Long-term treatment with eteplirsen in nonambulatory patients with Duchenne muscular dystrophy

Lindsay N Alfano, Jay S Charleston, Anne M Connolly, Linda Cripe, Cas Donoghue, Robert Dracker, Johannes Dworzak, Helen Eliopoulos, Diane E Frank, Sarah Lewis, Karin Lucas, Jessie Lynch, A J Milici, Amy Flynt, Emily Naughton, Louise R Rodino-Klapac, Zarife Sahenk, Frederick J Schnell, G David Young, Jerry R Mendell, Linda P Lowes, Lindsay N Alfano, Jay S Charleston, Anne M Connolly, Linda Cripe, Cas Donoghue, Robert Dracker, Johannes Dworzak, Helen Eliopoulos, Diane E Frank, Sarah Lewis, Karin Lucas, Jessie Lynch, A J Milici, Amy Flynt, Emily Naughton, Louise R Rodino-Klapac, Zarife Sahenk, Frederick J Schnell, G David Young, Jerry R Mendell, Linda P Lowes

Abstract

This analysis aims to describe the outcomes of two nonambulatory patients with Duchenne muscular dystrophy (DMD) who participated in two clinical studies. The two consecutive trials of eteplirsen (studies 201 and 202) were conducted in patients with DMD (N = 12) and confirmed genetic mutations amenable to exon 51 skipping.In study 201, 12 patients were randomized to receive once-weekly, double-blind intravenous infusions of eteplirsen 30 or 50 mg/kg or placebo for 24 weeks; patients then received open-label eteplirsen during weeks 25 through 28. All 12 patients continued onto open-label extension study 202 and received long-term treatment with eteplirsen. We compared cardiac, pulmonary, and upper limb function and dystrophin production in the nonambulatory twin patients versus the 10 ambulatory patients through 240 combined treatment weeks.Ten study patients remained ambulatory through both studies, while the identical twin patients both experienced early, rapid loss of ambulation. The twin patients had greater disease severity at baseline (6-minute walk test [6MWT], 330 and 256 m) versus the other patients (n = 10; 6MWT range, 341-418 m). They maintained cardiac and upper limb function through combined week 240, with outcomes similar to those of the patients who remained ambulatory. Dystrophin production was confirmed following eteplirsen treatment.Despite the loss of ambulation, other markers of disease progression remained relatively stable in the eteplirsen-treated twin patients and were similar to those of the ambulatory patients.

Conflict of interest statement

Conflicts of interest: Medical writing and editorial support were provided by Callie Grimes, PhD, and Barbara Zeman, PhD, of Peloton Advantage, LLC, an OPEN Health company (Parsippany, NJ), and funded by Sarepta Therapeutics, Inc. L. N. Alfano, L. Cripe, R. Dracker, Z. Sahenk, and L. Lowes have no conflicts to disclose. J. S. Charleston, J. Dworzak, H. Eliopoulos, D. E. Frank, K. Lucas, J. Lynch, E. Naughton, and F.J. Schnell are employees of Sarepta Therapeutics, Inc. and may own stock or options in the company. C. Donoghue was an employee at the time of the study. A. M. Connolly has served on advisory boards for AveXis, Inc., Cytokinetics, Inc., Sanofi Genzyme, and Sarepta Therapeutics, Inc. She serves on the data management safety board for Catabasis Pharmaceuticals, Inc., and as an investigator for AveXis, Inc., Biogen, Bristol-Myers Squibb, Cytokinetics, Inc., FibroGen, Italfarmeco SpA, NS Pharma, Inc., Pfizer Inc, PTC Therapeutics, Inc., and Sarepta Therapeutics, Inc. S. Lewis is an employee of Nationwide Children's Hospital, which received funding from Sarepta Therapeutics in the form of the study budget. A. J. Milici serves as a remunerated consultant for Sarepta Therapeutics, Inc. A. Flynt is an employee of PharPoint Research, which received funding from Sarepta Therapeutics for data analysis for the study. At the time of the study, L.R. Rodino-Klapac was an employee of Nationwide Children's Hospital, which received funding from Sarepta Therapeutics in the form of a research agreement; she is currently an employee of Sarepta Therapeutics, Inc., and may own stock/options in the company. She is the inventor of technology that has been exclusively optioned to Sarepta Therapeutics. G. D. Young serves as a remunerated consultant for Sarepta Therapeutics, Inc. J. R. Mendell serves on an advisory board for Sarepta Therapeutics, Inc.

Figures

Figure 1
Figure 1
Six-minute walk test distance for all patients at baseline (day 1 visit). 6MWT = 6-minute walk test.
Figure 2
Figure 2
Rapid decline and loss of ambulation in the eteplirsen-treated twin patients. 6MWT = 6-minute walk test.
Figure 3
Figure 3
Measures of pulmonary and cardiac function in patients A and B versus study mean through 240 weeks of eteplirsen treatment: (A) FVC%p and (B) LVEF. FVC%p = percent predicted forced vital capacity, LVEF = left ventricular ejection fraction.
Figure 4
Figure 4
Measures of upper limb function in patients A and B versus study mean through 240 weeks of eteplirsen treatment: (A) 9-Hole Peg Test; (B) grip strength; (C) elbow flexion; and (D) elbow extension. All data represent the right hand, which was the dominant hand for all 12 patients.
Figure 5
Figure 5
Dystrophin expression following 180 weeks of eteplirsen treatment, based on Western blot analysis and immunohistochemistry (A), Western blot image (B), PDPF (C), and the intensity of dystrophin fluorescent signal in muscle fibers (D). In panels A, C, and D, standard deviations are represented by error bars. In panel B, lanes in the Western blot image are identified left to right as the standard curve (4%, 2%, 1%, 0.5%, 0.25% of normal), patient samples (treated or untreated in blinded random order in next two lanes), and negative pooled DMD control sample. Patient samples representing patient A and patient B are identified in both figures. Note that either an unrelated patient sample representing an untreated (left figure) or treated (right figure) patient was run on the adjacent lane due to the random blinded nature that samples were evaluated. DMD = Duchenne muscular dystrophy, PDPF = percent dystrophin-positive fibers, tx = treated, untx = untreated.

References

    1. Kole R, Krieg AM. Exon skipping therapy for Duchenne muscular dystrophy. Adv Drug Deliv Rev 2015;87:104–7.
    1. Mendell JR, Rodino-Klapac LR, Sahenk Z, et al. Eteplirsen for the treatment of Duchenne muscular dystrophy. Ann Neurol 2013;74:637–47.
    1. McNeil DE, Davis C, Jillapalli D, et al. Duchenne muscular dystrophy: drug development and regulatory considerations. Muscle Nerve 2010;41:740–5.
    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;9:177–89.
    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. Fayssoil A, Nardi O, Orlikowski D, et al. Cardiomyopathy in Duchenne muscular dystrophy: pathogenesis and therapeutics. Heart Fail Rev 2010;15:103–7.
    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;13:987–96.
    1. Bushby K, Finkel R, Wong B, et al. Ataluren treatment of patients with nonsense mutation dystrophinopathy. Muscle Nerve 2014;50:477–87.
    1. Exondys 51 [package insert]. Cambridge, MA: Sarepta Therapeutics, Inc.; 2018.
    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;79:257–71.
    1. Charleston JS, Schnell FJ, Dworzak J, et al. Eteplirsen treatment for Duchenne muscular dystrophy: exon skipping and dystrophin production. Neurology 2018;90:e2146–54.
    1. Kinane TB, Mayer OH, Mendell JR, et al. Effects of long-term eteplirsen treatment on pulmonary function in patients with Duchenne muscular dystrophy: findings of two phase 2 clinical trials [poster]. Presented at: Annual Scientific Conference of the Muscular Dystrophy Association; March 19–22, 2017; Arlington, VA.
    1. Duda PW, Moody S, Colan S, et al. Effects of long-term treatment with eteplirsen on cardiac function in patients with DMD versus disease natural history [poster]. Presented at: Annual Scientific Conference of the Muscular Dystrophy Association; March 19–22, 2017; Arlington, VA.
    1. Poole JL, Burtner PA, Torres TA, et al. Measuring dexterity in children using the Nine-hole Peg Test. J Hand Ther 2005;18:348–51.
    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. Mazzone E, Vasco G, Sormani MP, et al. Functional changes in Duchenne muscular dystrophy: a 12-month longitudinal cohort study. Neurology 2011;77:250–6.
    1. LoMauro A, D’Angelo MG, Aliverti A. Assessment and management of respiratory function in patients with Duchenne muscular dystrophy: current and emerging options. Ther Clin Risk Manag 2015;11:1475–88.
    1. Roberto R, Fritz A, Hagar Y, et al. The natural history of cardiac and pulmonary function decline in patients with Duchenne muscular dystrophy. Spine 2011;36:E1009–1017.
    1. Mayer OH, Finkel RS, Rummey C, et al. Characterization of pulmonary function in Duchenne muscular dystrophy. Pediatr Pulmonol 2015;50:487–94.
    1. Connolly AM, Malkus EC, Mendell JR, et al. Outcome reliability in non-ambulatory boys/men with Duchenne muscular dystrophy. Muscle Nerve 2015;51:522–32.
    1. Mayhew JE, Florence JM, Mayhew TP, et al. Reliable surrogate outcome measures in multicenter clinical trials of Duchenne muscular dystrophy. Muscle Nerve 2007;35:36–42.
    1. Mayer OH, Henricson EK, McDonald CM, et al. Advances in pulmonary care in Duchenne muscular dystrophy. US Neurology 2017;13:35–41.
    1. Finder J, Mayer OH, Sheehan D, et al. Pulmonary endpoints in Duchenne muscular dystrophy. A workshop summary. Am J Respir Crit Care Med 2017;196:512–9.
    1. Connolly AM, Florence JM, Zaidman CM, et al. Clinical trial readiness in non-ambulatory boys and men with Duchenne muscular dystrophy: MDA-DMD network follow-up. Muscle Nerve 2016;54:681–9.
    1. Duboc D, Meune C, Lerebours G, et al. Effect of perindopril on the onset and progression of left ventricular dysfunction in Duchenne muscular dystrophy. J Am Coll Cardiol 2005;45:855–7.
    1. Tandon A, Villa CR, Hor KN, et al. Myocardial fibrosis burden predicts left ventricular ejection fraction and is associated with age and steroid treatment duration in Duchenne muscular dystrophy. J Am Heart Assoc 2015;4:e001338.
    1. Schram G, Fournier A, Leduc H, et al. All-cause mortality and cardiovascular outcomes with prophylactic steroid therapy in Duchenne muscular dystrophy. J Am Coll Cardiol 2013;61:948–54.
    1. Birnkrant DJ, Ararat E, Mhanna MJ. Cardiac phenotype determines survival in Duchenne muscular dystrophy. Pediatr Pulmonol 2016;51:70–6.
    1. Ricotti V, Evans MR, Sinclair CD, et al. Upper limb evaluation in Duchenne muscular dystrophy: fat-water quantification by MRI, muscle force and function define endpoints for clinical trials. PLoS One 2016;11:e0162542.
    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. Vestergaard P, Glerup H, Steffensen BF, et al. Fracture risk in patients with muscular dystrophy and spinal muscular atrophy. J Rehabil Med 2001;33:150–5.
    1. Archer JE, Gardner AC, Roper HP, et al. Duchenne muscular dystrophy: the management of scoliosis. J Spine Surg 2016;2:185–94.

Source: PubMed

3
Abonner