Increased dystrophin production with golodirsen in patients with Duchenne muscular dystrophy

Diane E Frank, Frederick J Schnell, Cody Akana, Saleh H El-Husayni, Cody A Desjardins, Jennifer Morgan, Jay S Charleston, Valentina Sardone, Joana Domingos, George Dickson, Volker Straub, Michela Guglieri, Eugenio Mercuri, Laurent Servais, Francesco Muntoni, SKIP-NMD Study Group, Christopher Clark, Rahul Phadke, Thomas Voit, Pierre G. Carlier, Teresa Gidaro, Andreea Mihaela Seferian, Elena Gargaun, Linda Popplewell, Adam Jones, Lucy Feng, Caroline Sewry, Daniela Leone, Maria Carmela Pera, Mauro Monforte, Marika Pane, Shande Tang, Diane E Frank, Frederick J Schnell, Cody Akana, Saleh H El-Husayni, Cody A Desjardins, Jennifer Morgan, Jay S Charleston, Valentina Sardone, Joana Domingos, George Dickson, Volker Straub, Michela Guglieri, Eugenio Mercuri, Laurent Servais, Francesco Muntoni, SKIP-NMD Study Group, Christopher Clark, Rahul Phadke, Thomas Voit, Pierre G. Carlier, Teresa Gidaro, Andreea Mihaela Seferian, Elena Gargaun, Linda Popplewell, Adam Jones, Lucy Feng, Caroline Sewry, Daniela Leone, Maria Carmela Pera, Mauro Monforte, Marika Pane, Shande Tang

Abstract

Objective: To report safety, pharmacokinetics, exon 53 skipping, and dystrophin expression in golodirsen-treated patients with Duchenne muscular dystrophy (DMD) amenable to exon 53 skipping.

Methods: Part 1 was a randomized, double-blind, placebo-controlled, 12-week dose titration of once-weekly golodirsen; part 2 is an ongoing, open-label evaluation. Safety and pharmacokinetics were primary and secondary objectives of part 1. Primary biological outcome measures of part 2 were blinded exon skipping and dystrophin protein production on muscle biopsies (baseline, week 48) evaluated, respectively, using reverse transcription PCR and Western blot and immunohistochemistry.

Results: Twelve patients were randomized to receive golodirsen (n = 8) or placebo (n = 4) in part 1. All from part 1 plus 13 additional patients received 30 mg/kg golodirsen in part 2. Safety findings were consistent with those previously observed in pediatric patients with DMD. Most of the study drug was excreted within 4 hours following administration. A significant increase in exon 53 skipping was associated with ∼16-fold increase over baseline in dystrophin protein expression at week 48, with a mean percent normal dystrophin protein standard of 1.019% (range, 0.09%-4.30%). Sarcolemmal localization of dystrophin was demonstrated by significantly increased dystrophin-positive fibers (week 48, p < 0.001) and a positive correlation (Spearman r = 0.663; p < 0.001) with dystrophin protein change from baseline, measured by Western blot and immunohistochemistry.

Conclusion: Golodirsen was well-tolerated; muscle biopsies from golodirsen-treated patients showed increased exon 53 skipping, dystrophin production, and correct dystrophin sarcolemmal localization.

Clinicaltrialsgov identifier: NCT02310906.

Classification of evidence: This study provides Class I evidence that golodirsen is safe and Class IV evidence that it induces exon skipping and novel dystrophin as confirmed by 3 different assays.

Copyright © 2020 The Author(s). Published by Wolters Kluwer Health, Inc. on behalf of the American Academy of Neurology.

Figures

Figure 1. Golodirsen study details
Figure 1. Golodirsen study details
(A) Study design. (B) Study flow diagram (part 1). 6MWT = 6-minute walk test; DSMB = data safety monitoring board; PDPF = percentage dystrophin-positive fibers.
Figure 2. Treatment with golodirsen demonstrated an…
Figure 2. Treatment with golodirsen demonstrated an increase in de novo dystrophin protein
(A) Western blot examples performed with baseline (labeled as BL) and on treatment (Tx) paired samples from 4 patients. Numbers above Tx gel lanes indicate the percent normal control dystrophin, calculated from the standard curve on each blot (lanes denoted with 4%, 2%, 1%, 0.5%, 0.25%). In both panels, lane 0 represents baseline untreated DMD control tissue with no normal control lysate. Arrows and values above gel indicate the percent normal dystrophin measured for the indicated lane. (B) Western blot data shown as averaged dystrophin percent of normal for each individual patient at baseline (open circles) and at part 2, week 48, (solid squares), with dystrophin levels on y-axis presented as linear values. *Sample reading was above the upper limit of quantification.
Figure 3. Exon 53 skipping and dystrophin
Figure 3. Exon 53 skipping and dystrophin
(A) Treatment with golodirsen demonstrates an increase in skipping of exon 53 in dystrophin mRNA. Percent exon 53 skipping for individual patients at baseline (open circles) and on treatment (filled squares). Data represent mean ± SD values for 4–8 replicates. (B) Positive correlation between percent exon 53 skipping and production of dystrophin protein. Percent exon 53 skipping change from baseline was plotted against the change in dystrophin protein from baseline, as measured by Western blot (Spearman r correlation coefficient: 0.500; p = 0.011).
Figure 4. Dystrophin expression, baseline and treated…
Figure 4. Dystrophin expression, baseline and treated part 2, week 48, biopsy samples from the same patient
(A) Example 1: Low magnification image of whole tissue section stained for laminin α2 (green, all fibers stained) and dystrophin (red). A total of 2,843 muscle fibers were present in the baseline whole-slide image (A.a) and 1,077 fibers in the part 2, week 48, image (A.b). Three highlighted regions from the whole tissue sections (green) are magnified below each image to illustrate the corresponding dystrophin staining in these regions (red). Image intensity was not enhanced and represents original captured images. Dystrophin protein at part 2, week 48, was 4.3% of normal, as measured by Western blot for this patient. (B) Example 2: Low magnification image of whole tissue section stained for laminin α2 (green, all fibers stained) and dystrophin (red). A total of 4,945 muscle fibers were present in the baseline whole-slide image (B.a) and 2,167 fibers in the part 2, week 48, image (B.b). Three highlighted regions from the whole tissue sections (green) are magnified below each image to highlight the corresponding dystrophin staining in these regions (red). Image intensity was not enhanced and represents original captured images. Dystrophin protein at part 2, week 48, was 1.91% of normal, as measured by Western blot for this patient.
Figure 5. Manual percentage dystrophin-positive fiber (PDPF)…
Figure 5. Manual percentage dystrophin-positive fiber (PDPF) analysis
(A) Baseline values of PDPF for each patient are shown as open circles and part 2, week 48, intensity is shown as solid squares. This graphical representation provides a ready way to visualize relative changes in each patient. Data represent the mean of 3–4 replicates with SD bars. (B) Western blot protein change from baseline is plotted on the x-axis and percent dystrophin-positive fiber change from baseline as assessed by manual scoring is plotted on the y-axis. Spearman analysis shows a significant correlation (p < 0.001; r = 0.663). Each point represents a single patient.

References

    1. 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.
    1. Emery AE. Population frequencies of inherited neuromuscular diseases: a world survey. Neuromuscul Disord 1991;1:19–29.
    1. Mendell JR, Shilling C, Leslie ND, et al. . Evidence-based path to newborn screening for Duchenne muscular dystrophy. Ann Neurol 2012;71:304–313.
    1. Mah JK, Korngut L, Dykeman J, Day L, Pringsheim T, Jette N. A systematic review and meta-analysis on the epidemiology of Duchenne and Becker muscular dystrophy. Neuromuscul Disord 2014;24:482–491.
    1. Kobayashi YM, Campbell KP. Skeletal muscle dystrophin-glycoprotein complex and muscular dystrophy. In: Hill JA, Olson EN, eds. Muscle: Fundamental Biology and Mechanisms of Disease, vol. 2. New York: Academic Press; 2012:935–942.
    1. Emery AE. The muscular dystrophies. Lancet 2002;359:687–695.
    1. Monaco AP, Bertelson CJ, Liechti-Gallati S, Moser H, Kunkel LM. An explanation for the phenotypic differences between patients bearing partial deletions of the DMD locus. Genomics 1988;2:90–95.
    1. Allen DG, Whitehead NP, Froehner SC. Absence of dystrophin disrupts skeletal muscle signaling: roles of Ca2+, reactive oxygen species, and nitric oxide in the development of muscular dystrophy. Physiol Rev 2016;96:253–305.
    1. Rando TA. The dystrophin-glycoprotein complex, cellular signaling, and the regulation of cell survival in the muscular dystrophies. Muscle Nerve 2001;24:1575–1594.
    1. Mah JK. Current and emerging treatment strategies for Duchenne muscular dystrophy. Neuropsychiatr Dis Treat 2016;12:1795–1807.
    1. Falzarano MS, Scotton C, Passarelli C, Ferlini A. Duchenne muscular dystrophy: from diagnosis to therapy. Molecules 2015;20:18168–18184.
    1. Wilton SD, Fall AM, Harding PL, McClorey G, Coleman C, Fletcher S. Antisense oligonucleotide-induced exon skipping across the human dystrophin gene transcript. Mol Ther 2007;15:1288–1296.
    1. Wilton SD, Fletcher S. RNA splicing manipulation: strategies to modify gene expression for a variety of therapeutic outcomes. Curr Gene Ther 2011;11:259–275.
    1. Wilton SD, Lloyd F, Carville K, et al. . Specific removal of the nonsense mutation from the mdx dystrophin mRNA using antisense oligonucleotides. Neuromuscul Disord 1999;9:330–338.
    1. Popplewell LJ, Trollet C, Dickson G, Graham IR. Design of phosphorodiamidate morpholino oligomers (PMOs) for the induction of exon skipping of the human DMD gene. Mol Ther 2009;17:554–561.
    1. Exondys 51 [package insert]. Cambridge: Sarepta Therapeutics, Inc.; 2016.
    1. 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.
    1. 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.
    1. 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.
    1. Frank D, Dworzak J, Lawlor M, et al. . Optimization and implementation of best practices for collection and preparation of muscle biopsies for analysis during clinical trials of neuromuscular disease therapeutics [poster]. International Annual Congress of the World Muscle Society; October 3–7, 2017; Saint Malo, France.
    1. Charleston JS, Schnell FJ, Dworzak J, et al. . Eteplirsen treatment for Duchenne muscular dystrophy: exon skipping and dystrophin production. Neurology 2018;90:e2146–e2154.
    1. Guidance for Industry. Duchenne muscular dystrophy and related dystrophinopathies: developing drugs for treatment. 2018. Available at: . Accessed February 5, 2019.
    1. Mendell JR, Rodino-Klapac LR, Sahenk Z, et al. . Eteplirsen for the treatment of Duchenne muscular dystrophy. Ann Neurol 2013;74:637–647.
    1. Alter J, Lou F, Rabinowitz A, et al. . Systemic delivery of morpholino oligonucleotide restores dystrophin expression bodywide and improves dystrophic pathology. Nat Med 2006;12:175–177.
    1. Yokota T, Lu QL, Partridge T, et al. . Efficacy of systemic morpholino exon-skipping in Duchenne dystrophy dogs. Ann Neurol 2009;65:667–676.
    1. van Putten M, Hulsker M, Young C, et al. . Low dystrophin levels increase survival and improve muscle pathology and function in dystrophin/utrophin double-knockout mice. FASEB J 2013;27:2484–2495.
    1. Anthony K, Arechavala-Gomeza V, Ricotti V, et al. . Biochemical characterization of patients with in-frame or out-of-frame DMD deletions pertinent to exon 44 or 45 skipping. JAMA Neurol 2014;71:32–40.
    1. Bello L, Morgenroth LP, Gordish-Dressman H, Hoffman EP, McDonald CM, Cirak S. DMD genotypes and loss of ambulation in the CINRG Duchenne natural history study. Neurology 2016;87:401–409.
    1. Moon D, Hu S, Bange J, et al. . Genotype-phenotype associations in a large cohort of Duchenne muscular dystrophy patients. Neuromuscul Disord 2017;27(suppl 2):S104–S105.
    1. Waldrop MA, Gumienny F, El Husayni S, Frank DE, Weiss RB, Flanigan KM. Low-level dystrophin expression attenuating the dystrophinopathy phenotype. Neuromuscul Disord 2018;28:116–121.
    1. Brogna C, Coratti G, Pane M, et al. ; on behalf on the International DMD Group. Long-term natural history data in Duchenne muscular dystrophy ambulant patients with mutations amenable to skip exons 44, 45, 51 and 53. PLoS One 2019;14:e0218683.
    1. Khan N, Eliopoulos H, Han L, et al. . Eteplirsen treatment attenuates respiratory decline in ambulatory and non-ambulatory patients with Duchenne muscular dystrophy. J Neuromuscul Dis 2019;6:213–225.
    1. Godfrey C, Muses S, McClorey G, et al. . How much dystrophin is enough: the physiological consequences of different levels of dystrophin in the mdx mouse. Hum Mol Genet 2015;24:4225–4237.
    1. Bello L, Flanigan KM, Weiss RB, et al. . Association study of exon variants in the NF-κB and TGFβ pathways identifies CD40 as a modifier of Duchenne muscular dystrophy. Am J Hum Genet 2016;99:1163–1171.
    1. Flanigan KM, Ceco E, Lamar KM, et al. . LTBP4 genotype predicts age of ambulatory loss in Duchenne muscular dystrophy. Ann Neurol 2013;73:481–488.
    1. European Medicines Agency. Guideline on the clinical investigation of medicinal products for the treatment of Duchenne and Becker muscular dystrophy. Available at: . Accessed November 5, 2018.

Source: PubMed

3
Subscribe