One Year of Newborn Screening for SMA - Results of a German Pilot Project

Katharina Vill, Heike Kölbel, Oliver Schwartz, Astrid Blaschek, Bernhard Olgemöller, Erik Harms, Siegfried Burggraf, Wulf Röschinger, Jürgen Durner, Dieter Gläser, Uta Nennstiel, Brunhilde Wirth, Ulrike Schara, Beate Jensen, Marc Becker, Katharina Hohenfellner, Wolfgang Müller-Felber, Katharina Vill, Heike Kölbel, Oliver Schwartz, Astrid Blaschek, Bernhard Olgemöller, Erik Harms, Siegfried Burggraf, Wulf Röschinger, Jürgen Durner, Dieter Gläser, Uta Nennstiel, Brunhilde Wirth, Ulrike Schara, Beate Jensen, Marc Becker, Katharina Hohenfellner, Wolfgang Müller-Felber

Abstract

Objective: Spinal muscular atrophy (SMA) is the most common neurodegenerative disease in childhood. The study was conducted to assess the impact of early detection of SMA by newborn screening (NBS) on the clinical course of the disease.

Methods: Screening was performed in two federal states of Germany, Bavaria and North Rhine Westphalia, between January 2018 and February 2019. The incidence in the screening population was calculated as number of detected patients with a homozygous deletion in the SMN1-gene per number of screened patients. To get an idea about the incidence of newly diagnosed SMA in the year prior to screening a survey covering all neuropediatric centers in the state of Bavaria was conducted, identifying all SMA-cases in 2017 and 2018. Following positive NBS and confirmatory diagnostic test, treatment was advised according to the recommendations of the "American SMA NBS Multidisciplinary Working Group". Immediate treatment with Nusinersen was recommended in children with 2 and 3 SMN2 copies and a conservative strict follow-up strategy in children with ≥4 copies. All children underwent regular standardized neuropediatric examination, CHOP INTEND and HINE-2 testing as well as electrophysiological exams every 2-3 months.

Results: 165,525 children were screened. 22 cases of SMA were identified, meaning an incidence rate of 1:7524. SMN2 copy number analysis showed 2 SMN2 copies in 45% of patients, 3 SMN2 copies in 19 % and 4 SMN2 copies in 36%. These findings are confirmed in the most recent statistical data-cut from 31st August 2019 (incidence 1:7089, 2 SMN2 copies in 44%, 3 in 15% and 4 in 38%). Comparison with up-to-date German data on SMA incidence and the Bavarian survey give evidence that NBS did not lead to a relevant increase in incidence. 10 patients with 2 or 3 SMN2 copies were treated with Nusinersen, starting between 15- 39 days after birth, in 7/10 patients before onset of symptoms. Presymptomatically treated patients (age at last examination: 1- 12 months, median 8 months) showed no muscle weakness by the age of one month to one year. One child with 4 SMN2 copies became symptomatic at the age of 8 months.

Conclusions: Newborn screening, resulting in presymptomatic treatment, improves outcome in children with genetically proven SMA. Newborn screening for SMA should be introduced in all countries where therapy is available. An immediate therapy in cases with 4 SMN2 copies should be considered.

Keywords: Nusinersen; SMA treatment; Spinal muscular atrophy; newborn screening; prognosis.

Conflict of interest statement

1. Serving on a scientific advisory board or data safety monitoring board.

2. Gifts (other than travel or compensation for consulting or for educational efforts) worth more than USD $1000.

3. Funding for travel or speaker honoraria to the individual from a commercial or non-profit entity not included in the study funding [Exclude CME activities and Grand Rounds].

4. Serving as a journal editor, an associate editor, or editorial advisory board member. This may include a journal published by your national medical/scientific organization. Please include regardless of whether you receive compensation.

5. Patents issued or pending.

6. Publishing Royalties (do not include honoraria for occasional writing).

7. Employment. If you are currently employed by a commercial entity, please disclose below. In addition, if your past employment at a commercial entity is directly related to this manuscript, please disclose below.

8. Consultancies.

9. Speakers’ bureau.

10. Other activities not covered in designations above (if in doubt, provide full disclosure).

11. Some published work has potential for financial gain for the study investigators or the sponsor. The following question seeks to provide transparency regarding any financial benefits to investigators or sponsors.

Katharina Vill, Heike Kölbel, Oliver Schwartz, Astrid Blaschek, Bernhard Olgemöller, Erik Harms, Uta Nennstiel, and Beate Jensen have nothing to declare.

Siegfried Burggraf, Wulf Röschinger, Jürgen Durner and Marc Becker are employed by/owner of a commercial entity (Laboratory Becker and colleagues MVZ GbR, Führichstraße 70, 81871 München, Germany).

Dieter Gläser: employed by/owner of a commercial entity (Genetikum®, Wegenerstr. 15, 89231 Neu-Ulm, Germany).

Ulrika Schara is serving on a scientific advisory board or data safety monitoring board for Biogen, Avexis and Novartis.

Brunhilde Wirth is serving on a scientific advisory board or data safety monitoring board for SMA Europe and received travel and speaker honoraria from Biogen.

Katharina Hohenfellner received commercial travel support and speaker honoraria from Ortphan Europe, Chiesi, and non-profit travel support from Cystinosis Foundation and Nephie.

Wolfgang Müller-Felber is serving on a scientific advisory board or data safety monitoring board for Biogen, Avexis, PTC, Sanofi-Aventis and Cytokinetics and received travel and speaker honoraria from Biogen, Avexis, PTC and Sanofi-Aventis.

Figures

Fig. 1
Fig. 1
CHOP-INTEND a) patients with 2 SMN2 copies b) patients with 3 SMN2 copies c) patients with 4 SMN2 copies.
Fig. 2
Fig. 2
HINE-2 a) patients with 2 SMN2 copies b) patients with 3 SMN2 copies c) patients with 4 SMN2 copies.
Fig. 3
Fig. 3
ulnar nerve CMAPs a) patients with 2 SMN2 copies b) patients with 3 SMN2 copies c) patients with 4 SMN2 copies.

References

    1. Sugarman EA, Nagan N, Zhu H, Akmaev VR, Zhou Z, Rohlfs EM, et al. Pan-ethnic carrier screening and prenatal diagnosis for spinal muscular atrophy: Clinical laboratory analysis of >72,400 specimens. European Journal of Human Genetics: EJHG. 2012;20(1):27–32.
    1. Wang CH, Finkel RS, Bertini ES, Schroth M, Simonds A, Wong B, et al. Consensus statement for standard of care in spinal muscular atrophy. Journal of Child Neurology. 2007;22(8):1027–49.
    1. Mercuri E, Finkel RS, Muntoni F, Wirth B, Montes J, Main M, et al. Diagnosis and management of spinal muscular atrophy: Part Recommendations for diagnosis, rehabilitation, orthopedic and nutritional care. Neuromuscular Disorders: NMD. 2018;28(2):103–15.
    1. Finkel RS, Mercuri E, Meyer OH, Simonds AK, Schroth MK, Graham RJ, et al. Diagnosis and management of spinal muscular atrophy: Part 2: Pulmonary and acute care; medications, supplements and immunizations; other organ systems; and ethics. Neuromuscular disorders: NMD. 2018;28(3):197–207.
    1. Lefebvre S, Burglen L, Reboullet S, Clermont O, Burlet P, Viollet L, et al. Identification and characterization of a spinal muscular atrophy-determining gene. Cell. 1995;80(1):155–65.
    1. Shorrock HK, Gillingwater TH, Groen EJN. Overview of current drugs and molecules in development for spinal muscular atrophy therapy. Drugs. 2018;78(3):293–305.
    1. Lorson CL, Hahnen E, Androphy EJ, Wirth B. A single nucleotide in the SMN gene regulates splicing and is responsible for spinal muscular atrophy. Proc Natl Acad Sci U S A. 1999;96(11):6307–11.
    1. Helmken C, Hofmann Y, Schoenen F, Oprea G, Raschke H, Rudnik-Schoneborn S, et al. Evidence for a modifying pathway in SMA discordant families: Reduced SMN level decreases the amount of its interacting partners and Htra2-beta1. Hum Genet. 2003;114(1):11–21.
    1. Wirth B, Garbes L, Riessland M. How genetic modifiers influence the phenotype of spinal muscular atrophy and suggest future therapeutic approaches. Current Opinion in Genetics & Development. 2013;23(3):330–8.
    1. Schrank B, Gotz R, Gunnersen JM, Ure JM, Toyka KV, Smith AG, et al. Inactivation of the survival motor neuron gene, a candidate gene for human spinal muscular atrophy, leads to massive cell death in early mouse embryos. Proc Natl Acad Sci U S A. 1997;94(18):9920–5.
    1. Foust KD, Wang X, McGovern VL, Braun L, Bevan AK, Haidet AM, et al. Rescue of the spinal muscular atrophy phenotype in a mouse model by early postnatal delivery of SMN. Nature Biotechnology. 2010;28(3):271–4.
    1. Passini MA, Bu J, Richards AM, Kinnecom C, Sardi SP, Stanek LM, et al. Antisense oligonucleotides delivered to the mouse CNS ameliorate symptoms of severe spinal muscular atrophy. Sci Transl Med. 2011;3(72):72ra18.
    1. Hua Y, Sahashi K, Rigo F, Hung G, Horev G, Bennett CF, et al. Peripheral SMN restoration is essential for long-term rescue of a severe spinal muscular atrophy mouse model. Nature. 2011;478(7367):123–6.
    1. Finkel RS, Chiriboga CA, Vajsar J, Day JW, Montes J, De Vivo DC, et al. Treatment of infantile-onset spinal muscular atrophy with nusinersen: A phase 2, open-label, dose-escalation study. Lancet (London, England). 2017;388(10063):3017–26.
    1. Mercuri E, Darras BT, Chiriboga CA, Day JW, Campbell C, Connolly AM, et al. Nusinersen versus sham control in later-onset spinal muscular atrophy. The New England Journal of Medicine. 2018;378(7):625–35.
    1. Mendell JR, Al-Zaidy S, Shell R, Arnold WD, Rodino-Klapac LR, Prior TW, et al. Single-dose gene-replacement therapy for spinal muscular atrophy. The New England Journal of Medicine. 2017;377(18):1713–22.
    1. Hua Y, Sahashi K, Hung G, Rigo F, Passini MA, Bennett CF, et al. Antisense correction of SMN2 splicing in the CNS rescues necrosis in a type III SMA mouse model. Genes Dev. 2010;24(15):1634–44.
    1. Talbot K, Tizzano EF. The clinical landscape for SMA in a new therapeutic era. Gene Therapy. 2017;24(9):529–33.
    1. Tizzano EF, Finkel RS. Spinal muscular atrophy: A changing phenotype beyond the clinical trials. Neuromuscular Disorders: NMD: 2017.
    1. Kolb SJ, Coffey CS, Yankey JW, Krosschell K, Arnold WD, Rutkove SB, et al. Natural history of infantile-onset spinal muscular atrophy. Annals of Neurology. 2017;82(6):883–91.
    1. Fingerhut R, Olgemoller B. Newborn screening for inborn errors of metabolism and endocrinopathies: An update. Anal Bioanal Chem. 2009;393(5):1481–97.
    1. De Vivo DC. Data from the Nurture Study (Biogen), presented at the MDA meeting in Arlington. 2018.
    1. Parsons J De Vivo DC BE, Hwu W-L, Crawford TO, Swoboda KJ, Finkel RS, Kirschner J, Kuntz N, Ryan MM, Butterfield RJ, Topaloglu H, Ben Omran T, 14 Sansone VA, Jong Y-J, Shu F, Reyna SP, Johnson K, Foster R, Bhan I, Fradette S, Farwell W, on behalf of the NURTURE Study Investigators. Nusinersen in Infants Who Initiate Treatment in a Presymptomatic Stage of Spinal Muscular Atrophy (SMA): Interim Efficacy and Safety Results From the Phase 2 NURTURE Study. 23rd Annual Spinal Muscular Atrophy Researcher Meeting, Anaheim, CA. 2019.
    1. Czibere L, Burggraf S, Fleige T, Gluck B, Keitel LM, Landt O, et al. High-throughput genetic newborn screening for spinal muscular atrophy by rapid nucleic acid extraction from dried blood spots and 384-well qPCR. European Journal of Human Genetics: EJHG. 2019.
    1. Arkblad EL, Darin N, Berg K, Kimber E, Brandberg G, Lindberg C, et al. Multiplex ligation-dependent probe amplification improves diagnostics in spinal muscular atrophy. Neuromuscular Disorders: NMD. 2006;16(12):830–8.
    1. Glascock J, Sampson J, Haidet-Phillips A, Connolly A, Darras B, Day J, et al. Treatment algorithm for infants diagnosed with spinal muscular atrophy through newborn screening. J Neuromuscul Dis. 2018.
    1. Glanzman AM, Mazzone E, Main M, Pelliccioni M, Wood J, Swoboda KJ, et al. The Children’s Hospital of Philadelphia Infant Test of Neuromuscular Disorders (CHOP INTEND): Test development and reliability. Neuromuscular Disorders: NMD. 2010;20(3):155–61.
    1. Bishop KM, Montes J, Finkel RS. Motor milestone assessment of infants with spinal muscular atrophy using the hammersmith infant neurological Exam-Part Experience from a nusinersen clinical study. Muscle & Nerve. 2018;57(1):142–6.
    1. Verhaart IEC, Robertson A, Wilson IJ, Aartsma-Rus A, Cameron S, Jones CC, et al. Prevalence, incidence and carrier frequency of 5q-linked spinal muscular atrophy - a literature review. Orphanet Journal of Rare Diseases. 2017;12(1):124.
    1. Jedrzejowska M, Szczaluba K, Sielska D. Homozygous deletion in the SMN1 gene in asymptomatic individual - genetic counselling issues in SMA-risk families. Medycyna Wieku Rozwojowego. 2011;15(2):126–31.
    1. Hahnen E, Forkert R, Marke C, Rudnik-Schoneborn S, Schonling J, Zerres K, et al. Molecular analysis of candidate genes on chromosome 5q13 in autosomal recessive spinal muscular atrophy: Evidence of homozygous deletions of the SMN gene in unaffected individuals. Human Molecular Genetics. 1995;4(10):1927–33.
    1. Riessland M, Kaczmarek A, Schneider S, Swoboda KJ, Lohr H, Bradler C, et al. Neurocalcin delta suppression protects against spinal muscular atrophy in humans and across species by restoring impaired endocytosis. American Journal of Human Genetics. 2017;100(2):297–315.
    1. Wang CH, Xu J, Carter TA, Ross BM, Dominski MK, Bellcross CA, et al. Characterization of survival motor neuron (SMNT) gene deletions in asymptomatic carriers of spinal muscular atrophy. Human Molecular Genetics. 1996;5(3):359–65.
    1. Cobben JM, van der Steege G, Grootscholten P, de Visser M, Scheffer H, Buys CH. Deletions of the survival motor neuron gene in unaffected siblings of patients with spinal muscular atrophy. American Journal of Human Genetics. 1995;57(4):805–8.
    1. Oprea GE, Krober S, McWhorter ML, Rossoll W, Muller S, Krawczak M, et al. Plastin 3 is a protective modifier of autosomal recessive spinal muscular atrophy. Science. 2008;320(5875):524–7.
    1. Heesen L, Peitz M, Torres-Benito L, Holker I, Hupperich K, Dobrindt K, et al. Plastin 3 is upregulated in iPSC-derived motoneurons from asymptomatic SMN1-deleted individuals. Cell Mol Life Sci. 2016;73(10):2089–104.
    1. Feldkotter 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. American Journal of Human Genetics. 2002;70(2):358–68.
    1. Jedrzejowska M, Milewski M, Zimowski J, Zagozdzon P, Kostera-Pruszczyk A, Borkowska J, et al. Incidence of spinal muscular atrophy in Poland–more frequent than predicted? Neuroepidemiology. 2010;34(3):152–7.
    1. Wirth B. An update of the mutation spectrum of the survival motor neuron gene (SMN1) in autosomal recessive spinal muscular atrophy (SMA). Human Mutation. 2000;15(3):228–37.
    1. Chien YH, Chiang SC, Weng WC, Lee NC, Lin CJ, Hsieh WS, et al. Presymptomatic diagnosis of spinal muscular atrophy through newborn screening. The Journal of Pediatrics. 2017.
    1. Kraszewski JN, Kay DM, Stevens CF, Koval C, Haser B, Ortiz V, et al. Pilot study of population-based newborn screening for spinal muscular atrophy in New York state. Genet Med. 2018;20(6):608–13.
    1. Calucho M, Bernal S, Alias L, March F, Vencesla A, Rodriguez-Alvarez FJ, et al. Correlation between SMA type and SMN2 copy number revisited: An analysis of 625 unrelated Spanish patients and a compilation of reported cases. Neuromuscular Disorders: NMD. 2018;28(3):208–15.
    1. Prior TW, Krainer AR, Hua Y, Swoboda KJ, Snyder PC, Bridgeman SJ, et al. A positive modifier of spinal muscular atrophy in the SMN2 gene. American Journal of Human Genetics. 2009;85(3):408–13.
    1. Wirth B, Brichta L, Schrank B, Lochmuller H, Blick S, Baasner A, et al. Mildly affected patients with spinal muscular atrophy are partially protected by an increased SMN2 copy number. Hum Genet. 2006;119(4):422–8.
    1. Cusco I, Barcelo MJ, Rojas-Garcia R, Illa I, Gamez J, Cervera C, et al. SMN2 copy number predicts acute or chronic spinal muscular atrophy but does not account for intrafamilial variability in siblings. Journal of Neurology. 2006;253(1):21–5.
    1. Ogino S, Gao S, Leonard DG, Paessler M, Wilson RB. Inverse correlation between SMN1 and SMN2 copy numbers: Evidence for gene conversion from SMN2 to SMN1. European Journal of Human Genetics: EJHG. 2003;11(3):275–7.
    1. Schorling DC, Becker J, Pechmann A, Langer T, Wirth B, Kirschner J. Discrepancy in redetermination of SMN2 copy numbers in children with SMA. Neurology. 2019;93(6):267–9.
    1. Finkel RS, Chiriboga CA, Vajsar J, Day JW, Montes J, De Vivo DC, et al. Treatment of infantile-onset spinal muscular atrophy with nusinersen: A phase 2, open-label, dose-escalation study. Lancet (London, England). 2016;388(10063):3017–26.
    1. Pane M, Palermo C, Messina S, Sansone VA, Bruno C, Catteruccia M, et al. Nusinersen in type 1 SMA infants, children and young adults: Preliminary results on motor function. Neuromuscular Disorders: NMD. 2018;28(7):582–5.
    1. Verde F, Steinacker P, Weishaupt JH, Kassubek J, Oeckl P, Halbgebauer S, et al. Neurofilament light chain in serum for the diagnosis of amyotrophic lateral sclerosis. Journal of Neurology, Neurosurgery, and Psychiatry. 2019;90(2):157–64.
    1. Lin CW, Kalb SJ, Yeh WS. Delay in diagnosis of spinal muscular atrophy: A systematic literature review. Pediatric Neurology. 2015.
    1. Kariya S, Obis T, Garone C, Akay T, Sera F, Iwata S, et al. Requirement of enhanced Survival Motoneuron protein imposed during neuromuscular junction maturation. The Journal of Clinical Investigation. 2014;124(2):785–800.
    1. Wolf B. Successful outcomes of older adolescents and adults with profound biotinidase deficiency identified by newborn screening. Genet Med. 2017;19(4):396–402.
    1. Mlcoch T, Puda R, Jesina P, Lhotakova M, Sterbova S, Dolezal T. Dietary patterns, cost and compliance with low-protein diet of phenylketonuria and other inherited metabolic diseases. Eur J Clin Nutr. 2018;72(1):87–92.
    1. Scoto M, Finkel RS, Mercuri E, Muntoni F. Therapeutic approaches for spinal muscular atrophy (SMA). Gene Therapy. 2017;24(9):514–9.

Source: PubMed

3
Subskrybuj