Safety, Tolerability, and Efficacy of Viltolarsen in Boys With Duchenne Muscular Dystrophy Amenable to Exon 53 Skipping: A Phase 2 Randomized Clinical Trial

Paula R Clemens, Vamshi K Rao, Anne M Connolly, Amy D Harper, Jean K Mah, Edward C Smith, Craig M McDonald, Craig M Zaidman, Lauren P Morgenroth, Hironori Osaki, Youhei Satou, Taishi Yamashita, Eric P Hoffman, CINRG DNHS Investigators, Paula R Clemens, Vamshi K Rao, Anne M Connolly, Amy D Harper, Jean K Mah, Edward C Smith, Craig M McDonald, Craig M Zaidman, Lauren P Morgenroth, Hironori Osaki, Youhei Satou, Taishi Yamashita, Eric P Hoffman, CINRG DNHS Investigators

Abstract

Importance: An unmet need remains for safe and efficacious treatments for Duchenne muscular dystrophy (DMD). To date, there are limited agents available that address the underlying cause of the disease.

Objective: To evaluate the safety, tolerability, and efficacy of viltolarsen, a novel antisense oligonucleotide, in participants with DMD amenable to exon 53 skipping.

Design, setting, and participants: This phase 2 study was a 4-week randomized clinical trial for safety followed by a 20-week open-label treatment period of patients aged 4 to 9 years with DMD amenable to exon 53 skipping. To enroll 16 participants, with 8 participants in each of the 2 dose cohorts, 17 participants were screened. Study enrollment occurred between December 16, 2016, and August 17, 2017, at sites in the US and Canada. Data were collected from December 2016 to February 2018, and data were analyzed from April 2018 to May 2019.

Interventions: Participants received 40 mg/kg (low dose) or 80 mg/kg (high dose) of viltolarsen administered by weekly intravenous infusion.

Main outcomes and measures: Primary outcomes of the trial included safety, tolerability, and de novo dystrophin protein production measured by Western blot in participants' biceps muscles. Secondary outcomes included additional assessments of dystrophin mRNA and protein production as well as clinical muscle strength and function.

Results: Of the 16 included boys with DMD, 15 (94%) were white, and the mean (SD) age was 7.4 (1.8) years. After 20 to 24 weeks of treatment, significant drug-induced dystrophin production was seen in both viltolarsen dose cohorts (40 mg/kg per week: mean [range] 5.7% [3.2-10.3] of normal; 80 mg/kg per week: mean [range] 5.9% [1.1-14.4] of normal). Viltolarsen was well tolerated; no treatment-emergent adverse events required dose reduction, interruption, or discontinuation of the study drug. No serious adverse events or deaths occurred during the study. Compared with 65 age-matched and treatment-matched natural history controls, all 16 participants treated with viltolarsen showed significant improvements in timed function tests from baseline, including time to stand from supine (viltolarsen: -0.19 s; control: 0.66 s), time to run/walk 10 m (viltolarsen: 0.23 m/s; control: -0.04 m/s), and 6-minute walk test (viltolarsen: 28.9 m; control: -65.3 m) at the week 25 visit.

Conclusions and relevance: Systemic treatment of participants with DMD with viltolarsen induced de novo dystrophin production, and clinical improvement of timed function tests was observed.

Trial registration: ClinicalTrials.gov Identifier: NCT02740972.

Conflict of interest statement

Conflict of Interest Disclosures: Dr Clemens has received grants from NS Pharma during the conduct of the study as well as grants from Spark Therapeutics, Amicus Therapeutics, Sanofi Genzyme, ReveraGen BioPharma, and TRiNDS and personal fees from Spark Therapeutics, UCB, Pfizer, and Roche outside the submitted work. Dr Rao has received nonfinancial support from Ann and Robert H. Lurie Children’s Hospital of Chicago during the conduct of the study as well as personal fees from PTC Therapeutics and Sarepta Therapeutics outside the submitted work. Dr Connelly has received grants from the Washington University School of Medicine and TRiNDS during the conduct of the study as well as grants from Sarepta Therapeutics and AveXis and personal fees from Sarepta Therapeutics, AveXis, Genentech-Roche, Acceleron Pharma, and Catabasis Pharmaceuticals outside the submitted work. Dr Harper has received grants from NS Pharma, Italfarmaco, ReveraGen BioPharma, Catabasis Pharmaceuticals, AveXis, CSL Behring, Teva Pharmaceutical Industries, National Institutes of Health, and US Centers for Disease Control and Prevention. Dr Mah has received grants from NS Pharma during the conduct of the study as well as grants from PTC Therapeutics, Pfizer, Roche, Sarepta Therapeutics, Italfarmaco, Novartis, Biogen, ReveraGen BioPharma, Catabasis Pharmaceuticals, Sanofi-Genzyme, and Bristol-Myers Squibb outside the submitted work. Dr Smith has received salary support from NS Pharma during the conduct of the study as well as personal fees from Pfizer and Sarepta Therapeutics and salary support from Pfizer and ReveraGen BioPharma outside the submitted work. Dr McDonald has received grants from NS Pharma during the conduct of the study as well as personal fees from Bristol-Myers Squibb, Astellas/Mitobridge, Cardero Therapeutics, Capricor Therapeutics, Catabasis Pharmaceuticals, Eli Lilly and Company, Epirium Bio, FibroGen, Halo Therapeutics, Italfarmaco, BioMarin Pharmaceutical, Novartis, Pfizer, Prosensa, PTC Therapeutics, Santhera Pharmaceuticals, and Sarepta Therapeutics outside the submitted work. Dr Zaidman has received nonfinancial support from Biogen outside the submitted work. Ms Morgenroth has received salary support from TRiNDS during the conduct of the study. Mr Osaki and Dr Yamashita are employed by NS Pharma. Mr Satou is employed by NS Pharma and has patent PCT/JP2011/070318 issued and licensed to Nippon Shinyaku and the National Center of Neurology and Psychiatry and has patent PCT/JP2019/026393 pending. Dr Hoffman has received grants from AGADA Biosciences and TRiNDS during the conduct of the study and grants from ReveraGen BioPharma outside the submitted work.

Figures

Figure 1.. CONSORT Diagram
Figure 1.. CONSORT Diagram
Participant flow throughout the trial.
Figure 2.. Evaluation of Dystrophin Induction
Figure 2.. Evaluation of Dystrophin Induction
Muscle biopsies randomized and tested by blinded analysts. A, Dystrophin Western blot. Examples of participants with Duchenne muscular dystrophy treated with 40 mg/kg per week (low-dose cohort) and 80 mg/kg per week (high-dose cohort) of viltolarsen are shown, together with standard curve for dystrophin (D) quantitation. The lanes indicate individual participants; samples were pretreatment (baseline) and posttreatment (week 25 visit) biopsies. Normalization was to both myosin heavy chains (M) and α-actinin (A). All biopsies were analyzed with triplicate gels. B, Percentage of normal dystrophin levels was graphed for each participant at baseline and posttreatment at the week 25 visit. Dystrophin was measured using Western blot and normalized to myosin heavy chains. The blue line denotes 3% of normal dystrophin levels. Each line indicates a different patient. C, Dystrophin mRNA reverse transcription–polymerase chain reaction (RT-PCR). Examples of pretreatment and posttreatment biopsies from the high-dose group are shown, each tested in duplicate assays. D, Immunofluorescence staining of muscle biopsies for dystrophin and colocalization controls. Serial cryosections were double stained with dystrophin and laminin α2 or dystrophin and α-sarcoglycan from a participant who received 80 mg/kg per week of viltolarsen.
Figure 3.. Change in Timed Function Tests…
Figure 3.. Change in Timed Function Tests in Viltolarsen-Treated Participants and Cooperative International Neuromuscular Research Group (CINRG) Duchenne Natural History Study (DNHS)
The change in timed function tests of viltolarsen-treated participants from both dose groups (blue) and CINRG DNHS steroid-treated, age-matched comparators (orange) are shown. Assessments were performed at baseline and the 13-week and 25-week visits.

References

    1. Hoffman EP, Brown RH Jr, Kunkel LM. Dystrophin: the protein product of the Duchenne muscular dystrophy locus. Cell. 1987;51(6):919-928. doi:10.1016/0092-8674(87)90579-4
    1. Uaesoontrachoon K, Srinivassane S, Warford J, et al. . Orthogonal analysis of dystrophin protein and mRNA as a surrogate outcome for drug development. Biomark Med. 2019;13(14):1209-1225. doi:10.2217/bmm-2019-0242
    1. Darin N, Tulinius M. Neuromuscular disorders in childhood: a descriptive epidemiological study from western Sweden. Neuromuscul Disord. 2000;10(1):1-9. doi:10.1016/S0960-8966(99)00055-3
    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. 2015;36(4):395-402. doi:10.1002/humu.22758
    1. Hoffman EP, Fischbeck KH, Brown RH, et al. . Characterization of dystrophin in muscle-biopsy specimens from patients with Duchenne’s or Becker’s muscular dystrophy. N Engl J Med. 1988;318(21):1363-1368. doi:10.1056/NEJM198805263182104
    1. Bushby K, Finkel R, Birnkrant DJ, et al. ; DMD Care Considerations Working Group . Diagnosis and management of Duchenne muscular dystrophy, part 1: diagnosis, and pharmacological and psychosocial management. Lancet Neurol. 2010;9(1):77-93. doi:10.1016/S1474-4422(09)70271-6
    1. Beggs AH, Hoffman EP, Snyder JR, et al. . Exploring the molecular basis for variability among patients with Becker muscular dystrophy: dystrophin gene and protein studies. Am J Hum Genet. 1991;49(1):54-67.
    1. Bello L, Gordish-Dressman H, Morgenroth LP, et al. ; CINRG Investigators . Prednisone/prednisolone and deflazacort regimens in the CINRG Duchenne Natural History Study. Neurology. 2015;85(12):1048-1055. doi:10.1212/WNL.0000000000001950
    1. Verhaart IEC, Aartsma-Rus A. Therapeutic developments for Duchenne muscular dystrophy. Nat Rev Neurol. 2019;15(7):373-386. doi:10.1038/s41582-019-0203-3
    1. Komaki H, Nagata T, Saito T, et al. . Systemic administration of the antisense oligonucleotide NS-065/NCNP-01 for skipping of exon 53 in patients with Duchenne muscular dystrophy. Sci Transl Med. 2018;10(437):eaan0713. doi:10.1126/scitranslmed.aan0713
    1. Watanabe N, Nagata T, Satou Y, et al. . NS-065/NCNP-01: an antisense oligonucleotide for potential treatment of exon 53 skipping in Duchenne muscular dystrophy. Mol Ther Nucleic Acids. 2018;13:442-449. doi:10.1016/j.omtn.2018.09.017
    1. McDonald CM, Henricson EK, Abresch RT, et al. ; Cinrg Investigators . 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(1):32-54. doi:10.1002/mus.23807
    1. McDonald CM, Henricson EK, Abresch RT, et al. ; CINRG Investigators . Long-term effects of glucocorticoids on function, quality of life, and survival in patients with Duchenne muscular dystrophy: a prospective cohort study. Lancet. 2018;391(10119):451-461. doi:10.1016/S0140-6736(17)32160-8
    1. Extension study of NS-065/NCNP-01 in boys with Duchenne muscular dystrophy (DMD). identifier: NCT03167255. Updated March 21, 2019. Accessed January 6, 2020.
    1. US Food and Drug Administration . Table of surrogate endpoints that were the basis of drug approval or licensure. Accessed December 6, 2019.
    1. Charleston JS, Schnell FJ, Dworzak J, et al. . Eteplirsen treatment for Duchenne muscular dystrophy: exon skipping and dystrophin production. Neurology. 2018;90(24):e2146-e2154. doi:10.1212/WNL.0000000000005680
    1. Muntoni F, Frank D, Sardone V, et al. . Golodirsen induces exon skipping leading to sarcolemmal dystrophin expression in Duchenne muscular dystrophy patients with mutations amenable to exon 53 skipping (S22.001). Neurology. 2018;90(15)(suppl):S22.001.
    1. van den Bergen JC, Wokke BH, Janson AA, et al. . Dystrophin levels and clinical severity in Becker muscular dystrophy patients. J Neurol Neurosurg Psychiatry. 2014;85(7):747-753. doi:10.1136/jnnp-2013-306350
    1. Bello L, Campadello P, Barp A, et al. . Functional changes in Becker muscular dystrophy: implications for clinical trials in dystrophinopathies. Sci Rep. 2016;6:32439. doi:10.1038/srep32439
    1. Beekman C, Janson AA, Baghat A, van Deutekom JC, Datson NA. Use of capillary Western immunoassay (Wes) for quantification of dystrophin levels in skeletal muscle of healthy controls and individuals with Becker and Duchenne muscular dystrophy. PLoS One. 2018;13(4):e0195850. doi:10.1371/journal.pone.0195850
    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(1):e83400. doi:10.1371/journal.pone.0083400
    1. Wang RT, Barthelemy F, Martin AS, et al. . DMD genotype correlations from the Duchenne Registry: endogenous exon skipping is a factor in prolonged ambulation for individuals with a defined mutation subtype. Hum Mutat. 2018;39(9):1193-1202. doi:10.1002/humu.23561
    1. US Food and Drug Administration . Duchenne muscular dystrophy and related dystrophinopathies: developing drugs for treatment guidance for industry. Accessed December 6, 2019.
    1. Institute for Clinical and Economic Review . Deflazacort, eteplirsen, and golodirsen for Duchenne muscular dystrophy: effectiveness and value. Accessed December 6, 2019.
    1. Cordova G, Negroni E, Cabello-Verrugio C, Mouly V, Trollet C. Combined therapies for Duchenne muscular dystrophy to optimize treatment efficacy. Front Genet. 2018;9:114. doi:10.3389/fgene.2018.00114
    1. Blake DJ, Weir A, Newey SE, Davies KE. Function and genetics of dystrophin and dystrophin-related proteins in muscle. Physiol Rev. 2002;82(2):291-329. doi:10.1152/physrev.00028.2001
    1. Mazzone ES, Pane M, Sormani MP, et al. . 24 month longitudinal data in ambulant boys with Duchenne muscular dystrophy. PLoS One. 2013;8(1):e52512. doi:10.1371/journal.pone.0052512
    1. Mendell JR, Goemans N, Lowes LP, et al. ; Eteplirsen Study Group and Telethon Foundation DMD Italian Network . Longitudinal effect of eteplirsen versus historical control on ambulation in Duchenne muscular dystrophy. Ann Neurol. 2016;79(2):257-271. doi:10.1002/ana.24555
    1. Mendell JR, Rodino-Klapac LR, Sahenk Z, et al. ; Eteplirsen Study Group . Eteplirsen for the treatment of Duchenne muscular dystrophy. Ann Neurol. 2013;74(5):637-647. doi:10.1002/ana.23982
    1. Goemans N, Mercuri E, Belousova E, et al. ; DEMAND III study group . A randomized placebo-controlled phase 3 trial of an antisense oligonucleotide, drisapersen, in Duchenne muscular dystrophy. Neuromuscul Disord. 2018;28(1):4-15. doi:10.1016/j.nmd.2017.10.004
    1. BioMarin announces withdrawal of market authorization application for Kyndrisa™ (drisapersen) in Europe. News release. Globe Newswire; May 31, 2016. Accessed December 6, 2019.
    1. Phase I/II study of SRP-4053 in DMD patients. identifier: NCT02310906. Updated June 12, 2019. Accessed December 6, 2019.

Source: PubMed

3
Předplatit