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

Maria Kinali, Virginia Arechavala-Gomeza, Lucy Feng, Sebahattin Cirak, David Hunt, Carl Adkin, Michela Guglieri, Emma Ashton, Stephen Abbs, Petros Nihoyannopoulos, Maria Elena Garralda, Mary Rutherford, Caroline McCulley, Linda Popplewell, Ian R Graham, George Dickson, Matthew J A Wood, Dominic J Wells, Steve D Wilton, Ryszard Kole, Volker Straub, Kate Bushby, Caroline Sewry, Jennifer E Morgan, Francesco Muntoni, Maria Kinali, Virginia Arechavala-Gomeza, Lucy Feng, Sebahattin Cirak, David Hunt, Carl Adkin, Michela Guglieri, Emma Ashton, Stephen Abbs, Petros Nihoyannopoulos, Maria Elena Garralda, Mary Rutherford, Caroline McCulley, Linda Popplewell, Ian R Graham, George Dickson, Matthew J A Wood, Dominic J Wells, Steve D Wilton, Ryszard Kole, Volker Straub, Kate Bushby, Caroline Sewry, Jennifer E Morgan, Francesco Muntoni

Abstract

Background: Mutations that disrupt the open reading frame and prevent full translation of DMD, the gene that encodes dystrophin, underlie the fatal X-linked disease Duchenne muscular dystrophy. Oligonucleotides targeted to splicing elements (splice switching oligonucleotides) in DMD pre-mRNA can lead to exon skipping, restoration of the open reading frame, and the production of functional dystrophin in vitro and in vivo, which could benefit patients with this disorder.

Methods: We did a single-blind, placebo-controlled, dose-escalation study in patients with DMD recruited nationally, to assess the safety and biochemical efficacy of an intramuscular morpholino splice-switching oligonucleotide (AVI-4658) that skips exon 51 in dystrophin mRNA. Seven patients with Duchenne muscular dystrophy with deletions in the open reading frame of DMD that are responsive to exon 51 skipping were selected on the basis of the preservation of their extensor digitorum brevis (EDB) muscle seen on MRI and the response of cultured fibroblasts from a skin biopsy to AVI-4658. AVI-4658 was injected into the EDB muscle; the contralateral muscle received saline. Muscles were biopsied between 3 and 4 weeks after injection. The primary endpoint was the safety of AVI-4658 and the secondary endpoint was its biochemical efficacy. This trial is registered, number NCT00159250.

Findings: Two patients received 0.09 mg AVI-4658 in 900 microL (0.9%) saline and five patients received 0.9 mg AVI-4658 in 900 microL saline. No adverse events related to AVI-4658 administration were reported. Intramuscular injection of the higher-dose of AVI-4658 resulted in increased dystrophin expression in all treated EDB muscles, although the results of the immunostaining of EDB-treated muscle for dystrophin were not uniform. In the areas of the immunostained sections that were adjacent to the needle track through which AVI-4658 was given, 44-79% of myofibres had increased expression of dystrophin. In randomly chosen sections of treated EDB muscles, the mean intensity of dystrophin staining ranged from 22% to 32% of the mean intensity of dystrophin in healthy control muscles (mean 26.4%), and the mean intensity was 17% (range 11-21%) greater than the intensity in the contralateral saline-treated muscle (one-sample paired t test p=0.002). In the dystrophin-positive fibres, the intensity of dystrophin staining was up to 42% of that in healthy muscle. We showed expression of dystrophin at the expected molecular weight in the AVI-4658-treated muscle by immunoblot.

Interpretation: Intramuscular AVI-4658 was safe and induced the expression of dystrophin locally within treated muscles. This proof-of-concept study has led to an ongoing systemic clinical trial of AVI-4658 in patients with DMD.

Funding: UK Department of Health.

Figures

Figure 1
Figure 1
Deletions and predicted results of exon skipping in the patients who were studied (A) Pre-mRNA transcripts and dystrophin protein products from full length DMD, in patients with Duchenne muscular dystrophy, and predicted protein sequences after exon skipping. (I) The normal dystrophin gene produces the full length dystrophin product. (II) Patients 1 and 2 had a deletion in exon 50 that disrupts the open reading frame, leading to a truncated and unstable dystrophin. (III) Skipping of exon 51 restores the reading frame, producing a truncated but functional dystrophin that lacks exons 50 and 51. (IV) Patient 7 is missing exons 49 and 50. (V) Patients 3 and 4 are missing exons 48–50. (VI) Patients 5 and 6 are missing exons 45–50. All the truncated dystrophins produced after skipping of exon 51 are missing the hinge 3 region and some of the rod domain but have been associated with the milder BMD phenotype., (B) Structure of the phosphorodiamidate morpholino modification of the antisense oligomer.
Figure 2
Figure 2
Procedure for prescreening of patients before injection of AVI-4658. Patient 3 is shown as an example; similar results were obtained for all patients. (A) Transverse MRI of the lower leg and coronal MRI of the extensor digitorum brevis muscle (arrow) confirmed the suitability of the muscle. (B) Skin fibroblasts from all patients were forced into myogenic differentiation and treated with an AVI-4658 congener to confirm exon skipping and dystrophin production. RT-PCR analysis shows two bands: the high molecular weight band corresponds to the unskipped transcript (including exons 46, 47, 51, and 52) and the low molecular weight band corresponds to the transcript fragment with size specific skipping of exon 51. (C) Exon 51 skipping was confirmed by sequencing.
Figure 3
Figure 3
Dystrophin expression in patients treated with high-dose AVI-4658 Transverse sections of treated and contralateral EDB muscles that were immunostained for dystrophin with MANDYS106. (A) Low-power micrograph of a whole section taken with ×10 objective lens shows widespread expression of dystrophin in fibres from the treated muscle in patient 4. (B) Higher magnification (×20 objective lens) of dystrophin immunolabelling in treated and untreated sections in patients 3–7. Scale bars=100 μm.
Figure 4
Figure 4
Intensity of dystrophin expression in patients treated with high-dose AVI-4658 relative to control (A) Mean random intensity measurements. (B) Measurement of mean dystrophin intensity in positive fibres: intensity measurements exclusively targeted to 100 dystrophin-positive and 100 dystrophin-negative fibres within the same area in patient 4. Bars are SEM.
Figure 5
Figure 5
Exon 51 skipping in amplified RNA from treated muscles (A) RT-PCR analysis of RNA extracted from treated (X), untreated (O), and control (C) muscle sections detects shorter transcript fragments in the treated muscles, with sizes that correspond to the specific skipping of exon 51. (B) Exon 51 skipping was confirmed by sequencing. (C) Western blot analysis of homogenates of treated and untreated muscle (20×10 μm sections) and control muscle (2×10 μm sections [to avoid overexposure]) shows dystrophin expression in extracts from the control muscles (C) and treated (X) extensor digitorum brevis but not in the contralateral muscles (O). Loading was monitored with protogold. Low dose=0·09 mg AV-4658. High dose=0·9 mg AV-4658.

References

    1. Simonds AK, Muntoni F, Heather S, Fielding S. Impact of nasal ventilation on survival in hypercapnic Duchenne muscular dystrophy. Thorax. 1998;53:949–952.
    1. Eagle M, Baudouin SV, Chandler C, Giddings DR, Bullock R, Bushby K. Survival in Duchenne muscular dystrophy: improvements in life expectancy since 1967 and the impact of home nocturnal ventilation. Neuromuscul Disord. 2002;12:926–929.
    1. Hoffman EP, Brown RH, Jr, Kunkel LM. Dystrophin: the protein product of the Duchenne muscular dystrophy locus. Cell. 1987;51:919–928.
    1. Morris GE, Ellis JM, Nguyen TM. A quantitative ELISA for dystrophin. J Immunol Methods. 1993;161:23–28.
    1. Koenig M, Beggs AH, Moyer M. The molecular basis for Duchenne versus Becker muscular dystrophy: correlation of severity with type of deletion. Am J Hum Genet. 1989;45:498–506.
    1. Bushby KM, Gardner-Medwin D, Nicholson LV. The clinical, genetic and dystrophin characteristics of Becker muscular dystrophy. II Correlation of phenotype with genetic and protein abnormalities. J Neurol. 1993;240:105–112.
    1. Klein CJ, Coovert DD, Bulman DE, Ray PN, Mendell JR, Burghes AH. Somatic reversion/suppression in Duchenne muscular dystrophy (DMD): evidence supporting a frame-restoring mechanism in rare dystrophin-positive fibers. Am J Hum Genet. 1992;50:950–959.
    1. Lu QL, Morris GE, Wilton SD. Massive idiosyncratic exon skipping corrects the nonsense mutation in dystrophic mouse muscle and produces functional revertant fibers by clonal expansion. J Cell Biol. 2000;148:985–996.
    1. Melis MA, Cau M, Muntoni F. Elevation of serum creatine kinase as the only manifestation of an intragenic deletion of the dystrophin gene in three unrelated families. Eur J Paediatr Neurol. 1998;2:255–261.
    1. Lesca G, Testard H, Streichenberger N. Family study allows more optimistic prognosis and genetic counselling in a child with a deletion of exons 50–51 of the dystrophin gene. Arch Pediatr. 2007;14:262–265.
    1. Muntoni F, Torelli S, Ferlini A. Dystrophin and mutations: one gene, several proteins, multiple phenotypes. Lancet Neurol. 2003;2:731–740.
    1. Aartsma-Rus A, Fokkema I, Verschuuren J. Theoretic applicability of antisense-mediated exon skipping for Duchenne muscular dystrophy mutations. Hum Mutat. 2009;30:293–299.
    1. Sazani P, Graziewicz MA, Kole R. In: Antisense drug technology: principles, strategies, and applications. Crooke ST, editor. Taylor and Francis; London: 2008. Splice switching oligonucleotides as potential therapeutics; pp. 89–114.
    1. Aartsma-Rus A, Janson AA, Kaman WE. Therapeutic antisense-induced exon skipping in cultured muscle cells from six different DMD patients. Hum Mol Genet. 2003;12:907–914.
    1. Aartsma-Rus A, van Ommen GJ. Antisense-mediated exon skipping: a versatile tool with therapeutic and research applications. RNA. 2007;13:1609–1624.
    1. McClorey G, Moulton HM, Iversen PL, Fletcher S, Wilton SD. Antisense oligonucleotide-induced exon skipping restores dystrophin expression in vitro in a canine model of DMD. Gene Ther. 2006;13:1373–1381.
    1. Lu QL, Mann CJ, Lou F. Functional amounts of dystrophin produced by skipping the mutated exon in the mdx dystrophic mouse. Nat Med. 2003;9:1009–1014.
    1. Yokota T, Lu QL, Partridge T. Efficacy of systemic morpholino exon-skipping in duchenne dystrophy dogs. Ann Neurol. 2009;65:667–676.
    1. Takeshima Y, Yagi M, Wada H. Intravenous infusion of an antisense oligonucleotide results in exon skipping in muscle dystrophin mRNA of Duchenne muscular dystrophy. Pediatr Res. 2006;59:690–694.
    1. van Deutekom JC, Janson AA, Ginjaar IB. Local dystrophin restoration with antisense oligonucleotide PRO051. N Engl J Med. 2007;357:2677–2686.
    1. Sazani P, Gemignani F, Kang SH. Systemically delivered antisense oligomers upregulate gene expression in mouse tissues. Nat Biotechnol. 2002;20:1228–1233.
    1. Fletcher S, Honeyman K, Fall AM, Harding PL, Johnsen RD, Wilton SD. Dystrophin expression in the mdx mouse after localised and systemic administration of a morpholino antisense oligonucleotide. J Gene Med. 2006;8:207–216.
    1. Alter J, Lou F, Rabinowitz A. Systemic delivery of morpholino oligonucleotide restores dystrophin expression bodywide and improves dystrophic pathology. Nat Med. 2006;12:175–177.
    1. Heemskerk HA, de Winter CL, de Kimpe SJ. In vivo comparison of 2′-O-methyl phosphorothioate and morpholino antisense oligonucleotides for Duchenne muscular dystrophy exon skipping. J Gene Med. 2009;11:257–266.
    1. Muntoni F, Bushby KD, van Ommen G. 149th ENMC International Workshop and 1st TREAT-NMD Workshop on: planning phase I/III clinical trials using systemically delivered antisense oligonucleotides in Duchenne muscular dystrophy. Neuromuscul Disord. 2008;18:268–275.
    1. Emery AE, Muntoni F. Duchenne muscular dystrophy. 3rd edn. Oxford University Press; Oxford: 2003.
    1. Mercuri E, Cini C, Counsell S. Muscle MRI findings in a three-generation family affected by Bethlem myopathy. Eur J Paediatr Neurol. 2002;6:309–314.
    1. Hawley RJ, Jr, Schellinger D, O'Doherty DS. Computed tomographic patterns of muscles in neuromuscular diseases. Arch Neurol. 1984;41:383–387.
    1. Dubowitz V, Sewry C. Muscle biopsy: a practical approach. 3rd edn. Saunders Elsevier; Netherlands: 2007.
    1. Macalister A. Additional observation on muscular anomalies in human anatomy (third series), with a catalogue of the principal muscular variations hitherto published. Trans Roy Irish Acad Sci. 1875;25:1–134.
    1. Mercuri E, Pichiecchio A, Allsop J, Messina S, Pane M, Muntoni F. Muscle MRI in inherited neuromuscular disorders: past, present, and future. J Magn Reson Imaging. 2007;25:433–440.
    1. Mercuri E, Pichiecchio A, Counsell S. A short protocol for muscle MRI in children with muscular dystrophies. Eur J Paediatr Neurol. 2002;6:305–307.
    1. Bailey D, Garralda ME. The use of social stress and support interview in families with deviant children: methodological issues. Social Psychiatry. 1987;22:209–215.
    1. Goodman R, Ford T, Simmons H, Gatward R, Meltzer H. Using the Strengths and Difficulties questionnaire to screen for child psychiatric disorders. Br J Psychiatry. 2000;177:534–539.
    1. Goldberg D. Manual of the General Health Questionnaire. NFER Publishing Company; Wiltshire: UK: 1978.
    1. Miller IW, Epstein NB, Bishop DS, Keitner GI. The McMaster Family Assessment Device: Reliability and Validity. J Marital Fam Ther. 1985;11:345–356.
    1. White D, Leach C, Atkinson M, Cottrell D. Validation of the Hospital Anxiety and Depression Scale for use with adolescents. Br J Psychiatry. 1999;175:452–454.
    1. Roest PA, van der Tuijn AC, Ginjaar HB. Application of in vitro Myo-differentiation of non-muscle cells to enhance gene expression and facilitate analysis of muscle proteins. Neuromuscul Disord. 1996;6:195–202.
    1. Arechavala-Gomeza V, Graham IR, Popplewell LJ. Comparative analysis of antisense oligonucleotide sequences for targeted skipping of exon 51 during dystrophin pre-mRNA splicing in human muscle. Hum Gene Ther. 2007;18:798–810.
    1. Ferrer A, Foster H, Wells KE, Dickson G, Wells DJ. Long-term expression of full-length human dystrophin in transgenic mdx mice expressing internally deleted human dystrophins. Gene Ther. 2004;11:884–893.
    1. Nguyen TM, Morris GE. Use of epitope libraries to identify exon-specific monoclonal antibodies for characterization of altered dystrophins in muscular dystrophy. Am J Hum Genet. 1993;52:1057–1066.
    1. Neri M, Torelli S, Brown S. Dystrophin levels as low as 30% are sufficient to avoid muscular dystrophy in the human. Neuromuscul Disord. 2007;17:913–918.

Source: PubMed

3
Iratkozz fel