Antisense oligonucleotide therapy reduces seizures and extends life span in an SCN2A gain-of-function epilepsy model.

De novo variation in SCN2A can give rise to severe childhood disorders. Biophysical gain of function in SCN2A is seen in some patients with early seizure onset developmental and epileptic encephalopathy (DEE). In these cases, targeted reduction in SCN2A expression could substantially improve clinical outcomes. We tested this theory by central administration of a gapmer antisense oligonucleotide (ASO) targeting Scn2a mRNA in a mouse model of Scn2a early seizure onset DEE (Q/+ mice). Untreated Q/+ mice presented with spontaneous seizures at P1 and did not survive beyond P30. Administration of the ASO to Q/+ mice reduced spontaneous seizures and significantly extended life span. Across a range of behavioral tests, Scn2a ASO-treated Q/+ mice were largely indistinguishable from WT mice, suggesting treatment is well tolerated. A human SCN2A gapmer ASO could likewise impact the lives of patients with SCN2A gain-of-function DEE.

[1]  I. Aznarez,et al.  Antisense oligonucleotides increase Scn1a expression and reduce seizures and SUDEP incidence in a mouse model of Dravet syndrome , 2020, Science Translational Medicine.

[2]  Jacy L. Wagnon,et al.  Scn8a Antisense Oligonucleotide Is Protective in Mouse Models of SCN8A Encephalopathy and Dravet Syndrome , 2020, Annals of neurology.

[3]  Roy Ben-Shalom,et al.  The Autism-Associated Gene Scn2a Contributes to Dendritic Excitability and Synaptic Function in the Prefrontal Cortex , 2019, Neuron.

[4]  K. Yamakawa,et al.  Scn2a haploinsufficient mice display a spectrum of phenotypes affecting anxiety, sociability, memory flexibility and ampakine CX516 rescues their hyperactivity , 2019, Molecular Autism.

[5]  C. Bennett Therapeutic Antisense Oligonucleotides Are Coming of Age. , 2019, Annual review of medicine.

[6]  A. Ludolph,et al.  Intrathecal administration of nusinersen in adolescent and adult SMA type 2 and 3 patients , 2018, Journal of Neurology.

[7]  Y. Yanagawa,et al.  Nav1.2 haplodeficiency in excitatory neurons causes absence-like seizures in mice , 2018, Communications Biology.

[8]  S. Willard,et al.  A comprehensive institutional overview of intrathecal nusinersen injections for spinal muscular atrophy , 2018, Pediatric Radiology.

[9]  Stephan J Sanders,et al.  Progress in Understanding and Treating SCN2A-Mediated Disorders , 2018, Trends in Neurosciences.

[10]  I. Scheffer,et al.  Dynamic action potential clamp predicts functional separation in mild familial and severe de novo forms of SCN2A epilepsy , 2018, Proceedings of the National Academy of Sciences.

[11]  I. Scheffer,et al.  Ion Channels in Genetic Epilepsy: From Genes and Mechanisms to Disease-Targeted Therapies , 2018, Pharmacological Reviews.

[12]  R. J. Ramamurthi,et al.  Nusinersen versus Sham Control in Infantile‐Onset Spinal Muscular Atrophy , 2017, The New England journal of medicine.

[13]  C. Reid,et al.  Models for discovery of targeted therapy in genetic epileptic encephalopathies , 2017, Journal of neurochemistry.

[14]  H. Bui,et al.  Antisense oligonucleotides targeting translation inhibitory elements in 5′ UTRs can selectively increase protein levels , 2017, Nucleic acids research.

[15]  L. Lagae,et al.  Genetic and phenotypic heterogeneity suggest therapeutic implications in SCN2A-related disorders , 2017, Brain : a journal of neurology.

[16]  Edouard Hirsch,et al.  ILAE classification of the epilepsies: Position paper of the ILAE Commission for Classification and Terminology , 2017, Epilepsia.

[17]  David R Corey,et al.  Nusinersen, an antisense oligonucleotide drug for spinal muscular atrophy , 2017, Nature Neuroscience.

[18]  A. Krieg,et al.  FDA Approves Eteplirsen for Duchenne Muscular Dystrophy: The Next Chapter in the Eteplirsen Saga , 2017, Nucleic acid therapeutics.

[19]  Roy Ben-Shalom,et al.  Opposing Effects on NaV1.2 Function Underlie Differences Between SCN2A Variants Observed in Individuals With Autism Spectrum Disorder or Infantile Seizures , 2017, Biological Psychiatry.

[20]  R. Finkel,et al.  Treatment of infantile-onset spinal muscular atrophy with nusinersen: a phase 2, open-label, dose-escalation study , 2016, The Lancet.

[21]  S. Crooke,et al.  Translation efficiency of mRNAs is increased by antisense oligonucleotides targeting upstream open reading frames , 2016, Nature Biotechnology.

[22]  Jacqueline Montes,et al.  Results from a phase 1 study of nusinersen (ISIS-SMNRx) in children with spinal muscular atrophy , 2016, Neurology.

[23]  Shinichi Hirose,et al.  Retigabine, a Kv7.2/Kv7.3-Channel Opener, Attenuates Drug-Induced Seizures in Knock-In Mice Harboring Kcnq2 Mutations , 2016, PloS one.

[24]  V. Wimmer,et al.  'Neonatal' Nav1.2 reduces neuronal excitability and affects seizure susceptibility and behaviour. , 2015, Human molecular genetics.

[25]  Y. Hua,et al.  Pharmacology of a Central Nervous System Delivered 2′-O-Methoxyethyl–Modified Survival of Motor Neuron Splicing Oligonucleotide in Mice and Nonhuman Primates , 2014, The Journal of Pharmacology and Experimental Therapeutics.

[26]  K. Blomgren,et al.  Brain development in rodents and humans: Identifying benchmarks of maturation and vulnerability to injury across species , 2013, Progress in Neurobiology.

[27]  J. Shendure,et al.  Targeted resequencing in epileptic encephalopathies identifies de novo mutations in CHD2 and SYNGAP1 , 2013, Nature Genetics.

[28]  A. Guimond,et al.  Oligo safety working group exaggerated pharmacology subcommittee consensus document. , 2013, Nucleic acid therapeutics.

[29]  L. Shihabuddin,et al.  Sustained Therapeutic Reversal of Huntington's Disease by Transient Repression of Huntingtin Synthesis , 2012, Neuron.

[30]  A. Becker,et al.  Molecular correlates of age-dependent seizures in an inherited neonatal-infantile epilepsy. , 2010, Brain : a journal of neurology.

[31]  C. Bennett,et al.  RNA targeting therapeutics: molecular mechanisms of antisense oligonucleotides as a therapeutic platform. , 2010, Annual review of pharmacology and toxicology.

[32]  A. Walf,et al.  The use of the elevated plus maze as an assay of anxiety-related behavior in rodents , 2007, Nature Protocols.

[33]  I. Whishaw,et al.  Bilateral alteration in stepping pattern after unilateral motor cortex injury: A new test strategy for analysis of skilled limb movements in neurological mouse models , 2006, Journal of Neuroscience Methods.

[34]  I. Scheffer,et al.  Channelopathies as a genetic cause of epilepsy , 2003, Current opinion in neurology.

[35]  George Paxinos,et al.  The Mouse Brain in Stereotaxic Coordinates , 2001 .

[36]  A. L. Goldin,et al.  A gain-of-function mutation in the sodium channel gene Scn2a results in seizures and behavioral abnormalities , 2001, Neuroscience.

[37]  H. Lerche,et al.  Activity of Na V 1 . 2 promotes neurodegeneration in an animal model of multiple sclerosis , 2018 .

[38]  F. Rigo,et al.  Antisense oligonucleotide-based therapies for diseases caused by pre-mRNA processing defects. , 2014, Advances in experimental medicine and biology.

[39]  De novo mutations in epileptic encephalopathies , 2013 .

[40]  E. Masliah,et al.  Neuronal death and perinatal lethality in voltage-gated sodium channel alpha(II)-deficient mice. , 2000, Biophysical journal.

[41]  M. Takano,et al.  Neurotoxic effects of phenytoin on postnatal mouse brain development following neonatal administration. , 1999, Neurotoxicology and teratology.