Ataluren delays loss of ambulation and respiratory decline in nonsense mutation Duchenne muscular dystrophy patients

Craig M McDonald, Francesco Muntoni, Vinay Penematsa, Joel Jiang, Allan Kristensen, Francesco Bibbiani, Elizabeth Goodwin, Heather Gordish-Dressman, Lauren Morgenroth, Christian Werner, James Li, Richard Able, Panayiota Trifillis, Már Tulinius, Study 019 investigators, M Ryan, K Jones, N Goemans, C Campbell, J K Mah, K Selby, B Chabrol, Y Pereon, T Voit, T Gidaro, U Schara, J B Kirschner, Y Nevo, G P Comi, E Bertini, E Mercuri, J Colomer, A Nascimento, J J Vilchez, M Tulinius, T Sejersen, F Muntoni, K Bushby, M Guglieri, Craig M McDonald, Francesco Muntoni, Vinay Penematsa, Joel Jiang, Allan Kristensen, Francesco Bibbiani, Elizabeth Goodwin, Heather Gordish-Dressman, Lauren Morgenroth, Christian Werner, James Li, Richard Able, Panayiota Trifillis, Már Tulinius, Study 019 investigators, M Ryan, K Jones, N Goemans, C Campbell, J K Mah, K Selby, B Chabrol, Y Pereon, T Voit, T Gidaro, U Schara, J B Kirschner, Y Nevo, G P Comi, E Bertini, E Mercuri, J Colomer, A Nascimento, J J Vilchez, M Tulinius, T Sejersen, F Muntoni, K Bushby, M Guglieri

Abstract

Aim: We investigated the effect of ataluren plus standard of care (SoC) on age at loss of ambulation (LoA) and respiratory decline in patients with nonsense mutation Duchenne muscular dystrophy (nmDMD) versus patients with DMD on SoC alone. Patients & methods: Study 019 was a long-term Phase III study of ataluren safety in nmDMD patients with a history of ataluren exposure. Propensity score matching identified Study 019 and CINRG DNHS patients similar in disease progression predictors. Results & conclusion: Ataluren plus SoC was associated with a 2.2-year delay in age at LoA (p = 0.0006), and a 3.0-year delay in decline of predicted forced vital capacity to <60% in nonambulatory patients (p = 0.0004), versus SoC. Ataluren plus SoC delays disease progression and benefits ambulatory and nonambulatory patients with nmDMD. ClinicalTrials.gov registration: NCT01557400.

Keywords: Study 019; ataluren; dystrophin; efficacy; loss of ambulation; nonsense mutation Duchenne muscular dystrophy; respiratory function.

Conflict of interest statement

Financial & competing interests disclosure

This work was supported by PTC Therapeutics. Study 019 was sponsored by PTC Therapeutics. The CINRG DNHS was funded by grants from the US Department of Education/NIDRR (#H133B031118, #H133B090001), the US Department of Defense (#W81XWH-09-1-0592), the National Institutes of Health (#UL1RR031988, #U54HD053177, #UL1RR024992, #U54RR026139, #2U54HD053177, #G12RR003051, #1R01AR061875, #RO1AR062380) and Parent Project Muscular Dystrophy. CM McDonald has acted as a consultant on clinical trials of DMD for Astellas, BioMarin, Capricor Therapeutics, Catabasis Pharmaceuticals, Edgewise Therapeutics, Eli Lilly and company, Epirium Bio (formerly Cardero Therapeutics), FibroGen, Gilead, Hoffmann-La Roche, Italfarmaco, Pfizer, PTC Therapeutics, Santhera Pharmaceuticals and Sarepta Therapeutics, and has received research support for clinical trials from BioMarin, Capricor Therapeutics, Eli Lilly and company, Italfarmaco, Pfizer, PTC Therapeutics, Hoffmann-La Roche, Santhera Pharmaceuticals and Sarepta Therapeutics. F Muntoni has received consultancy fees from AveXis, Biogen, Capricor Therapeutics, Catabasis Pharmaceuticals, Dyne Therapeutics, Novartis, Pfizer, PTC Therapeutics, Roche, Santhera Pharmaceuticals, Sarepta Therapeutics and Wave Therapeutics, and is supported by the National Institute for Health Research Biomedical Research Centre at Great Ormond Street Hospital for Children National Health Service Foundation Trust and University College London. V Penematsa, J Jiang, A Kristensen, E Goodwin, C Werner, James Li, R Able and P Trifillis are employees of PTC Therapeutics. F Bibbiani is a former employee of PTC Therapeutics. H Gordish-Dressman has served as a consultant for AGADA Biosciences, Audentes Therapeutics, ReveraGen BioPharma and Solid GT, and is a cofounder and part owner of TRiNDS. L Morgenroth reports no disclosures relevant to this manuscript. M Tulinius has received lecture fees from Biogen and PTC Therapeutics, has acted as a consultant on DMD clinical trials for BioMarin, Catabasis Pharmaceuticals, PTC Therapeutics, ReveraGen BioPharma and Sarepta Therapeutics, and as an advisory board member for AveXis, Biogen, PTC Therapeutics and Sarepta Therapeutics. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

Medical writing and editorial support were provided by E Colbeck, PhD, an employee of PharmaGenesis London, London, UK, and were funded by PTC Therapeutics.

Figures

Figure 1.. The different study populations used…
Figure 1.. The different study populations used in the analyses presented herein.
aThe as-treated population included patients with nmDMD who received at least one dose of ataluren during Study 019. bThis patient had not participated in a previous trial involving ataluren and was included in Study 019 due to a successful petition. cThe four covariates used for propensity score matching were: age at first clinical symptoms; age at initiation of corticosteroids; duration of deflazacort use (<1 month, ≥1 month to <12 months, and ≥12 months); and duration of other corticosteroid use (<1 month, ≥1 month to <12 months and ≥12 months). dThe four respiratory outcome measures assessed were: predicted FVC <60%; predicted FVC <50%; predicted FVC <30%; and absolute FVC <1 l. These respiratory outcome measures were only performed in nonambulatory patients. eStudy 007; Study 007e; Study 004; Study 004e. fThe 33 patients comprised patients who (a) received placebo in Study 007 and ataluren 80 mg/kg/day in Study 007e, (b) received ataluren 80 mg/kg/day in Study 007 and ataluren 80 mg/kg/day in Study 007e, or (c) received ataluren 80 mg/kg/day in Study 004 and ataluren 80 mg/kg/day in Study 004e. FVC: Forced vital capacity; LoA: Loss of ambulation; nmDMD: Nonsense mutation Duchenne muscular dystrophy; SoC: Standard of care.
Figure 2.. Age at loss of ambulation…
Figure 2.. Age at loss of ambulation for Study 019 patients with nonsense mutation Duchenne muscular dystrophy who received ataluren 40 mg/kg/day plus standard of care in at least Study 019† compared with propensity-score matched patients with Duchenne muscular dystrophy receiving standard of care alone in the CINRG DNHS (each n = 60).
†The 60 patients in this analysis are comprised of 27 patients who received 40 mg/kg/day ataluren plus standard of care (SoC) in prior ataluren studies and 33 patients who received placebo and then ataluren 80 mg/kg/day or only ataluren 80 mg/kg/day plus SoC in prior clinical trials, and began treatment with ataluren 40 mg/kg/day plus SoC upon entry to Study 019, and were ambulatory at entry to Study 019.
Figure 3.. Age at (A) predicted forced…
Figure 3.. Age at (A) predicted forced vital capacity
FVC: Forced vital capacity.

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. 9(1), 77–93 (2010a).
    1. Ellis JA, Vroom E, Muntoni F. 195th ENMC International Workshop: newborn screening for Duchenne muscular dystrophy 14–16th December, 2012, Naarden, The Netherlands. Neuromusc. Disord. 23(8), 682–689 (2013).
    1. Duan D, Goemans N, Takeda S, Mercuri E, Aartsma-Rus A. Duchenne muscular dystrophy. Nat. Rev. Dis. Primers 7(1), 13 (2021).
    1. Ryder S, Leadley RM, Armstrong N et al. The burden, epidemiology, costs and treatment for Duchenne muscular dystrophy: an evidence review. Orphanet J. Rare Dis. 12(1), 79 (2017).
    1. Landfeldt E, Thompson R, Sejersen T, McMillan HJ, Kirschner J, Lochmuller H. Life expectancy at birth in Duchenne muscular dystrophy: a systematic review and meta-analysis. Eur. J. Epidemiol. 35(7), 643–653 (2020).
    1. Passamano L, Taglia A, Palladino A et al. Improvement of survival in Duchenne muscular dystrophy: retrospective analysis of 835 patients. Acta Myol. 31(2), 121–125 (2012).
    1. Rall S, Grimm T. Survival in Duchenne muscular dystrophy. Acta Myol. 31(2), 117–120 (2012).
    1. Kenneson A, Bobo JK. The effect of caregiving on women in families with Duchenne/Becker muscular dystrophy. Health Social Care Commun. 18(5), 520–528 (2010).
    1. Humbertclaude V, Hamroun D, Bezzou K et al. Motor and respiratory heterogeneity in Duchenne patients: implication for clinical trials. Eur. J. Paediatr. Neurol. 16(2), 149–160 (2012).
    1. Birnkrant DJ, Bushby K, Bann CM et al. Diagnosis and management of Duchenne muscular dystrophy, part 2: respiratory, cardiac, bone health, and orthopaedic management. Lancet Neurol. 17(4), 347–361 (2018).
    1. Bushby K, Finkel R, Birnkrant DJ et al. Diagnosis and management of Duchenne muscular dystrophy, part 2: implementation of multidisciplinary care. Lancet Neurol. 9(2), 177–189 (2010).
    1. McDonald CM, Gordish-Dressman H, Henricson EK et al. Longitudinal pulmonary function testing outcome measures in Duchenne muscular dystrophy: long-term natural history with and without glucocorticoids. Neuromusc. Disord. 28(11), 897–909 (2018).
    2. •• Describes results from an analysis of changes in pulmonary function measures across time in patients with DMD treated with corticosteroids for more than 1 year compared with corticosteroid-naive patients in the CINRG DNHS.

    1. Phillips MF, Quinlivan RC, Edwards RH, Calverley PM. Changes in spirometry over time as a prognostic marker in patients with Duchenne muscular dystrophy. Am. J. Respir. Crit. Care Med. 164(12), 2191–2194 (2001).
    1. 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. 36(4), 395–402 (2015).
    1. Mah JK. Current and emerging treatment strategies for Duchenne muscular dystrophy. Neuropsychiatr. Dis. Treat. 12, 1795–1807 (2016).
    1. Gao QQ, McNally EM. The dystrophin complex: structure, function, and implications for therapy. Comp. Physiol. 5(3), 1223–1239 (2015).
    1. PTC Therapeutics International Ltd. Summary of product characteristics, Translarna. European Medicines Agency, 2018. Available from: (2020).
    1. Welch EM, Barton ER, Zhuo J et al. PTC124 targets genetic disorders caused by nonsense mutations. Nature 447(7140), 87–91 (2007).
    1. Bushby K, Finkel R, Wong B et al. Ataluren treatment of patients with nonsense mutation dystrophinopathy. Muscle Nerve 50(4), 477–487 (2014).
    2. •• Describes the safety and efficacy results of ataluren in a Phase IIb randomized, double-blind, placebo-controlled trial.

    1. McDonald CM, Campbell C, Torricelli RE et al. Ataluren in patients with nonsense mutation Duchenne muscular dystrophy (ACT DMD): a multicentre, randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 390(10101), 1489–1498 (2017a).
    2. •• Describes the safety and efficacy results of ataluren in a Phase III randomized, double-blind, placebo-controlled trial.

    1. Campbell C, Barohn RJ, Bertini E et al. Meta-analyses of ataluren randomized controlled trials in nonsense mutation Duchenne muscular dystrophy. J. Comp. Eff. Res. 9(14), 973–984 (2020).
    2. •• A meta-analyses of the 6-minute walk test data from the Phase IIb and Phase III clinical studies of ataluren.

    1. Li D, McDonald CM, Elfring GL et al. Assessment of treatment effect with multiple outcomes in 2 clinical trials of patients with duchenne muscular dystrophy. JAMA Netw. Open 3(2), e1921306 (2020).
    2. •• A totality of evidence analysis of data from the Phase IIb and Phase III clinical studies of ataluren.

    1. Mercuri E, Muntoni F, Osorio AN et al. Safety and effectiveness of ataluren: comparison of results from the STRIDE Registry and CINRG DMD Natural History Study. J. Comp. Eff. Res. 9(5), 341–360 (2020).
    2. •• Describes long-term safety and effectiveness data on ataluren from the ongoing, observational STRIDE Registry.

    1. Muntoni F, Desguerre I, Guglieri M et al. Ataluren use in patients with nonsense mutation Duchenne muscular dystrophy: patient demographics and characteristics from the STRIDE Registry. J. Comp. Eff. Res. 8(14), 1187–1200 (2019).
    2. •• Describes demographics and disease characteristics of patients in the ongoing, observational ataluren STRIDE Registry.

    1. Birnkrant DJ, Bushby K, Bann CM et al. Diagnosis and management of Duchenne muscular dystrophy, part 1: diagnosis, and neuromuscular, rehabilitation, endocrine, and gastrointestinal and nutritional management. Lancet Neurol. 17(3), 251–267 (2018).
    1. Birnkrant DJ, Bushby K, Bann CM et al. Diagnosis and management of Duchenne muscular dystrophy, part 3: primary care, emergency management, psychosocial care, and transitions of care across the lifespan. Lancet Neurol. 17(5), 445–455 (2018).
    1. Henricson EK, Abresch RT, Cnaan A et al. The cooperative international neuromuscular research group Duchenne natural history study: glucocorticoid treatment preserves clinically meaningful functional milestones and reduces rate of disease progression as measured by manual muscle testing and other commonly used clinical trial outcome measures. Muscle Nerve 48(1), 55–67 (2013).
    1. McDonald CM, Henricson EK, Abresch RT et al. Long-term effects of glucocorticoids on function, quality of life, and survival in patients with Duchenne muscular dystrophy: a prospective cohort study. Lancet 391(10119), 451–461 (2018).
    2. •• Describes the long-term effects of corticosteroids on disease progression in patients with DMD in the Cooperative International Neuromuscular Research Group Duchenne Natural History Study (CINRG DNHS).

    1. McDonald CM, Henricson EK, Abresch RT et al. The cooperative international neuromuscular research group Duchenne natural history study--a longitudinal investigation in the era of glucocorticoid therapy: design of protocol and the methods used. Muscle Nerve 48(1), 32–54 (2013).
    1. Inacio MC, Chen Y, Paxton EW, Namba RS, Kurtz SM, Cafri G. Statistics in brief: an introduction to the use of propensity scores. Clin. Orthop. Relat. Res. 473(8), 2722–2726 (2015).
    1. Ciafaloni E, Kumar A, Liu K et al. Age at onset of first signs or symptoms predicts age at loss of ambulation in Duchenne and Becker Muscular Dystrophy: Data from the MD STARnet. J. Pediatr. Rehab. Med. 9(1), 5–11 (2016).
    1. Ricotti V, Ridout DA, Scott E et al. Long-term benefits and adverse effects of intermittent versus daily glucocorticoids in boys with Duchenne muscular dystrophy. J. Neurol. Neurosurg. Psych. 84(6), 698–705 (2013).
    1. Coca-Perraillon M. Local and global optimal propensity score matching. SAS Global Forum 185 (2007).
    1. Finkel RS, Flanigan KM, Wong B et al. Phase IIa study of ataluren-mediated dystrophin production in patients with nonsense mutation Duchenne muscular dystrophy. PLoS ONE 8(12), e81302 (2013).
    1. Trucco F, Domingos JP, Tay CG et al. Cardiorespiratory progression over 5 years and role of corticosteroids in Duchenne muscular dystrophy: a single-site retrospective longitudinal study. Chest 158(4), 1606–1616 (2020).
    1. Peay HL, Hollin I, Fischer R, Bridges JF. A community-engaged approach to quantifying caregiver preferences for the benefits and risks of emerging therapies for Duchenne muscular dystrophy. Clin. Ttherapeut. 36(5), 624–637 (2014b).

Source: PubMed

3
Subscribe