholino-oligomer-mediated exon-skipping. Mol Ther. 2010;18:198–205.

    Article  PubMed  CAS  Google Scholar 

  18. Yokota T, et al. Efficacy of systemic morpholino exon-skipping in Duchenne dystrophy dogs. Ann Neurol. 2009;65:667–76.

    Article  PubMed  Google Scholar 

  19. van Deutekom JC, et al. Local dystrophin restoration with antisense oligonucleotide PRO051. N Engl J Med. 2007;357:2677–86.

    Article  PubMed  Google Scholar 

  20. Kinali M, 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;8:918–28.

    Article  PubMed  CAS  Google Scholar 

  21. Goemans NM, et al. Systemic administration of PRO051 in Duchenne’s muscular dystrophy. N Engl J Med. 2011;364:1513–22.

    Article  PubMed  CAS  Google Scholar 

  22. Cirak S, 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.

    Article  PubMed  CAS  Google Scholar 

  23. •• 17th International Congress of The World Muscle Society. Neuromuscul Disord 2012;22:771–922. Systemically delivered morpholino in DMD patients demonstrated clinical benefit.

    Google Scholar 

  24. Moulton HM, et al. Cellular uptake of antisense morpholino oligomers conjugated to arginine-rich peptides. Bioconjug Chem. 2004;15:290–9.

    Article  PubMed  CAS  Google Scholar 

  25. Yin H, et al. Functional rescue of dystrophin-deficient mdx mice by a chimeric peptide-PMO. Mol Ther. 2010;18:1822–9.

    Article  PubMed  CAS  Google Scholar 

  26. Malerba A, Boldrin L, Dickson G. Long-term systemic administration of unconjugated morpholino oligomers for therapeutic expression of dystrophin by exon skipping in skeletal muscle: implications for cardiac muscle integrity. Nucleic Acid Ther. 2011;21:293–8.

    PubMed  CAS  Google Scholar 

  27. Casazza F, et al. Cardiac transplantation in Becker muscular dystrophy. J Neurol. 1988;235:496–8.

    Article  PubMed  CAS  Google Scholar 

  28. Stollberger C, Finsterer J. Left ventricular synchronization by biventricular pacing in Becker muscular dystrophy as assessed by tissue Doppler imaging. Heart Lung. 2005;34:317–20.

    Article  PubMed  Google Scholar 

  29. Doing AH, Renlund DG, Smith RA. Becker muscular dystrophy-related cardiomyopathy: a favorable response to medical therapy. J Heart Lung Transplant. 2002;21:496–8.

    Article  PubMed  Google Scholar 

  30. Lai Y, Duan D. Progress in gene therapy of dystrophic heart disease. Gene Ther. 2012;19:678–85.

    Article  PubMed  CAS  Google Scholar 

  31. Rafael-Fortney JA, et al. Early treatment with lisinopril and spironolactone preserves cardiac and skeletal muscle in Duchenne muscular dystrophy mice. Circulation. 2011;124:582–8.

    Article  PubMed  CAS  Google Scholar 

  32. Mendell JR, et al. Diagnosis of Duchenne dystrophy by enhanced detection of small mutations. Neurology. 2001;57:645–50.

    Article  PubMed  CAS  Google Scholar 

  33. Barton-Davis ER, et al. Aminoglycoside antibiotics restore dystrophin function to skeletal muscles of mdx mice. J Clin Invest. 1999;104:375–81.

    Article  PubMed  CAS  Google Scholar 

  34. Welch EM, et al. PTC124 targets genetic disorders caused by nonsense mutations. Nature. 2007;447:87–91.

    Article  PubMed  CAS  Google Scholar 

  35. Hirawat S, et al. Safety, tolerability, and pharmacokinetics of PTC124, a nonaminoglycoside nonsense mutation suppressor, following single- and multiple-dose administration to healthy male and female adult volunteers. J Clin Pharmacol. 2007;47:430–44.

    Article  PubMed  CAS  Google Scholar 

  36. Clinical Trials. http://www.clinicaltrials.gov/ct2/results?term=ptc124.

  37. Kayali R, et al. Read-through compound 13 restores dystrophin expression and improves muscle function in the mdx mouse model for Duchenne muscular dystrophy. Hum Mol Genet. 2012;21:4007–20.

    Article  PubMed  CAS  Google Scholar 

  38. Tinsley J, et al. Expression of full-length utrophin prevents muscular dystrophy in mdx mice. Nat Med. 1998;4:1441–4.

    Article  PubMed  CAS  Google Scholar 

  39. Gilbert R, et al. Adenovirus-mediated utrophin gene transfer mitigates the dystrophic phenotype of mdx mouse muscles. Hum Gene Ther. 1999;10:1299–310.

    Article  PubMed  CAS  Google Scholar 

  40. Krag TO, et al. Heregulin ameliorates the dystrophic phenotype in mdx mice. Proc Natl Acad Sci USA. 2004;101:13856–60.

    Article  PubMed  CAS  Google Scholar 

  41. Moorwood C, et al. Drug discovery for Duchenne muscular dystrophy via utrophin promoter activation screening. PLoS One. 2011;6:e26169.

    Article  PubMed  CAS  Google Scholar 

  42. Tinsley JM, et al. Daily treatment with SMTC1100, a novel small molecule utrophin upregulator, dramatically reduces the dystrophic symptoms in the mdx mouse. PLoS One. 2011;6:e19189.

    Article  PubMed  CAS  Google Scholar 

  43. Hnia K, et al. L-arginine decreases inflammation and modulates the nuclear factor-kappaB/matrix metalloproteinase cascade in mdx muscle fibers. Am J Pathol. 2008;172:1509–19.

    Article  PubMed  CAS  Google Scholar 

  44. Goonasekera SA, et al. Mitigation of muscular dystrophy in mice by SERCA overexpression in skeletal muscle. J Clin Invest. 2011;121:1044–52.

    Article  PubMed  CAS  Google Scholar 

  45. Martin PT, et al. Overexpression of Galgt2 in skeletal muscle prevents injury resulting from eccentric contractions in both mdx and wild-type mice. Am J Physiol Cell Physiol. 2009;296:C476–88.

    Article  PubMed  CAS  Google Scholar 

  46. Burkin DJ, et al. Enhanced expression of the alpha 7 beta 1 integrin reduces muscular dystrophy and restores viability in dystrophic mice. J Cell Biol. 2001;152:1207–18.

    Article  PubMed  CAS  Google Scholar 

  47. Amenta AR, et al. Biglycan recruits utrophin to the sarcolemma and counters dystrophic pathology in mdx mice. Proc Natl Acad Sci USA. 2011;108:762–7.

    Article  PubMed  CAS  Google Scholar 

  48. Gehrig SM, et al. Hsp72 preserves muscle function and slows progression of severe muscular dystrophy. Nature. 2012;484:394–8.

    Article  PubMed  CAS  Google Scholar 

  49. Partridge TA, et al. Conversion of mdx myofibres from dystrophin-negative to -positive by injection of normal myoblasts. Nature. 1989;337:176–9.

    Article  PubMed  CAS  Google Scholar 

  50. Skuk D, et al. Dystrophin expression in myofibers of Duchenne muscular dystrophy patients following intramuscular injections of normal myogenic cells. Mol Ther. 2004;9:475–82.

    Article  PubMed  CAS  Google Scholar 

  51. Mendell JR, et al. Myoblast transfer in the treatment of Duchenne’s muscular dystrophy. N Engl J Med. 1995;333:832–8.

    Article  PubMed  CAS  Google Scholar 

  52. Asakura A, Rudnicki MA. Side population cells from diverse adult tissues are capable of in vitro hematopoietic differentiation. Exp Hematol. 2002;30:1339–45.

    Article  PubMed  Google Scholar 

  53. Buckingham M. Myogenic progenitor cells and skeletal myogenesis in vertebrates. Curr Opin Genet Dev. 2006;16:525–32.

    Article  PubMed  CAS  Google Scholar 

  54. Cossu G, Bianco P. Mesoangioblasts–vascular progenitors for extravascular mesodermal tissues. Curr Opin Genet Dev. 2003;13:537–42.

    Article  PubMed  CAS  Google Scholar 

  55. Dezawa M, et al. Bone marrow stromal cells generate muscle cells and repair muscle degeneration. Science. 2005;309:314–7.

    Article  PubMed  CAS  Google Scholar 

  56. Qu-Petersen Z, et al. Identification of a novel population of muscle stem cells in mice: potential for muscle regeneration. J Cell Biol. 2002;157:851–64.

    Article  PubMed  CAS  Google Scholar 

  57. Tagliafico E, et al. TGFbeta/BMP activate the smooth muscle/bone differentiation programs in mesoangioblasts. J Cell Sci. 2004;117:4377–88.

    Article  PubMed  CAS  Google Scholar 

  58. Sampaolesi M, et al. Mesoangioblast stem cells ameliorate muscle function in dystrophic dogs. Nature. 2006;444:574–9.

    Article  PubMed  CAS  Google Scholar 

  59. Davies KE, Grounds MD. Treating muscular dystrophy with stem cells? Cell. 2006;127:1304–6.

    Article  PubMed  CAS  Google Scholar 

  60. Tedesco FS, Cossu G. Stem cell therapies for muscle disorders. Curr Opin Neurol. 2012;25:597–603.

    Article  PubMed  Google Scholar 

  61. Tedesco FS, et al. Transplantation of genetically corrected human iPSC-derived progenitors in mice with limb-girdle muscular dystrophy. Sci Transl Med. 2012;4:140ra89.

    Article  PubMed  Google Scholar 

  62. Darabi R, et al. Human ES- and iPS-derived myogenic progenitors restore DYSTROPHIN and improve contractility upon transplantation in dystrophic mice. Cell Stem Cell. 2012;10:610–9.

    Article  PubMed  CAS  Google Scholar 

Download references

Disclosure

L.R. Rodino-Klapac: none; J.R. Mendell: principal investigator in the eteplirsen trial, for which he received no personal compensation; Z. Sahenk: none.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Louise R. Rodino-Klapac or Zarife Sahenk.

Additional information

This article is part of the Topical Collection on Nerve and Muscle

Rights and permissions

Reprints and permissions

About this article

Cite this article

Rodino-Klapac, L.R., Mendell, J.R. & Sahenk, Z. Update on the Treatment of Duchenne Muscular Dystrophy. Curr Neurol Neurosci Rep 13, 332 (2013). https://doi.org/10.1007/s11910-012-0332-1

Download citation

  • Published:

  • DOI: https://doi.org/10.1007/s11910-012-0332-1

Keywords

Navigation