Nusinersen initiated in infants during the presymptomatic stage of spinal muscular atrophy: Interim efficacy and safety results from the Phase 2 NURTURE study

Darryl C De Vivo, Enrico Bertini, Kathryn J Swoboda, Wuh-Liang Hwu, Thomas O Crawford, Richard S Finkel, Janbernd Kirschner, Nancy L Kuntz, Julie A Parsons, Monique M Ryan, Russell J Butterfield, Haluk Topaloglu, Tawfeg Ben-Omran, Valeria A Sansone, Yuh-Jyh Jong, Francy Shu, John F Staropoli, Douglas Kerr, Alfred W Sandrock, Christopher Stebbins, Marco Petrillo, Gabriel Braley, Kristina Johnson, Richard Foster, Sarah Gheuens, Ishir Bhan, Sandra P Reyna, Stephanie Fradette, Wildon Farwell, NURTURE Study Group, Darryl C De Vivo, Enrico Bertini, Kathryn J Swoboda, Wuh-Liang Hwu, Thomas O Crawford, Richard S Finkel, Janbernd Kirschner, Nancy L Kuntz, Julie A Parsons, Monique M Ryan, Russell J Butterfield, Haluk Topaloglu, Tawfeg Ben-Omran, Valeria A Sansone, Yuh-Jyh Jong, Francy Shu, John F Staropoli, Douglas Kerr, Alfred W Sandrock, Christopher Stebbins, Marco Petrillo, Gabriel Braley, Kristina Johnson, Richard Foster, Sarah Gheuens, Ishir Bhan, Sandra P Reyna, Stephanie Fradette, Wildon Farwell, NURTURE Study Group

Abstract

Spinal muscular atrophy (SMA) is a neurodegenerative disease associated with severe muscle atrophy and weakness in the limbs and trunk. We report interim efficacy and safety outcomes as of March 29, 2019 in 25 children with genetically diagnosed SMA who first received nusinersen in infancy while presymptomatic in the ongoing Phase 2, multisite, open-label, single-arm NURTURE trial. Fifteen children have two SMN2 copies and 10 have three SMN2 copies. At last visit, children were median (range) 34.8 [25.7-45.4] months of age and past the expected age of symptom onset for SMA Types I or II; all were alive and none required tracheostomy or permanent ventilation. Four (16%) participants with two SMN2 copies utilized respiratory support for ≥6 h/day for ≥7 consecutive days that was initiated during acute, reversible illnesses. All 25 participants achieved the ability to sit without support, 23/25 (92%) achieved walking with assistance, and 22/25 (88%) achieved walking independently. Eight infants had adverse events considered possibly related to nusinersen by the study investigators. These results, representing a median 2.9 years of follow up, emphasize the importance of proactive treatment with nusinersen immediately after establishing the genetic diagnosis of SMA in presymptomatic infants and emerging newborn screening efforts.

Keywords: Clinical trial; Neurofilament; Newborn screening; Nusinersen; Presymptomatic; Spinal muscular atrophy.

Copyright © 2019 The Authors. Published by Elsevier B.V. All rights reserved.

Figures

Fig. 1
Fig. 1
NURTURE study design. Intention-to-treat population is all infants who received ≥1 dose of study drug (n= 25). aInfants who attended or had the opportunity to attend the visit. bInfants treated with nusinersen 12 mg; some infants received a 12-mg scaled equivalent dose before the protocol was revised in March 2017.
Fig. 2
Fig. 2
Kaplan–Meier plot for age at death or respiratory intervention.a SMN2, survival motor neuron 2. No participants have died or required tracheostomy or permanent ventilation (defined as ≥16 h/day continuously for >21 days in the absence of an acute reversible event or tracheostomy). aRespiratory intervention was defined as ventilator use for ≥6 h per day for ≥7 days or tracheostomy.
Fig. 3
Fig. 3
Site- or caregiver-reporteda age at first WHO motor milestone achievement. SMN2, survival motor neuron 2; WHO, World Health Organization. aIf caregiver-reported, achievement was confirmed by the study site at the next study visit with a yes or no response. bWHO motor milestone windows of achievement were determined based on the WHO Multicenter Growth Reference Study windows of achievement in healthy children .
Fig. 4
Fig. 4
Mean HINE-2 motor milestone scores over time. HINE-2, Hammersmith Infant Neurologic Examination, Section 2; SE, standard error; SMN2, survival motor neuron 2. Time points with n ≥ 5 included. HINE-2 score was assessed in NURTURE participants up until the Day 778 study visit.
Fig. 5
Fig. 5
(A) Mean CHOP INTEND score over time, (B) Kaplan–Meier plot for time to first achievement of maximum CHOP INTEND score of 64 points.a CHOP INTEND, Children's Hospital of Philadelphia Infant Test of Neuromuscular Disorders; SE, standard error; SMN2, survival motor neuron 2. For Fig. 5(A), time points with n ≥ 5 included. aIn the original protocol, CHOP INTEND was to be assessed in participants at each visit up to Day 778; however, this was amended to be until they had a maximum score of 64. Once a score of 64 was achieved, CHOP INTEND was no longer assessed.
Fig. 6
Fig. 6
(A) Plasma pNF-H levels at baseline in NURTURE infants and infants a CI, confidence interval; ELLA, enzyme-linked lectin assay; pNF-H, phosphorylated neurofilament heavy chain; SMA, spinal muscular atrophy; SMN2, survival motor neuron 2. pNF-H levels were evaluated using a pNF-H ELLA from ProteinSimple. 7.46 pg/mL was used as the imputed value if the pNF-H concentration was below the limit of quantification. Baseline pNF-H values in NURTURE infants were obtained on Study Visit Day 1, either prior to nusinersen administration or four h post-dose. Samples from infants without SMA were provided by Boston Children's Hospital. aTime points with n ≥ 5 included. The number of decimal places reported in summary statistics is not indicative of biomarker assay precision or sensitivity. In Panel A, some data points have an x-value offset of +/−0.2 months for better visualization.
Fig. 7
Fig. 7
Plasma pNF-H levels at Day 64 and achievement of WHO motor milestone of walking alone pNF-H, phosphorylated neurofilament heavy chain; SMN2, survival motor neuron 2; WHO, World Health Organization. 1WHO 99th percentile for age of achievement for development in healthy children .

References

    1. Darras B.T., Monani U.R., De Vivo D.C. Genetic disorders affecting the motor neuron: spinal muscular atrophy. In: Swaiman KF, editor. Swaiman's pediatric neurology: principles and practice. Elsevier; Edinburgh: 2017. pp. 1057–1064.
    1. Mercuri E., Bertini E., Iannaccone S.T. Childhood spinal muscular atrophy: controversies and challenges. Lancet Neurol. 2012;11:443–452.
    1. Markowitz J.A., Singh P., Darras B.T. Spinal muscular atrophy: a clinical and research update. Pediatr Neurol. 2012;46:1–12.
    1. Farrar M.A., Kiernan M.C. The genetics of spinal muscular atrophy: progress and challenges. Neurotherapeutics. 2015;12:290–302.
    1. Kubo Y., Nishio H., Saito K. A new method for SMN1 and hybrid SMN gene analysis in spinal muscular atrophy using long-range PCR followed by sequencing. J Hum Genet. 2015;60:233–239.
    1. Finkel R., Bertini E., Muntoni F., Mercuri E. 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. Talbot K., Tizzano E.F. The clinical landscape for SMA in a new therapeutic era. Gene Ther. 2017;24:529–533.
    1. Finkel R.S., Mercuri E., Darras B.T., Connolly A.M., Kuntz N.L., Kirschner J. Nusinersen versus sham control in infantile-onset spinal muscular atrophy. N Engl J Med. 2017;377:1723–1732.
    1. Mercuri E., Darras B.T., Chiriboga C.A., Day J.W., Campbell C., Connolly A.M. Nusinersen versus sham control in later-onset spinal muscular atrophy. N Engl J Med. 2018;378:625–635.
    1. Calucho M., Bernal S., Alías L., March F., Venceslá A., Rodríguez-Álvarez F.J. Correlation between SMA type and SMN2 copy number revisited: an analysis of 625 unrelated Spanish patients and a compilation of 2834 reported cases. Neuromuscul Disord. 2018;28:208–215.
    1. Feldkötter M., Schwarzer V., Wirth R., Wienker T.F., 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. Crawford T., Sumner C., Finkel R., De Vivo D.C., Oskoui M., Tizzano E. Phosphorylated neurofilament heavy chain (pNF-H) levels in infants and children with SMA: evaluation of pNF-H as a potential biomarker of sma disease activity. Neuromuscul Disord. 2018;28:S110–S111.
    1. Darras B., Finkel R., Mercuri E., Sumner C., Oskoui M., Tizzano E. Association of phosphorylated neurofilament heavy chain (pNF-H) with nusinersen treatment of SMA: analyses from the ENDEAR and CHERISH studies. Neuromuscul Disord. 2018;28(Suppl 2):S31.
    1. Yuan A., Rao M.V., Veeranna, Nixon R.A. Neurofilaments and neurofilament proteins in health and disease. Cold Spring Harb Perspect Biol. 2017;9
    1. Petzold A. Neurofilament phosphoforms: surrogate markers for axonal injury, degeneration and loss. J Neurol Sci. 2005;233:183–198.
    1. El-Hattab A.W., Almannai M., Sutton V.R. Newborn screening: history, current status, and future directions. Pediatr Clin North Am. 2018;65:389–405.
    1. Taylor J.L., Lee F.K., Yazdanpanah G.K., Staropoli J.F., Liu M., Carulli J.P. Newborn blood spot screening test using multiplexed real-time PCR to simultaneously screen for spinal muscular atrophy and severe combined immunodeficiency. Clin Chem. 2015;61:412–419.
    1. Chien Y.H., Chiang S.C., Weng W.C., Lee N.C., Lin C.J., Hsieh W.S. Presymptomatic diagnosis of spinal muscular atrophy through newborn screening. J Pediatr. 2017;190:124–129.
    1. Kraszewski J.N., Kay D.M., Stevens C.F., Koval C., Haser B., Ortiz V. Pilot study of population-based newborn screening for spinal muscular atrophy in New York state. Genet Med. 2018;20:608–613.
    1. Lopes JM. SMA added to list of recommended screenings for disease given to newborns in US. 2018[cited February 5, 2019]; Available from: .
    1. Hua Y., Sahashi K., Hung G., Rigo F., Passini M.A., Bennett C.F. 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 M.A., Bu J., Richards A.M., Kinnecom C., Sardi S.P., Stanek L.M. Antisense oligonucleotides delivered to the mouse CNS ameliorate symptoms of severe spinal muscular atrophy. Sci Transl Med. 2011;3:72ra18.
    1. Chiriboga C.A., Swoboda K.J., Darras B.T., Iannaccone S.T., Montes J., De Vivo D.C. Results from a Phase 1 study of nusinersen (ISIS-SMNRx) in children with spinal muscular atrophy. Neurology. 2016;86:890–897.
    1. Hoy S.M. Nusinersen: first global approval. Drugs. 2017;77:473–479.
    1. Finkel R.S., Chiriboga C.A., Vajsar J., Day J.W., Montes J., De Vivo D.C. Treatment of infantile-onset spinal muscular atrophy with nusinersen: a Phase 2, open-label, dose-escalation study. Lancet. 2016;388:3017–3026.
    1. Jones C.C., Cook S.F., Hobby K., Jarecki J. Presented at: 2016 Annual Spinal Muscular Atrophy Conference, June 16–19, 2016. Anaheim, CA; 2016. SMA subtype concordance in siblings: findings from the Cure SMA cohort. (unpublished results)
    1. Finkel R.S., McDermott M.P., Kaufmann P., Darras B.T., Chung W.K., Sproule D.M. Observational study of spinal muscular atrophy type I and implications for clinical trials. Neurology. 2014;83:810–817.
    1. WHO Multicentre Growth Reference Study Group. WHO Motor Development Study: windows of achievement for six gross motor development milestones. Acta Paediatr Suppl. 2006;450:86–95.
    1. Haataja L., Mercuri E., Regev R., Cowan F., Rutherford M., Dubowitz V. Optimality score for the neurologic examination of the infant at 12 and 18 months of age. J Pediatr. 1999;135:153–161.
    1. Glanzman A.M., Mazzone E., Main M., Pelliccioni M., Wood J., Swoboda K.J. The Children's Hospital of Philadelphia Infant Test of Neuromuscular Disorders (CHOP INTEND): test development and reliability. Neuromuscul Disord. 2010;20:155–161.
    1. Glanzman A.M., McDermott M.P., Montes J., Martens W.B., Flickinger J., Riley S. Validation of the Children's Hospital of Philadelphia Infant Test of Neuromuscular Disorders (CHOP INTEND) Pediatr Phys Ther. 2011;23:322–326.
    1. Lewelt A., Krosschell K.J., Scott C., Sakonju A., Kissel J.T., Crawford T.O. Compound muscle action potential and motor function in children with spinal muscular atrophy. Muscle Nerve. 2010;42:703–708.
    1. Swoboda K.J., Prior T.W., Scott C.B., McNaught T.P., Wride M.C., Reyna S.P. Natural history of denervation in SMA: relation to age, SMN2 copy number, and function. Ann Neurol. 2005;57:704–712.
    1. Julious S.A. Two-sided confidence intervals for the single proportion: comparison of seven methods by Robert G. Newcombe, Statistics in Medicine. 1998;17:857–872. . Stat Med 2005;24:3383-4.
    1. World Health Organization. Child growth standards. [cited December 16, 2018]; Available from:.
    1. . Study to evaluate sodium phenylbutyrate in pre-symptomatic infants with spinal muscular atrophy (STOPSMA). 2015[cited February 5, 2019]; NCT00528268]. Available from:.
    1. Kolb S.J., Coffey C.S., Yankey J.W., Krosschell K., Arnold W.D., Rutkove S.B. Natural history of infantile-onset spinal muscular atrophy. Ann Neurol. 2017;82:883–891.
    1. Boardman F.K., Sadler C., Young P.J. Newborn genetic screening for spinal muscular atrophy in the UK: the views of the general population. Mol Genet Genomic Med. 2018;6:99–108.
    1. Lin P.J., Yeh W.S., Neumann P.J. Willingness to pay for a newborn screening test for spinal muscular atrophy. Pediatr Neurol. 2017;66:69–75.
    1. Glascock J., Sampson J., Haidet-Phillips A., Connolly A., Darras B., Day J. Treatment algorithm for infants diagnosed with spinal muscular atrophy through newborn screening. J Neuromuscul Dis. 2018;5:145–158.
    1. Darras B.T., De Vivo D.C. Precious SMA natural history data: a benchmark to measure future treatment successes. Neurology. 2018;91:337–339.
    1. Benatar M., Wuu J. Presymptomatic studies in ALS: rationale, challenges, and approach. Neurology. 2012;79:1732–1739.
    1. Eisen A., Kiernan M., Mitsumoto H., Swash M. Amyotrophic lateral sclerosis: a long preclinical period? J Neurol Neurosurg Psychiatry. 2014;85:1232–1238.
    1. Paulsen J.S., Langbehn D.R., Stout J.C., Aylward E., Ross C.A., Nance M. Detection of Huntington's disease decades before diagnosis: the Predict-HD study. J Neurol Neurosurg Psychiatry. 2008;79:874–880.
    1. Sperling R.A., Aisen P.S., Beckett L.A., Bennett D.A., Craft S., Fagan A.M. Toward defining the preclinical stages of Alzheimer's disease: recommendations from the National Institute on Aging-Alzheimer's Association workgroups on diagnostic guidelines for Alzheimer's disease. Alzheimers Dement. 2011;7:280–292.

Source: PubMed

3
Subscribe