Longitudinal effect of eteplirsen versus historical control on ambulation in Duchenne muscular dystrophy

Jerry R Mendell, Nathalie Goemans, Linda P Lowes, Lindsay N Alfano, Katherine Berry, James Shao, Edward M Kaye, Eugenio Mercuri, Eteplirsen Study Group and Telethon Foundation DMD Italian Network, M Pane, E Mazzone, S Messina, G L Vita, A D Amico, Bertini, A Berardinelli, Y Torrente, F Magri, G P Comi, G Baranello, T Mongini, A Pini, R Battini, E Pegoraro, C Bruno, L Politano, S Previtali, Hoda Abdel Hamid, Barry J Byrne, Anne M Connolly, Robert A Dracker, L Matthew Frank, Peter T Heydemann, Kevin C O'Brien, Susan E Sparks, Linda A Specht, Louise Rodino-Klapac, Zarife Sahenk, Samiah Al-Zaidy, Linda H Cripe, Sarah Lewis, Jerry R Mendell, Nathalie Goemans, Linda P Lowes, Lindsay N Alfano, Katherine Berry, James Shao, Edward M Kaye, Eugenio Mercuri, Eteplirsen Study Group and Telethon Foundation DMD Italian Network, M Pane, E Mazzone, S Messina, G L Vita, A D Amico, Bertini, A Berardinelli, Y Torrente, F Magri, G P Comi, G Baranello, T Mongini, A Pini, R Battini, E Pegoraro, C Bruno, L Politano, S Previtali, Hoda Abdel Hamid, Barry J Byrne, Anne M Connolly, Robert A Dracker, L Matthew Frank, Peter T Heydemann, Kevin C O'Brien, Susan E Sparks, Linda A Specht, Louise Rodino-Klapac, Zarife Sahenk, Samiah Al-Zaidy, Linda H Cripe, Sarah Lewis

Abstract

Objective: To continue evaluation of the long-term efficacy and safety of eteplirsen, a phosphorodiamidate morpholino oligomer designed to skip DMD exon 51 in patients with Duchenne muscular dystrophy (DMD). Three-year progression of eteplirsen-treated patients was compared to matched historical controls (HC).

Methods: Ambulatory DMD patients who were ≥7 years old and amenable to exon 51 skipping were randomized to eteplirsen (30/50mg/kg) or placebo for 24 weeks. Thereafter, all received eteplirsen on an open-label basis. The primary functional assessment in this study was the 6-Minute Walk Test (6MWT). Respiratory muscle function was assessed by pulmonary function testing (PFT). Longitudinal natural history data were used for comparative analysis of 6MWT performance at baseline and months 12, 24, and 36. Patients were matched to the eteplirsen group based on age, corticosteroid use, and genotype.

Results: At 36 months, eteplirsen-treated patients (n = 12) demonstrated a statistically significant advantage of 151m (p < 0.01) on 6MWT and experienced a lower incidence of loss of ambulation in comparison to matched HC (n = 13) amenable to exon 51 skipping. PFT results remained relatively stable in eteplirsen-treated patients. Eteplirsen was well tolerated. Analysis of HC confirmed the previously observed change in disease trajectory at age 7 years, and more severe progression was observed in patients with mutations amenable to exon skipping than in those not amenable. The subset of patients amenable to exon 51 skipping showed a more severe disease course than those amenable to any exon skipping.

Interpretation: Over 3 years of follow-up, eteplirsen-treated patients showed a slower rate of decline in ambulation assessed by 6MWT compared to untreated matched HC.

Trial registration: ClinicalTrials.gov NCT01396239 NCT01540409.

© 2016 The Authors. Annals of Neurology published by Wiley Periodicals, Inc. on behalf of American Neurological Association.

Figures

Figure 1
Figure 1
Study design: 4658‐201 and 4658‐202. Twelve patients with Duchenne muscular dystrophy were randomly assigned to 1 of 3 cohorts receiving weekly intravenous (IV) infusions in a 24‐week, double‐blind, placebo‐controlled study (study 201): eteplirsen 30mg/kg (solid line), eteplirsen 50mg/kg (dashed line), or placebo (dotted line). At week 25, eteplirsen‐treated patients continued the same weekly dose open‐label and placebo patients were randomized to open‐label treatment with eteplirsen 30mg/kg or 50mg/kg weekly IV (study 202). Functional clinical assessments including the 6‐Minute Walk Test and pulmonary function tests were performed at each week shown on the time axis. Muscle biopsies for evaluation of dystrophin were obtained from the upper arm at the time points specified; data reported elsewhere.1 BL = baseline.
Figure 2
Figure 2
Subgroup identification. The hierarchical chart shows the predefined filters by which patients were evaluated to select the most appropriate subgroups for comparison, as well as the most appropriate matched cohorts for comparison to eteplirsen‐treated patients. 6MWT = 6‐Minute Walk Test.
Figure 3
Figure 3
Historical control longitudinal 6‐minute walk distance over 3 years (mean ± standard error of the mean). (A) Age dichotomy predicts a change in disease trajectory at age 7 years. Disease progression trajectory is shown for steroid‐treated Duchenne muscular dystrophy historical controls who were older and younger than 7 years and amenable to skipping any exon. (B) 6‐Minute Walk Test (6MWT) performance declines more rapidly in patients with mutations amenable to exon skipping compared to those not amenable to exon skipping. Disease progression trajectory is compared in steroid‐treated patients ≥7 years of age with genotypes amenable or not amenable to exon skipping therapy. (C) 6MWT performance declines more rapidly in patients amenable to exon 51 skipping versus patients amenable to skipping other exons. Disease progression trajectory is shown in steroid‐treated patients ≥7 years of age with genotypes amenable to exon 51 skipping or amenable to skipping other exons. y.o. = years old.
Figure 4
Figure 4
Longitudinal 6‐minute walk distance (mean ± standard error of the mean) and loss of ambulation over 3 years. (A) Eteplirsen‐treated patients experience slower disease progression than matched historical controls. Disease progression trajectory is shown for steroid‐treated historical controls, ≥7 years old amenable to exon 51 skipping (n = 13) and eteplirsen‐treated patients (n = 12). Baseline characteristics for eteplirsen‐treated patients and those of the matched historical controls amenable to exon 51 skipping were comparable on mean age (difference of

Figure 5

Stability of respiratory muscle function…

Figure 5

Stability of respiratory muscle function as assessed by pulmonary function testing (mean ±…

Figure 5
Stability of respiratory muscle function as assessed by pulmonary function testing (mean ± standard error of the mean). Observed percentage of predicted (% Pred) maximum expiratory pressure (MEP), maximum inspiratory pressure (MIP), and forced vital capacity (FVC) demonstrate relative stability of respiratory muscle strength in all patients over >3 years of treatment with eteplirsen. % Pred MEP, MIP, and FVC at month 36 were 74.3, 89.5, and 91.9, respectively. BL = baseline.
Figure 5
Figure 5
Stability of respiratory muscle function as assessed by pulmonary function testing (mean ± standard error of the mean). Observed percentage of predicted (% Pred) maximum expiratory pressure (MEP), maximum inspiratory pressure (MIP), and forced vital capacity (FVC) demonstrate relative stability of respiratory muscle strength in all patients over >3 years of treatment with eteplirsen. % Pred MEP, MIP, and FVC at month 36 were 74.3, 89.5, and 91.9, respectively. BL = baseline.

References

    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. Kole R, Krieg AM. Exon skipping therapy for Duchenne muscular dystrophy. Adv Drug Deliv Rev 2015;87:104–107.
    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 2013;48:55–67.
    1. Muntoni F, Wood MJ. Targeting RNA to treat neuromuscular disease. Nat Rev Drug Discov 2011;10:621–637.
    1. Ciafaloni E, Fox DJ, Pandya S, et al. Delayed diagnosis in Duchenne muscular dystrophy: data from the Muscular Dystrophy Surveillance, Tracking, and Research Network (MD STARnet). J Pediatrics 2009;155:380–385.
    1. Brooke MH, Fenichel GM, Griggs RC, et al. Duchenne muscular dystrophy: patterns of clinical progression and effects of supportive therapy. Neurology 1989;39:475–481.
    1. van Deutekom JC, Janson AA, Ginjaar IB, et al. Local dystrophin restoration with antisense oligonucleotide PRO051. N Engl J Med 2007;357:2677–2686.
    1. Eagle M, Baudouin SV, Chandler C, et al. 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. Kole R, Krainer AR, Altman S. RNA therapeutics: beyond RNA interference and antisense oligonucleotides. Nat Rev Drug Discov 2012;11:125–140.
    1. 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.
    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. Cirak S, Feng L, Anthony K, et al. Restoration of the dystrophin‐associated glycoprotein complex after exon skipping therapy in Duchenne muscular dystrophy. Mol Ther 2012;20:462–467.
    1. McDonald CM, Henricson EK, Han JJ, et al. The 6‐minute walk test as a new outcome measure in Duchenne muscular dystrophy. Muscle Nerve 2010;41:500–510.
    1. Goemans N, Klingels K, van den Hauwe M, et al. Six‐minute walk test: reference values and prediction equation in healthy boys aged 5 to 12 years. PloS One 2013;8:e84120.
    1. Goemans N, van den Hauwe M, Wilson R, et al. Ambulatory capacity and disease progression as measured by the 6‐minute‐walk‐distance in Duchenne muscular dystrophy subjects on daily corticosteroids. Neuromuscul Disord 2013;23:618–623.
    1. Henricson E, Abresch R, Han JJ, et al. The 6‐minute walk test and person‐reported outcomes in boys with Duchenne muscular dystrophy and typically developing controls: longitudinal comparisons and clinically‐meaningful changes over one year. PLoS Curr 2013(Jul 8);5.
    1. Mazzone E, Vasco G, Sormani MP, et al. Functional changes in Duchenne muscular dystrophy: a 12‐month longitudinal cohort study. Neurology 2011;77:250–256.
    1. Mazzone ES, Pane M, Sormani MP, et al. 24 month longitudinal data in ambulant boys with Duchenne muscular dystrophy. PloS One 2013;8:e52512.
    1. McDonald CM, Henricson EK, Han JJ, et al. The 6‐minute walk test in Duchenne/Becker muscular dystrophy: longitudinal observations. Muscle Nerve 2010;42:966–974.
    1. 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.
    1. 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.
    1. Ferlini A, Goemans N, Tulinius EH, et al. Exon Skipping and PRO044 in Duchenne muscular dystrophy: extending the program. Neuromuscul Disord 2013;23:847.
    1. Henricson E, Abresch R, Han JJ, et al. Percent‐predicted 6‐minute walk distance in Duchenne muscular dystrophy to account for maturational influences. PLoS Curr 2012;4:RRN1297.
    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 2013;48:32–54.
    1. Pane M, Mazzone ES, Sormani MP, et al. 6 minute walk test in Duchenne MD patients with different mutations: 12 month changes. PloS One 2014;9:e83400.
    1. 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.
    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. Bushby K, Finkel R, Birnkrant DJ, et al. Diagnosis and management of Duchenne muscular dystrophy, part 2: implementation of multidisciplinary care. Lancet Neurol 2010;9:177–189.
    1. McDonald CM, Henricson EK, Abresch RT, et al. The 6‐minute walk test and other clinical endpoints in Duchenne muscular dystrophy: reliability, concurrent validity, and minimal clinically important differences from a multicenter study. Muscle Nerve 2013;48:357–368.
    1. Mazzone E, Martinelli D, Berardinelli A, et al. North Star Ambulatory Assessment, 6‐minute walk test and timed items in ambulant boys with Duchenne muscular dystrophy. Neuromuscul Disord 2010;20:712–716.
    1. Khirani S, Ramirez A, Aubertin G, et al. Respiratory muscle decline in Duchenne muscular dystrophy. Pediatr Pulmonol 2014;49:473–481.
    1. Mayer OH, Finkel RS, Rummey C, et al. Characterization of pulmonary function in Duchenne muscular dystrophy. Pediatr Pulmonol 2015;50:487–494.
    1. 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.
    1. Wilson SH, Cooke NT, Edwards RH, Spiro SG. Predicted normal values for maximal respiratory pressures in Caucasian adults and children. Thorax 1984;39:535–538.
    1. Polgar GP, Varuni. Pulmonary function testing in children: techniques and standards. Philadelphia, PA: Saunders, 1971.
    1. Alexander MA, Johnson EW, Petty J, Stauch D. Mechanical ventilation of patients with late stage Duchenne muscular dystrophy: management in the home. Arch Phys Med Rehabil 1979;60:289–292.
    1. Kieny P, Chollet S, Delalande P, et al. Evolution of life expectancy of patients with Duchenne muscular dystrophy at AFM Yolaine de Kepper centre between 1981 and 2011. Ann Phys Rehabil Med 2013;56:443–454.
    1. Phillips MF, Smith PE, Carroll N, et al. Nocturnal oxygenation and prognosis in Duchenne muscular dystrophy. Am J Respir Crit Care Med 1999;160:198–202.
    1. Griggs RC, Donohoe KM, Utell MJ, et al. Evaluation of pulmonary function in neuromuscular disease. Arch Neurol 1981;38:9–12.
    1. Hahn A, Bach JR, Delaubier A, et al. Clinical implications of maximal respiratory pressure determinations for individuals with Duchenne muscular dystrophy. Arch Phys Med Rehabil 1997;78:1–6.
    1. Sazani P, Weller DL, Shrewsbury SB. Safety pharmacology and genotoxicity evaluation of AVI‐4658. Int J Toxicol 2010;29:143–156.
    1. Sazani P, Ness KP, Weller DL, et al. Repeat‐dose toxicology evaluation in cynomolgus monkeys of AVI‐4658, a phosphorodiamidate morpholino oligomer (PMO) drug for the treatment of Duchenne muscular dystrophy. Int J Toxicol 2011;30:313–321.
    1. Sazani P, Ness KP, Weller DL, et al. Chemical and mechanistic toxicology evaluation of exon skipping phosphorodiamidate morpholino oligomers in mdx mice. Int J Toxicol 2011;30:322–333.
    1. Heald AE, Charleston JS, Iversen PL, et al. AVI‐7288 for Marburg virus in nonhuman primates and humans. N Engl J Med 2015;373:339–348.
    1. Muntoni F, Bushby KD, van Ommen G. 149th ENMC International Workshop and 1st TREAT‐NMD Workshop on: “planning phase i/ii clinical trials using systemically delivered antisense oligonucleotides in Duchenne muscular dystrophy.” Neuromuscul Disord 2008;18:268–275.
    1. Kishnani PS, Corzo D, Nicolino M, et al. Recombinant human acid [alpha]‐glucosidase: major clinical benefits in infantile‐onset Pompe disease. Neurology 2007;68:99–109.
    1. Ceprotin prescribing information. Deerfield, IL: Baxter International, 2011.

Source: PubMed

3
Abonnieren