Safety and efficacy of nusinersen in spinal muscular atrophy: The EMBRACE study

Gyula Acsadi, Thomas O Crawford, Wolfgang Müller-Felber, Perry B Shieh, Randal Richardson, Niranjana Natarajan, Diana Castro, Daniela Ramirez-Schrempp, Giulia Gambino, Peng Sun, Wildon Farwell, Gyula Acsadi, Thomas O Crawford, Wolfgang Müller-Felber, Perry B Shieh, Randal Richardson, Niranjana Natarajan, Diana Castro, Daniela Ramirez-Schrempp, Giulia Gambino, Peng Sun, Wildon Farwell

Abstract

Introduction: The EMBRACE study (Clinical Trials No. NCT02462759) evaluated nusinersen in infants/children with infantile- or later-onset spinal muscular atrophy (SMA) who were ineligible for the ENDEAR and CHERISH studies.

Methods: Participants were randomized to intrathecal nusinersen (12-mg scaled equivalent dose; n = 14) or sham procedure (n = 7) in part 1 (~14 months) and subsequently received open-label nusinersen for ~24 months in part 2 of the study.

Results: Part 1 was stopped early after the demonstration of motor function benefit with nusinersen in ENDEAR. There were no nusinersen-related adverse events (AEs) and no study discontinuations due to nusinersen-related AEs. The most common AEs included pyrexia, cough, pneumonia, and upper respiratory tract infections. Motor milestone responder rates were higher in those receiving nusinersen at last available assessment (93%) than in those receiving sham procedure in part 1 (29%) or transitioned from sham to nusinersen in part 2 (83%). This functional improvement was observed despite the small sample size and shortened part 1 trial duration that undermined the power of the study to demonstrate such treatment effects at a significant level.

Discussion: Nusinersen demonstrated a favorable long-term benefit-risk profile in this broad population of individuals with infantile- or later-onset SMA.

Keywords: clinical tria; nusinersen; safety; spinal muscular atrophy; therapeutic use.

Conflict of interest statement

G.A. has participated in advisory boards for AveXis/Novartis, Biogen, Genentech, and Sarepta; and has participated in consultation and speaker engagements for Biogen. T.O.C. has been an advisor/consultant for AveXis/Novartis, Biogen, Catalyst, Cure SMA, Cytokinetics, Marathon, Novartis, Roche, Sarepta, and the SMA Foundation. W.M.‐F. has participated in advisory boards for AveXis/Novartis, Biogen, PTC, Roche, and Sarepta; has received honoraria from AveXis/Novartis, Biogen, and PTC; has received research funding from BMBF; and has clinical trial research contracts with Biogen, Santhera, and Sarepta. P.B.S. has participated in advisory boards for Biogen and, outside of the submitted work, has participated in advisory boards for AveXis/Novartis, PTC, and Sarepta. R.R. has participated in advisory boards for AveXis/Novartis and Biogen, and has clinical trial research contracts with Biogen. N.N. has received research funding from Biogen for execution of clinical trial projects and National Institutes of Health funding for site principal investigator work on the HEAL‐EEG study. D.C. has participated in advisory board for AveXis/Novartis, Biogen, PTC, and Sarepta; has received research funding from AveXis/Novartis, Biogen, FibroGen, PTC, ReveraGen, and Sarepta; and has participated in medical advisory boards for GBS‐CIDP Foundation, Myasthenia Gravis Foundation, and the Cure SMA Care Center Network. D.R.‐S., G.G., P.S., and W.F. are employees of and hold stock/stock options in Biogen.

© 2021 The Authors. Muscle & Nerve published by Wiley Periodicals LLC.

Figures

FIGURE 1
FIGURE 1
Participant disposition. Part 2 was terminated early to allow participants to transition to the SHINE study [Color figure can be viewed at wileyonlinelibrary.com]
FIGURE 2
FIGURE 2
Mean (standard error) data for: (A) predose CSF nusinersen concentrations (ng/mL; CSF‐evaluable population), (B) change from baseline in HINE‐2 total milestones, and (C) HINE‐2 total milestones. In A, values considered as outliers were excluded from the analyses and included observed concentrations in day 1 predose samples. Only visits with five or more participants are included. In B and C, only visits with four or more participants are included. For the purposes of comparison, the group that initiated nusinersen in part 2 is graphed from day 0 (first nusinersen dose), and in B the initial baseline HINE‐2 score reset to 0. In all graphs, data at each timepoint are offset to allow visualization of all participant groups. CSF, cerebrospinal fluid; HINE‐2, Hammersmith Infant Neurological Examination Section 2; SE, standard error [Color figure can be viewed at wileyonlinelibrary.com]
FIGURE 3
FIGURE 3
Plasma pNF‐H levels over time in part 1 of the study: (A) geometric mean (95% CI) and (B) geometric mean ratio (95% CI). One participant in the sham procedure group had a pNF‐H level below the limit of quantification at day 64 that was not included in the graph. CI, confidence interval; pNF‐H, phosphorylated neurofilament heavy chain [Color figure can be viewed at wileyonlinelibrary.com]

References

    1. Crawford TO , Pardo CA. The neurobiology of childhood spinal muscular atrophy. Neurobiol Dis. 1996;3:97‐110.
    1. Markowitz JA, Singh P, Darras BT. Spinal muscular atrophy: a clinical and research update. Pediatr Neurol. 2012;46:1‐12.
    1. Finkel R, Bertini E, Muntoni F, Mercuri E, Workshop Study ENMCSMA. Group. 209th ENMC international workshop: outcome measures and clinical trial readiness in spinal muscular atrophy 7‐9 November 2014, Heemskerk, The Netherlands. Neuromuscul Disord. 2015;25:593‐602.
    1. Russman BS. Spinal muscular atrophy: clinical classification and disease heterogeneity. J Child Neurol. 2007;22:946‐951.
    1. Lefebvre S, Burlet P, Liu Q, et al. Correlation between severity and SMN protein level in spinal muscular atrophy. Nat Genet. 1997;16:265‐269.
    1. Feldkötter M, Schwarzer V, Wirth R, Wienker TF, Wirth B. Quantitative analyses of SMN1 and SMN2 based on real‐time LightCycler PCR: fast and highly reliable carrier testing and prediction of severity of spinal muscular atrophy. Am J Hum Genet. 2002;70:358‐368.
    1. European Medicines Agency . Spinraza 12 mg solution for injection. 2017. . Accessed June 30, 2020.
    1. Biogen. Spinraza (nusinersen) injection, for intrathecal use. 2020. . Accessed June 30, 2020.
    1. Hua Y, Sahashi K, Hung G, et al. Antisense correction of SMN2 splicing in the CNS rescues necrosis in a type III SMA mouse model. Genes Dev. 2010;24:1634‐1644.
    1. Passini MA, Bu J, Richards AM, et al. Antisense oligonucleotides delivered to the mouse CNS ameliorate symptoms of severe spinal muscular atrophy. Sci Transl Med. 2011;3:72ra18.
    1. Chiriboga CA, Swoboda KJ, Darras BT, et al. Results from a phase 1 study of nusinersen (ISIS‐SMNRx) in children with spinal muscular atrophy. Neurology. 2016;86:890‐897.
    1. Finkel RS, Chiriboga CA, Vajsar J, et al. Treatment of infantile‐onset spinal muscular atrophy with nusinersen: a phase 2, open‐label, dose‐escalation study. Lancet. 2016;388:3017‐3026.
    1. Finkel RS, Mercuri E, Darras BT, et al. ENDEAR Study Group. Nusinersen versus sham control in infantile‐onset spinal muscular atrophy. N Engl J Med. 2017;377:1723‐1732.
    1. Mercuri E, Darras BT, Chiriboga CA, et al. Nusinersen versus sham control in later‐onset spinal muscular atrophy. N Engl J Med. 2018;378:625‐635.
    1. De Vivo DC, Bertini E, Swoboda KJ, et al. NURTURE Study Group. Nusinersen initiated in infants during the presymptomatic stage of spinal muscular atrophy: interim efficacy and safety results from the phase 2 NURTURE study. Neuromuscul Disord. 2019;29:842‐856.
    1. Harada Y, Sutomo R, Sadewa AH, et al. Correlation between SMN2 copy number and clinical phenotype of spinal muscular atrophy: three SMN2 copies fail to rescue some patients from the disease severity. J Neurol. 2002;249:1211‐1219.
    1. Matsuzawa J, Matsui M, Konishi T, et al. Age‐related volumetric changes of brain gray and white matter in healthy infants and children. Cereb Cortex. 2001;11:335‐342.
    1. Darras BT, Crawford TO , Finkel RS, et al. Neurofilament as a potential biomarker for spinal muscular atrophy. Ann Clin Transl Neurol. 2019;6:932‐944.
    1. Frisone MF, Mercuri E, Laroche S, et al. Prognostic value of the neurologic optimality score at 9 and 18 months in preterm infants born before 31 weeks' gestation. J Pediatr. 2002;140:57‐60.
    1. Bishop KM, Montes J, Finkel RS. Motor milestone assessment of infants with spinal muscular atrophy using the Hammersmith Infant Neurological Exam—part 2: experience from a nusinersen clinical study. Muscle Nerve. 2018;57:142‐146.
    1. Haataja L, Mercuri E, Regev R, et al. Optimality score for the neurologic examination of the infant at 12 and 18 months of age. J Pediatr. 1999;135:153‐161.
    1. World Health Organization . WHO child growth standards. 2006. . Accessed June 29, 2020.
    1. Busner J, Targum SD. The clinical global impressions scale: applying a research tool in clinical practice. Psychiatry (Edgmont). 2007;4:28‐37.
    1. Haché M, Swoboda KJ, Sethna N, et al. Intrathecal injections in children with spinal muscular atrophy: nusinersen clinical trial experience. J Child Neurol. 2016;31:899‐906.
    1. Ebinger F, Kosel C, Pietz J, Rating D. Headache and backache after lumbar puncture in children and adolescents: a prospective study. Pediatrics. 2004;113:1588‐1592.
    1. Morgenlander JC. Lumbar puncture and CSF examination. Answers to three commonly asked questions. Postgrad Med. 1994;95:125‐128.
    1. Crooke ST, Baker BF, Witztum JL, et al. The effects of 2'‐O‐methoxyethyl containing antisense oligonucleotides on platelets in human clinical trials. Nucleic Acid Ther. 2017;27:121‐129.
    1. Crooke ST, Baker BF, Pham NC, et al. The effects of 2'‐O‐methoxyethyl oligonucleotides on renal function in humans. Nucleic Acid Ther. 2018;28:10‐22.

Source: PubMed

3
Abonner