U.S. flag

An official website of the United States government

NCBI Bookshelf. A service of the National Library of Medicine, National Institutes of Health.

Noebels JL, Avoli M, Rogawski MA, et al., editors. Jasper's Basic Mechanisms of the Epilepsies. 5th edition. New York: Oxford University Press; 2024. doi: 10.1093/med/9780197549469.003.0005

Cover of Jasper's Basic Mechanisms of the Epilepsies

Jasper's Basic Mechanisms of the Epilepsies. 5th edition.

Show details

Chapter 5Epilepsy Genomics

Disease-Causing Sequence Variants

.

Abstract

Approximately 1% of 25,000 genes and 25 million single-nucleotide polymorphisms in the genome of modern humans are epilepsy variants. Epilepsy variants are the molecular basis of fatal progressive myoclonus epilepsies, epileptic encephalopathies, and other genetic epilepsies. This chapter annotates the early success and key turning points in the quest to cure fatal epilepsies. The first key turning point occurred in 2018 when “every-2-weeks” intraventricular infusion of cerliponase alfa, enzyme replacement therapy, halted progression of an early childhood progressive myoclonus epilepsy (CLN2 Batten disease), as reported by Schulz et al. In 2021, Schaeffer et al. reported 2 years of intraventricular cerliponase alfa delayed or prevented onset of symptoms in presymptomatic CLN2 infants. Earlier, in 2017, the Food and Drug Administration (FDA) and European Medicines Agency approved Nusinersen, an antisense oligonucleotide, and AVXS-101 (Zolgensma), a viral-mediated gene replacement therapy, as disease halting and potential cures for Werdnig Hoffmann type I spinal muscular atrophy of infancy (SMN1). Even now, breakthrough advances by others include (1) ex vivo HSPC transduction gene therapy in metachromatic keukodystrophy, and (2) genome editing with zinc finger nuclease opening DNA strands and inserting a correct alpha-L-iduronidase into the genome of Hurler mucopolysaccharidoses patients. Recently, the FDA proclaimed that they expect to approve 10–20 cell and gene therapies a year from 2025 onward. As clinical trials raise hopes for cures, vigilance for adverse effects and ethical concerns must not waiver.

Introduction

The epilepsy genome constitutes approximately 1% of 25,000 genes contained in the genome of the modern human (Homo sapiens sapiens). Of 25 million common and rare single-nucleotide polymorphisms or variants (SNPs), about 1% are epilepsy variants. Epilepsy variants include its nucleotide sequences, coding and noncoding DNA (enhancers, promoters and terminators, transposable elements, and repetitive DNA), and mitochondrial DNA. Epilepsy genomics, the study of the epilepsy variants’ function, structure, analysis, regulation, editing, and evolution (Cristescu, 2019), together with the natural history of epilepsy syndromes, laid the platform for the five genetic and clinical ingredients necessary for a quest to cure epilepsies. The “concept of genome” came from Hans Winkler (1920).

This fifth edition of Jasper’s Basic Mechanisms of the Epilepsies will be remembered as the edition when the tools for the medical interpretation of the human epilepsy genome and discovery of epilepsy variants were in high gear, when epilepsy genomics laid the foundation for precision medicine in epilepsy, and when genomics-based therapeutics in epilepsy moved from symptomatic alleviation of disease to modification and halting disease progression and to the courageous quest for cures. One by one, genomics-based functional studies are defining disease mechanisms of fatal and nonfatal, nonprogressive genetic epilepsies of infancy, childhood, adolescence, and adulthood. The “single, one-and-done” gene replacement therapy (GRT, or introducing a wild-type sequence to replace the defective sequence) and disease mechanisms, targeted, down-regulated, or up-regulated by antisense oligonucleotides (ASOs), and disease mechanisms modified or edited by the genius of our researchers in basic mechanisms of epilepsies and the pharmaceutical industry provide the paths toward (a) the future curing of nonfatal common genetic epilepsies and (b) the beginning of the end of some fatal genetic epilepsies.

This chapter describes the progress in epilepsy genomics that transformed the quest to cure epilepsies by establishing five key genetic and clinical ingredients necessary for this quest. The chapter also identifies the key turning points when proof of principles were obtained in epilepsy clinical trials that a fatal epilepsy can have its disease progression halted, when symptoms can be reversed, and when a possible cure can be proven in presymptomatic individuals. The “every-3-months” intrathecal ASO injections are contrasted with the “single, one-in-a-lifetime” intrathecal injection of enduring viral-vector based GRT. At the start, the chapter provides a brief summary of the history of epilepsy variants and epileptogenesis concepts generated by disease mechanisms of epilepsy variants. In closing, the chapter emphasizes the need for continuing vigilance for unexpected adverse effects of novel therapeutics and highlights key social and ethical concerns in the quest to cure the epilepsies.

History of Curating the Epilepsy Genome: The First Ten Years of Epilepsy Variants

Epilepsy variants are the molecular basis of specific genetic epilepsy syndromes, causing their phenotypes and traits. Historically, the complexity and variability of phenotypes of age-dependent genetic epileptic syndromes and their overlapping clinical features limited the resolution of phenotype-based epilepsy classification and confounded epilepsy nosology.

Then came the first pioneers who discovered epilepsy genetic variants in the decade from 1995 to 2005 (see Table 5–1 for list of epilepsy genetic variants). In 1995, Steinlein and her team were among the first pioneers to show germline epilepsy causing variants/mutations in the alpha4 subunit of nACH receptor (CHRNA4) (Steinlein et al., 1995, 1997). But the story of curating the epilepsy genome started earlier than 1995 with Mark Leppert and Elving Anderson. In 1989, Leppert and his team at the University of Utah, in collaboration with Anderson’s group at the University of Minnesota, established genetic linkage of the long arm of chromosome 20 to benign familial neonatal seizures (BFNS). This was the first report of a genetic linkage of a chromosome site to an epilepsy syndrome (Leppert et al., 1989). Nine years later, in 1998, the same collaborating teams reported KCNQ2 as the potassium channel mutated in an inherited epilepsy of newborn. At the time it was considered a novel potassium channel gene. Now we know that KCNQ2 variants (more than 80 variants described) are the most common cause of BFNS (Singh et al., 1998; Charlier et al., 1998; Biervert & Steinlein, 1999; Biervert et al., 1998).

Table Icon

Table 5–1

Some Epilepsy Genes and Their Phenotypes.

At about the same time, while the first epilepsy variants were being discovered in Germany and Utah, Scheffer and Berkovic, in 1997, taught us about a then unrecognized syndrome of generalized epilepsy with febrile seizures plus (GEFS+). Over the following 2–3 years, this Australian team showed that a variant in SCN1B could cause GEFS+, and that variants in SCN1A are the most common genetic causes of GEFS+ and febrile seizures (Scheffer and Berkovic, 1997; Wallace et al., 1998; Escayg et al., 2000, 2001).

As the twenty-first century turned, in 2002, Cossette with Rouleau and colleagues, in Montreal, identified, for the first time, that a variant in an inhibitory neurotransmitter receptor, namely GABA-A receptor (an Ala322Asp variant in GABRA1), could cause epilepsy in clinically affected members of a large multiplex French-Canadian family with juvenile myoclonic epilepsy (JME) (Cossette et al., 2002). Since then, variants in GABA-A gamma 3 subunit in chromosome 15q11 and variants in GABA-A receptor gamma 2 subunit in chromosome 5 and variants in GABA-A receptor beta 3 subunit have been reported in absence epilepsy syndromes. It is now accepted that Absence epilepsies, as part of the GEFS+ syndrome or as a separate and exclusive absence syndrome, are caused by GABR variants (Baulac et al., 2001; Wallace et al., 2001; Tanaka et al., 2008).

In 2003, one year after the first discoveries of variants in GABA-A receptors causing absence epilepsies and JME, Chen et al. (2003) reported association between variants in a calcium channel, CACN1A and childhood absence. Variants in CACN1A and CACNB4 have now also been observed in childhood absence with ataxia (Imbrici et al., 2004). Thus, in the first decade of epilepsy variants, “proof of principle” evidence was documented mainly for ion channelopathies and transmitter-receptoropathies.

In 2004, the first report of variants in a non-ion channel and non-transmitter receptor gene was made by Suzuki et al. (2004) in families with JME. The variants were in EFHC1, a novel gene, which encodes a microtubule-associated protein with an EF-hand motif whose variants disturb R-type VDCC and TRPM2-calcium currents. Among 17 cohorts with 54 EFHC1 variants, reported in literature by 2018, 23 were pathogenic or likely pathogenic. According to the National Human Research Institute and American College of Medical Genetics and Genomics, gene-level evidence, and variant-level evidence, EFHC1 is the first non-ion channel associated with JME. Since then, variants in other non-ion channel genes, such as intestinal cell kinase (ICK) and carboxypeptidase A6 (CPA6), have also been observed in JME (Bailey et al., 2017, 2018; Sapio et al., 2015). Ottman et al. (2010) were the first to report variants in a non-ion channel gene, leucine-rich glioma-inactivated 1 (LGI1), in a focal epilepsy known as autosomal dominant lateral temporal lobe epilepsy (ADLTLE with auditory features). About half of ADLTLE families have variants in LGI1 or epitempin (Kalachikov et al., 2002; Micheluchi et al., 2003).

Benefits of Curating the Epilepsy Genome

Genetic Testing for Epilepsy Variants Improves Diagnosis Treatment and Prognosis

Curating the epilepsy genome sharpened the diagnostic view of the medical community to genetic testing of epilepsy syndromes. But it was the 2001 discovery of somatic recurrent de novo mutations in a sporadic epilepsy syndrome first described by Dravet (Claes et al., 2001) that sealed the deal for genetic testing—for example, in the alpha1 subunit of the sodium channel (SCN1A) arising mainly from paternally derived chromosomes, the beta1 subunit of sodium channel (SCN1B) (Wallace et al., 1998; Escayg et al., 2000, 2001), and in protocadherin 19 (PCDH19) in X-linked females with Dravet syndrome (Depienne et al., 2009; Dibbens et al., 2008). This recognition of de novo mutations in sporadic epilepsies led to whole exome sequencing (WES) of developmental disabilities mixed with infantile epileptic encephalopathies, such as infantile spasms in West syndrome and Dravet syndrome. In 2013, WES revealed de novo variants in GABRb3, CACNA1A, CHD2, FLNA, GABRa1, GRIN1, GRIN2B, HNRNPU, IQSEC2, MTOR, and NEDD4L (Allen et al., 2013) and, in 2017, de novo variants in NTRK2, GABRB2, CLTC, DHDDS, NUS1, RAB11A, GABBR2, and SNAP25 (Hamdan et al., 2017).

With association of disease-causing variants with specific epileptic syndromes also came realization of specific antiseizure drugs (ASDs) as “keys that fit the lock” of disease mechanisms of epileptic syndromes. The concept of “antiseizure drugs that act as sodium channel blockers worsen genetic epilepsies” had to be modified. De novo SCN1A variants Dravet syndrome or epileptic encephalopathy is worse with ASD sodium channel blockers (Dravet et al., 1992, 2005), but de novo SCN8A or SCN2A variants Dravet syndrome can respond favorably with ASD sodium channel blockers. Some KCNT1-deficient malignant migrating partial epilepsies in infancy respond to quinidine (Abdelnour et al., 2018) and some to carbamazepine. Sodium channel blockers can also be effective in variant KCNQ2, while ketogenic diet has diverse efficacy in variant SCN1A, CDKL5, KCNQ2, STXBP1, and SCN2A (Ko et al., 2018).

There are now 90 to 172 disease-causing variants primarily expressed as epilepsies or epileptic encephalopathies targeted in gene-panel sequencing platforms by commercial genome centers (Table 5–1) and about 40 genetic diseases whose phenotype significantly includes epilepsy (Table 5–1). There is an ever-growing list of epilepsy genes, and a complete list here and now would not be possible.

Novel Molecular Concepts of Epileptogenesis in Genetic Epilepsy Syndromes

The concepts of epilepsy syndromes as epilepsy diseases (Roger et al., 1985), their appearance as developmental epileptic encephalopathies, and their association with autism spectrum disorder (Specchio et al, 2022) were bolstered when the epilepsy causing pathogenic variants/mutations and disease mechanisms were established.

These, in turn, led to new concepts of epileptogenesis:

  • Concept 1. Developmental and epileptic encephalopathies (DEEs) of infancy and childhood can be genetic and involve epilepsy variants that affect synaptic function and structure and transcription regulation. This is exemplified by STXBP1/MUNC18-1 or ARX or KCNQ2 mutations in Ohtahara syndrome (Kato et al., 2007; Saitsu et al., 2008; Weckhuysen et al., 2012); SHANK3, CDKL5, NRXN1, PCDH19, and CASK in generalized epilepsies with autism spectrum; and SYNGAP1 in myoclonic absence epilepsy and epilepsy with myoclonic absence with autism spectrum and SRPX2 mutations in disorders of language cortex and cognition with epilepsy (Roll et al., 2006). MECP2 involved in chromatin remodeling and autistic behavior is a further example (Shibata et al., 2020; Specchio et al., 2022).
  • Concept 2. Ion channelopathies can cause simple febrile seizures and GEFS+ syndrome as well as benign neonatal, infantile epilepsy syndromes and epileptic encephalopathies. This is exemplified by de novo SCN1A, SCN1B, KCNQ2, and KCNQ3 pathogenic variants in Dravet syndrome (Claes et al., 2001; Dravet et al., 2005a,b; Harkin etal., 2007; Wallace et al., 1998, 2001a, 2001b, 2003; Hirose et al., 2000; Marini et al., 2007, 2009; Nabbout et al., 2003; Oguni et al., 2001, 2005; Singh et al., 2009) and autosomal dominant transmitted variants of KCNQ1 gene in Long Q-T syndrome and SUDEP (Tester et al., 2005).
  • Concept 3. Receptoropathies can cause genetic focal epilepsies (ADNFLE) (Rodrigues-Pinguet et al., 2003) as well as genetic generalized epilepsies. For example, pathogenic variants in nACH or GABR (Steinlein et al., 1995, 1997; De Fusco et al., 2000; Hirose et al., 1999; Wallace et al., 2001) can cause absence epilepsies of childhood and adolescence (Baulac et al., 2001; Cossette et al., 2002; Dibbens et al., 2004; Wallace et al., 2001a; Kanamara et al., 2002); GRIN2A genetic variants in epilepsy plus aphasia spectrum disorder, which is composed by Rolandic epilepsy, atypical benign partial epilepsy, epileptic encephalopathy of continuous spike-and-waves during slow-wave sleep, and Landau-Kleffner syndrome, were first reported in 2012 by Szeptowski’s group in Aix-Marseilles University (Lesca et al., 2013). Replication came from Seattle and Australia (Carvill et al, 2013; von Stülpnagel et al., 2017). GRIN2B and GRIN2D each associates with DEE.
  • Concept 4. Developmental non-ion channel and non-receptor genetic variants can cause common epilepsies. Some examples include Myoclonin1/EFHC1, Intestinal cell kinase (ICK), Importin 8 (IPO8), PROSER1, MYOFERLIN, BRD2, EL4, TBC1D24, CDKL5, STXBP1, and MAG 12, which can cause common epilepsies, such as juvenile myoclonic epilepsy, Rolandic epilepsy, familial infantile myoclonic epilepsy, and infantile spasms (Suzuki et al., 2004; de Nijs et al., 2009; Cavalleri et al., 2007a,b; Pal et al., 2003; Greenberg et al, 2005; Lorenz et al., 2006; Strug et al., 2009; Falace et al., 2010; Corbett et al., 2010; Weaving et al., 2004; Deprez et al., 2010; Marshall et al., 2008; Mei et al., 2009; Gazzerro et al., 2011).
  • Concept 5. Transporter genes’ variants cause both genetic generalized epilepsies and DEE. Examples are SLC1A2 (solute carrier family 1-glial high affinity glutamate transporter) in DEE; SLC12A5 (solute carrier family 12-potassium/chloride transporter) in DEE, as well as genetic generalized epilepsy; SLC13A5 (solute carrier family 13–sodium dependent citrate transporter) in DEE; SLC25A12 (solute carrier family 25-mitochondrial carrier-aspartate-glutamate carrier 1 transporter) in DEE; SLC25A22 (solute carrier family 25-mitochondrial carrier-glutamate transporter) in DEE; SLC2A1 (solute carrier family 2-facilitated glucose transporter 1) in genetic generalized early childhood absence epilepsy (Nakagama et al., 2012; Suls et al, 2008, 2009; Wang et al., 2002, 2007, 2009; Leen et al., 2010; Yang et al., 2011); and SLC6A1 (solute carrier family 6, neurotransmitter GABA transporter) in myoclonic atonic Doose syndrome (Specchio et al., 2022; Kodera et al., 2013).
  • Concept 6. Genes involved in membrane trafficking and exocytosis. Examples of this include STX1B in GEFS+, STXBP1 in DEE, and SYNGAP1 (synaptic RAS-GTPase- activating protein) in DEE plus autism spectrum (Specchio et al., 2022).
  • Concept 7. Genes encoding cell adhesion molecules. Examples of this include a GPI-anchored neuronal membrane protein contactin2 or its gene CNTN2 in familial adult myoclonic epilepsy (FAME) and Protocadherin 19 (PCDH19) (Marini et al., 2010) variants in chromosome X, now associated with sporadic infantile epileptic encephalopathy, female restricted form of Dravet syndrome, and also childhood absence epilepsy and other DEEs (Shibata et al., 2020; Specchio et al., 2022).
  • Concept 8. Genes involved in cell death (Laforin/DSP and cystatin B [Minassian et al., 1998; Serratosa et al., 1999; Pennachio et al., 1996], and the proteasome degradation pathways (Malin/UbiquitinE3 ligase [Chen et al., 2003; Rao et al., 2010]), as well as errors in glycogen metabolism (Turnbull et al., 2011) can cause progressive myoclonus epilepsy syndromes.
  • Concept 9. Specific developmental genes involved in neuronal cell proliferation (e.g., EMX2 homeobox gene in schizencephaly) and (Stromme et al., 2002) progenitor cell migration (e.g., filamin 1 gene in periventricular nodular heterotopia; double cortin gene in X-linked lissencephaly and subcortical band heterotopia; EFHC1, ECK, and IPO8 in JME (Suzuki et al., 2004; Bailey et al., 2017, 2018; Nganou et al., 2018; Dobyns et al., 1993) and cell differentiation (G protein–coupled receptor gene 6 or GPR56 in bilateral frontoparietal polymicrogyria) cause epilepsies in malformations of the cerebral cortex (Bingham et al., 1998; Guerrini and Marini, 2006; Katsarou et al., 2018; Kitamura et al., 2002).
  • Concept 10. mTOR (mammalian target of rapamycin) signaling pathways (Wong, 2010) or mTORpathies, where variants along the genes encoding mTOR pathway regulatory proteins have been linked to focal cortical dysplasia types IIa and IIb, hemimegalencephaly and megalencephaly, such as variants in DEPDC5, NPRL2, and NPRL3, which are linked to focal cortical dysplasia and tuberous sclerosis (TSC1 and TSC2); and hemimegalencephaly is frequently linked to P13K-AKT variants (Iffland et al., 2019).
  • Concept 11. Microscopic rearrangements, microdeletions, and chromosomal defects are common causes of epilepsy, dysmorphisms, and mild mental retardation (Dibbens et al., 2009; Helbig et al., 2009; Schinzel et al., 2001; Marshall et al., 2008).
Table Icon

Table 5–2

Some Genetic Diseases Whose Phenotype Includes Epilepsy.

Epilepsy Genomics: At Last a Road to Halting and Reversing Disease Progression and Possible Cures

Key Elements in the Genetic and Clinical Platform Needed in the Quest to Cure Each Epilepsy

Epilepsy genomics in studying structure and function of epilepsy variants, their epilepsy-causing mechanisms in knockout and knock-in mouse models of epilepsy phenotypes, and the rescue of epilepsy and disease symptomatology of epilepsy syndromes in conditional knock-in mouse models helped identify the key and critical genetic ingredients.

Epileptologists who specialize in rare epilepsies by studying the natural history of these rare epilepsy syndromes can identify the critical window or time when treatment should start in their quest to cure the disease. Additional elements in the road to the quest are the earliest clinical, electroencephalogram (EEG), and biological biomarkers that can be used to gauge onset and progression of disease.

These key elements are as follows:

1.

The monogenic fully penetrant epilepsy-causing variant must be defined and its structure and disease mechanism(s) offered as a therapeutic target.

2.

Transgenic mouse models for the clinical epilepsy must be available for the preclinical trials of potentially curative therapeutic agents.

3.

Potentially curative therapeutic agents must cross the blood–brain barrier (BBB); otherwise intrathecal or intracerebral-intraventricular route will be route of administration.

4.

The critical window to start treatment in clinical trials for proof of a cure. Natural history studies of the clinical epilepsy syndrome must define the following:

(a)

The onset and trajectories of disease progression (but not only this)

(b)

The EEG spike wave indices, sleep architecture, and cognitive conditions of the presymptomatic states

(c)

When clinical disease starts versus when pathology starts and the optimal time to start treatment to prove a cure

5.

A population of genotyped probands and affected family members must be available for the actual clinical curative trial (see Table 5–3).

Table Icon

Table 5–3

Key Turning Points in the Quest for Cures.

Key Turning Points

There are several key turning points in epilepsy genomics, specifically in human clinical trials and in knockout or knock-in mouse models that provided proof of principle that a progressive epilepsy disease can be halted, symptoms reversed, and a possible cure obtained.

Proof of Principle: Disease Progression Can Be Halted

The First Epilepsy/Neurodegenerative Syndrome in Which the First Evidence of Halting Disease Progression Was Obtained by Enzyme Replacement Therapy: Intraventricular Cerliponase Alfa for CLN2 Form of Batten Disease

All the five above genetic and clinical ingredients were present in the quest to cure neuronal ceroid lipofuscinoses (NCL), a previously incurable epilepsy/neurodegenerative disease of childhood. There are 13 CLN pathogenic variants in genes that process lysosomal enzymes and receptors and that are transmitted as an autosomal recessive trait. There is one pathogenic CLN variant that is inherited as an autosomal dominant trait (CLN4). The CLN4 pathogenic variant is in DNAJC5 gene, which encodes the presynaptic co-chaperone cysteine string protein. Disruption of lysosome functions lead to the buildup of cellular aggregates composed by lipofuscin, fine yellow and brown pigments with autofluorescent granules—hence the name, lipofuscinoses. Cell aggregates of lipofuscin accumulate in neurons of the central nervous system (CNS), retinal and ganglion cells of the eye, liver, heart, spleen, adrenals, and kidney. Clinically, NCL patients share common signs of epileptic seizures, visual and cognitive decline, and regression of motor skills and balance. Age at disease onset varies according to the pathogenic variant.

A major problem of these NCL diseases is that the disease mechanisms of the 14 pathogenic variants known to cause their clinical phenotypes are not completely understood and remain unknown in some. Because of this, most research efforts in NCL diseases in the last decade were devoted to developing human gene therapy. Gene therapy as defined by the Food and Drug Administration (FDA) (Cellular & Gene Therapy Guidances, July 20, 2018) and the European Union Commission (Directive 2001/83/EC, Part IV of Annex I) is a “biological medicinal product containing recombinant nucleic acid used in or administered to a human to regulate, repair, replace, add or delete a genetic sequence with the aim to cure or treat diseases” (Jensen et al., 2021).

Gene therapy includes (a) “in vivo” vector-mediated gene replacement, (b) “ex vivo” cell transduction gene therapy, and (c) genome editing. Most research programs in NCL chose an in vivo viral-vector-mediated GRT program that targets loss-of-function mutation diseases. This GRT approach reached high hopes for a cure when the FDA and European Medicines Agency (EMA) both approved Zolgensma (onasemnogene abeparvovec) for in vivo adeno-associated virus mediated GRT for spinal muscular atrophy. More recently, EMA approved an ex vivo gene therapy with lentivirus vector transduced autologous CD-34- positive stem cells for therapy of metachromatic leucodystrophy (Libmeldy). FDA approval of cerlifonase alfa (Brineura) for CLN2 disease has given more hope for families and parents who have children with NCL.

Stopping Disease Progression by “Every-Two-Weeks” Intraventricular Infusion of Cerliponase Alfa

In 2018, Schulz et al. were the first to show a halt to disease progression for a neurodegenerative epilepsy syndrome, neuronal ceroid lipofuscinosis type 2 (CLN2) (Schulz et al. 2018). CLN2 disease has a rapidly progressive and invariably fatal outcome, which was modified by intraventricular enzyme replacement therapy. CLN2 disease is caused by pathogenic variants in the gene encoding lysosomal enzyme tripeptidyl peptidase 1 or TPP1. CLN2-deficient children are normal until 2 to 4 years; then the disease starts as an epilepsy (tonic-clonic or partial seizures in 70%) and delayed language development (57%). Slowed psychomotor development may be followed by a clinical standstill, after which delayed language development is recognized. Cognitive, motor, and visual functions regress, leading to death by age 12 years due to accumulation of lysosomal storage material in the CNS and retina.

Diagnosis is confirmed by homozygote or compound heterozygote mutations in the TPP1 gene and deficiency or absence of TPP1 enzyme activity. EEG shows slow and attenuated background activities, epileptiform activities in occipital regions, and photoparoxysmal responses at low frequencies (1 to 2 Hz) of photic stimulation. Brain MRI shows early evidence of cerebral and cerebellar atrophy. Optic coherence tomography may show retinal degeneration and accumulation of hyperreflective material, abnormal visual evoked potentials, and diminished electroretinographic amplitudes.

In the study by Schulz et al. (2018), 24 children had their CLN2 disease documented by TPP1 enzyme activity and CLN2 genotype. Median age at enrollment was 5 years, range 3.3 years to 9 years, with subjects classified as having mild to moderate disease progression as measured by a combined score of 3–6 on the motor and language domains of the CLN2 clinical rating scale.

Subjects received during the stable dose phase, 300 mg of intraventricular cerliponase alfa (a recombinant proenzyme form of human TPP1, also called zymogen, developed by BioMarin pharmaceuticals) every 2 weeks for at least 96 weeks, after initial doses of 30 mg, 100 mg, or 300 mg. Intraventricular infusion rate at 2.5 ml per hour for 4 hours through a surgically implanted Ommaya or Rickham ventricular reservoir is meant to replace the deficient enzyme in affected brain tissues. Cerliponase alfa is activated upon uptake into the acidic pH environment of lysosomes in vivo and becomes the mature proteolytic form of rhTPP1 that catabolyzes lysosomal aggregated storage material. Concentrations peak in CSF at the end of ~4-hour infusion and 8 hours thereafter in plasma.

The primary efficacy outcome was the time until an “unreversed” first two-point decline in the motor and language domains of the CLN2 clinical rating scale, occurred, compared to 42 historical controls. The treated children had slower decline in motor and language function than that in CLN2 historical control patients. Adverse effects consisted of convulsions, pyrexia, vomiting, hypersensitivity reactions, and device-related complications, including grade-3 infection, leakage, and an increase in white cell count in cerebrospinal fluid in half the patients. There was no apparent correlation between cerebrospinal fluid and plasma peak concentrations and incidence of adverse effects. There was no relationship between magnitude of csfexposure and changes in CLN2 score, indicating maximum benefit across the range of exposure with 300 mg every 2 weeks.

This clinical trial that started as the first in human phase I/II study and one subsequent phase I/II extension study still in progress were made possible by a safety and efficacy study and pharmacology/pharmacokinetics study in young dogs with spontaneous homozygous TPP1 deficiency and safety studies in monkeys. Cerliponase alfa clears lysosomal storage material and preserves neuron morphology and reduces brain inflammation in CLN2-deficient dogs. Treated dogs had delayed onset and slower progression of neurologic signs, brain atrophy, preserved cognitive functions, and an extended lifespan (Schulz et al., 2018).

Nusinersen (Spinraza) for Spinal Muscular Atrophy

Nusinersen provided the first proof of principle that disease progression can be halted, symptoms decreased, and risk for death reduced in a non-epilepsy disorder (spinal muscular atrophy [SMA] rarely manifests with myoclonus epilepsy) (Chen, 2020; Finkel et al., 2016). SMA is caused by a loss-of-function variant in the “survival motor neuron1 or SMN1 gene.” SMN1 gene encodes a protein essential for survival of alpha motor neurons in the spinal cord. Most commonly, this is a homozygous deletion of exon 7 of the survival motor neuron 1 gene resulting in SMN protein deficiency. The human genome has a second and similar gene, SMN2, which is different from SMN1 only by a few nucleotides, notably, a nucleotide variant 7. This variant in SMN2 leads to exclusion of exon 7, resulting in an unstable truncated protein without exon 7. In SMA, a pathogenic variant in SMN1 is present and a few copies of SMN2, which is not enough to transcribe/translate into functional SMN proteins.

Clinically, four separate syndromes of SMA are present.  Type Zero or Type 0 with one SMN2 copy starts in utero. Infants are dependent on mechanical ventilation at birth, and death usually follows before 6 months of age. Type 1 SMA or Werdnig Hoffmann disease with one to three SMN2 copies is the most common clinical syndrome, accounting for approximately 60% of all SMA cases. Type 1 SMA starts before 6 months of age, as hypotonia and muscle weakness require posture, respiratory, and feeding support. Median age to ventilation is at 10.5 months and to exitus is at 13.5 months. Type 2 SMA or Dubowith disease with one to two SMN2 copies is often included in the clinical trials because it also starts early, after 6 months of age, usually 7 to 18 months, with infants able to sit up but unable to walk independently, also eventually needing respiratory and feeding support. Life expectancy may reach adulthood. Type 3 Kugelberg-Welander disease, with three to four SMN2 copies, starts at 18 months with inability to stand and walk. Eventually gaining strength, patients have a normal life expectancy. Life expectancy is also normal in Type 4 with four SMN2 copies with adult-onset mild muscle weakness.

In 2006, an intron-splicing silencer N1 sequence in intron 7 of the SMN2 gene, which enhances skipping of exon 7 and produces full and functional SMN2 protein, was identified. ASOs were then designed that bind to the intron-splicing silencer N1 region of the SMN2 pre-messenger promoter exon 7, upregulating transcription of full-length SMN2. This led to the original open-label phase 1/2 clinical trial in 2016 for “the every-3-to-4-months” intrathecal nusinersen (brand name Spinraza) that was followed by an open-label extension study (Chen, 2020). Then, in 2017, Finkel et al. (2017) led a double-blind, placebo-controlled trial with type 1 and type 2 SMA patients.

All trials showed that multiple intrathecal nusinersen reduced symptoms, halted disease progression, and reduced the risk of death or permanent ventilation by 47%. All three clinical trials also showed a favorable safety profile. Among the three clinical trials, a total of 194 type 1 SMA or Werdnig-Hoffman type SMA were treated with nusinersen with similar positive results of improvement (Finkel et al., 2016, 2017; Aragon-Gawinska et al., 2018; Pechmann et al., 2018).

Proof of Principle: Preventing or Delaying Onset of Symptoms: Proving a Possible Cure in a Presymptomatic CLN2 Individual

Since its first reports, in 2018, cerliponase alfa (Brineura) had its first global approval by the FDA in the United States for CLN2 patients that are over 3 years of age and in the EMA for CLN2 patients of all ages. Real-world experience of effectiveness of cerliponase alfa in CLN2 patients has been reported in Colombia (eight patients) and in France (seven patients) with similar success in halting disease progression and even improvement of clinical signs when patients with a CLN2 score greater than 3 were enrolled for intraventricular enzyme GRT.

Remarkably, Schaefers et al. (2021) reported that a 23-month-old presymptomatic CLN2 individual was treated with intraventricular cerliponase alfa for 2 years and demonstrated a delay in onset of symptoms. The individual had walked at 14 months, running and climbing stairs with fine motor skills. He spoke two-word sentences, had normal vocabulary for age, and had good nonverbal communication skills. He had no symptoms of CLN2 disease at the start of treatment. This 23-month-old subject had maximum scores (total score of 12) on all subdomains of the CLN2 rating scale, which remained the same over the 2-year course of treatment. An increase in cognitive development from 21 to 39 months was observed, and cerebral and cerebellar atrophy was not observed on MRI after 2 years of treatment when the subject reached 4 years of age. Remaining asymptomatic at 4 years of age, the subject did have a prolonged P100 latency time on visual evoked potential studies. Results, nonetheless, suggest that disease onset has been delayed and a cure could be possible with continued treatment. Studies with a larger population of presymptomatic individuals with pathogenic variants in CLN2 are needed to truly establish a cure, but this single case report is most encouraging.

Pilot programs testing if presymptomatic SMN1 newborns can be cured by intrathecal nusinersen support the studies on presymptomatic CLN2 children and FDA and EMA approval of AVXS-101 or Zolgensma for in vivo adeno-associated viral-mediated GRT for SMN1 patients represent groundbreaking milestones in the quest to cure epilepsy and neurodegenerative diseases (Jensen et al., 2021; Aragon-Gawinska et al., 2018; Bertini etal., 2017; Mendell et al., 2021; Chiriboga et al., 2016; Mercuri et al., 2018; Day et al., 2021).

About a year after Nusinersen was shown to be effective in type 1 SMA, AveXis pharma (recently acquired by Novartis Pharmaceuticals) with Mendell et al. (2017) introduced AVXS-101 or onasemnogene abeparvovec (brand name Zolgensma), a “one-and-done, single intravenous injection” of GRT (Jensen et al., 2021). AVXS-101 is contained in a self-complementary AAV9 that crosses the BBB when administered before 2 years of age. AVXS 101 also has a constitutively active promoter that provides persistent expression of SMN1 protein. Three open-label, single-arm, single-dose phase 3 trials were conducted in the United States, Europe, and Asia after a one-time intravenous infusion of AVXS-101 in 22 type 1 SMA infants younger than 6 months. All three clinical trials established safety and efficacy (Mendell et al., 2017, 2021; Al-Zaidy al., 2019a,b; Lowes et al., 2019; Finkel et al., 2019, 2020). End-of-study analysis showed AVXS-101 improved permanent ventilation-free survival and improved motor function and milestones in infants with SMA type 1. At 14 months, 91% survived free from permanent ventilation support. At 18 months, 59% were able to sit independently.

Within 2 years of introduction, a one-time intravenous dosing of AVXS-101 in three presymptomatic infants (age up to 42 days) was started, realizing that a “one-and-done, single intravenous injection” of AVXS-101 will cross the BBB in infants. Results are not yet available. Newborn screening for SMN1 variants has also started in some countries in anticipation of intravenous dosing with AVXS-101 (Jensen et al., 2021).

Eventually, pilot programs for presymptomatic newborns with three copies of SMN2 began in Taiwan, New York, Belgium, and France. Twenty patients have been enrolled (Bertini et al., 2017). Twelve infants have reported typical normal development so far and have been presented in international meetings (Gidaro and Servais, 2019). Long-term efficacy and normality until adulthood remain to be established.

Proof of Principle: One Single Lifetime Injection with Enduring Results

Contrasting “one single lifetime” intrathecal injection with enduring results versus every 2 weeks intraventricular GRT led to ongoing clinical trials testing “one single lifetime” intrathecal injections of CLN2, CLN3, CLN6, and CLN7 ceroid lipofucsinoses.

Recombinant AAV Vectors Have Made Long-Term Enduring Results Possible

With “every-2-weeks” intraventricular cerliponase alfa GRT considered a promising treatment to cure CLN2 disease, a “single-injection treatment strategy of GRT” was developed for CLN2 and other genotypes of CLN with the ultimate goal of achieving a long-term, enduring, if not lifelong cure. The single-treatment strategy of GRT has been made possible by the newer generation of AAV gene therapy vector, self-complementary AAV (scAAV). scAAV vectors deliver long-term expression of the packaged gene which has been demonstrated to be present for as long as 10 years in humans and 15 years in nonhuman primates (Sehara et al., 2017; Chu et al., 2020; Jiang et al., 2006). AAV vectors induce expression in both dividing and nondividing cells and remain within the cell nucleus in episomal form (Salganik et al., 2015). scAAV has a desirable safety profile with less vector-associated toxicity, lack of pathogenicity, and the least immunogenic in humans. scAAV has strong neuronal tropism, is effective in transduction, and is unable to self-replicate. Self-complementary AAV carries a genome that resembles a double-stranded DNA template ready for transcription upon infection. Improved transgene expression cassette designs, and the use of split-vector systems have overcome its small genome size (4.7 kb) packaging.

After two phase1/2 trials using AAVrh10-CNL2 were completed in a mice model and safety profiles in rats and nonhuman primates deemed acceptable, clinical trials using “single injection treatment strategy of intrathecal GRT with the ultimate goal of achieving a long-term, if not life-long cure” soon followed (Sondhi et al., 2007).

Clinical trials.gov now lists such trials in CLN2, CLN3, CLN6, and CLN7 disease testing single injection of intrathecal GRT. Clinical trials.gov lists “Gene Therapy for Children with CLN3 Batten Disease” sponsored by Amicus Therapeutics (NCTO3770572). The human trial is described as an open-label, single-dose escalation clinical trial to evaluate safety and efficacy of AT-GTX-502 (AAV9 .P546.-CLN3) delivered intrathecally in the lumbar spinal cord region of subjects with CLN3 Batten disease. The “one-time intrathecal injection” has seven participants and completion date is estimated as September 2023.

CLN3 in chromosome 16p12.1 encodes a transmembrane 438-amino acid hyrophobic protein (Battenin), which is trafficked to and expressed on lysosomal membranes. Pathogenic variants in CLN3 lead to accumulation and buildup of autofluorescent lipofuscin-like-deposits with lysosomes of affected neurons. CLN3 juvenile CLN is the most prevalent of CLN and presents with rapidly progressive retinal degeneration between 4 and 7 years. In the ensuing decade, seizures, language problems, and ataxia lead ultimately to death during the second or third decades of life.

Results of the open-label, single intrathecal injection of low-dose (6 ×1013) vector genomes and high-dose (1.2 × 1014) vector genomes of AT-GTX-502 were presented by Amicus Therapeutics as a poster in the 17th Annual World Symposium, held online February 8–12, 2021. Early safety and efficacy results included four children; three received low dose and one at high dose. Analyses indicated AT-GTX-502 was safe and well tolerated. Four reported side effects consisted of elevated levels of liver enzymes that resolved.

Intraocular gene therapy with AAV-meditated expression of CLN3 in a mice model of CLN3 eye disease prevented decline in inner retinal function resulting from death of bipolar disease (Klein Holthaus et al., 2020).

Clinical trials.gov now also lists as ongoing “Gene Therapy for Children with CLN6 Batten Disease” sponsored by Amicus Therapeutics (NCTO2725580) and a 15-year follow-up study to evaluate long-term safety and efficacy (NCT04273243). The human trials are described as an open-label, single-dose escalation clinical trial to evaluate safety and efficacy of AT-GTX-501 (AAV9-CNL6) delivered intrathecally in the lumbar spinal cord region of subjects with CLN6 Batten disease. The “one-time intrathecal injection” has 13 human participants with variant late infantile/early juvenile NCL. Preliminary results were presented at the joint meetings of the 16th Child Neurology Congress and 49th Annual Child Neurology Society. Interim analyses for efficacy showed stabilization of motor and language function in treated individuals versus individuals in the natural history cohort (see (see Section 7, Chapter by Kayani et al. and Chapter by Sarah Mole, 2022).).

CLN6 disease is caused by one of the 70 well-characterized pathogenic variants in a 311-amino acid protein with seven predicted transmembrane domains, predominantly localized in the endoplasmic reticulum. Most pathogenic variants lead to a complete loss of CNL6 protein or a truncated nonfunctional CNL6 protein. Two clinical phenotypes result: (1) variant late infantile/early juvenile NCL, which is the relatively more common syndrome, and (2) adult type A Kufs disease. In the variant late infantile syndrome, the disease usually starts at 16 months to 6 years with impaired language and delayed motor and cognitive development in early childhood. Visual, motor, and mental deterioration lead to death at 12 to 15 years of age.

Cain et al. (2019) dosed three 4-year-old cynomolgus macaques with intrathecal sc AAV serotype 9.chicken B-actin.CLN6 and followed these macaques for 6 months. High levels of transgene expression were found in brain and spinal cord of all animals. No adverse effects or pathology were observed. Cain et al. (2019) also delivered scAAV9.CB.CLN6 into cerebrospinal fluid by a single postnatal intracerebroventricular injection into the Cln6nclf mouse model. The single injection prevented accumulation of autofluorescent storage material, including ATP synthase subunit C, prevented reactive gliosis, and loss of dendritic spines. It also prevented motor, memory, and learning deficits and improved survival of the Cln6nclf mouse. These results strongly supported the initiation of the clinical trial (NCTO2725580) for the single-dose AT-GTX-501 (AAV9-CNL6) for the variant late infantile/early juvenile NCL syndrome.

Clinical trials.gov now also lists NCTO4737460 as enrolling and ongoing “Gene Therapy for Children with CLN7 Batten Disease.” The human trial is described as an open-label, single-dose clinical trial to evaluate safety and efficacy of intrathecal AAV9-CNL7. The “one-time intrathecal injection” is in the enrollment stage.

CLN7 disease is caused by pathogenic variants in the major facilitator superfamily of transporter genes with domain containing 8 (MSFD8), which encodes a lysosomal membrane protein of unknown function. Two common mutations are known that causes disease at ages 2 to 7 years of age. Epilepsy, developmental delay, and regression with myoclonus, ataxia, loss of vision, and speech impairment worsen until death at 11.5 years of age.

CLN7 disease recently received attention and merited publication in the New England Journal of Medicine as an example of true personalized medicine, a novel concept known as “N of 1 treatment” where a specific ASO was developed to correct mis-splicing of the MSFD8 gene. The patient was compound heterozygous for an insertion of an SVA retrotransposon and c.1102G>C.RNA sequencing and reverse-transcriptase-PCR revealed missplicing of exon 6 into a cryptic splice-acceptor site in MSFD8 intron 6, about 119 bp upstream from the SVA insertion site. This missplicing was predicted to lead to premature translation termination. A 22-nucleotide antisense oligonucleotide was designed to target the intron 6 SA cryptic splice-acceptor site and nearby splicing enhancers (dubbed as Milasen). RNA sequence from the patient’s fibroblasts showed that Milasen more than tripled the amount of normal (exon6-exon7) splicing. Clinically, after intrathecal dose escalation followed by maintenance doses of 42 mg every 3 months for 1 year, seizures were substantially decreased and several neurologic and neuropsychological subscores stabilized after 7 months of treatment (Kim et al., 2019).

Remaining Challenges and Trends in the Next Ten Years

What about other epilepsies? Is a cure coming for them also in the next 10 years? Recently, the FDA proclaimed that they expect to be approving 10–20 cell and gene therapies a year from 2025 on (Jensen et al., 2021).

Ex Vivo HSPC Transduction Gene Therapy Can Cure MLD

Ex vivo hematopoietic stem and progenitor cell (HSPC) transduction gene therapy has been central in the treatment of lysosomal storage disorders that include, aside from CNS symptomatology, widespread systemic and major organ signs of disease such as metachromatic leukodystrophy (MLD) and mucopolysaccharidoses (MPS).

Epilepsy specialists encounter MLD as a rare cause of seizures in late infancy, early childhood, and even more rarely as seizures in adults. With cerebral demyelination and neuronal death in central and peripheral nervous systems caused by a loss-of-function mutation in arylsulfatase A (ARSA), muscle weakness and wasting plus dementia and seizures characterize MLD. To correct the loss of function of ARSA, three presymptomatic children had their stem cells isolated and transduced ex vivo with a lentivirus-vector carrying the ARSA gene. The autologous CD34-positive HSPCs (OTL-200 from Orel Therapeutics and San Raffaele-Telethon Institute for Gene Therapy) were then reinfused and transplanted into the bone marrow of three children (Biffi et al., 2013).

Progression of MLD was arrested in the three children with stable engraftment of transduced HSPCs; ARSA activity was reconstituted in CSF (Sessa et al., 2016).

As a result of these encouraging results in treatment during the presymptomatic state, 33 other children symptomatic with early-onset MLD were treated with OTL-200. With up to 7.5 years of follow-up, long-term stabilization of motor functions was reported during the 46th Annual Meeting of the European Society for Blood and Marrow Transplantation. These preliminary results were encouraging, together with stable engraftment of OTL-200 in bone marrow and restoration of ARSA activity in cerebrospinal fluid (Calbi et al., 2020; Fumagalli et al., 2022). In a short time, namely, the same year of 2020, the EMA approved OTL-200 (trade name Libmeldy) for treatment of MLD.

Ex vivo transduced CD34-positive HSPCs with a Lenti virus containing N-sulfoglucosamine sulfohydrolase (SGSH, also known as heparan-sulfatase) has been administered to patients with MPS III or Sanfilippo syndrome A (NCT04201405). Sanfilippo syndrome A is the most common and the most severe of MPS with the lowest survival rate. With the study still ongoing, no results are available.

Genome Editing in Hurler Mucopolysaccharidosis I

Mucopolysaccharidosis I (MPS I), an autosomal recessive lysosomal storage disorder, also called Hurler MPS I, is one of the severe forms of MPS I. It is caused by alpha-L-iduronidase enzyme deficiency (IDUA); IDUA is located in chromosome 4 and catalyzes degradation of glycosaminoglycans (GAGs). The enzyme deficiency results in long chains of sugar carbohydrates (GAGs heparan and dermatan sulfates) accumulating in cartilage, tendons, cornea, skin, and connective tissues. Infants develop bone deformities months after birth. Children remain behind in development, with many bodily system disorders such as mental retardation, speech impediments, short stature, stiff joints, heart, and respiratory systems affected. Patients usually succumb to heart failure or pneumonia.

Genome editing using zinc finger nucleases (ZFN) platform studies where AAV8 virus vectors were injected intravenously to genome edit living cells described encouraging findings in rodents (Ou et al., 2020). ZFN binds to triplet DNA sequences and opens up DNA strands with its intrinsic nuclease activity. At the present time, ZFN genome editing uses an intravenous-injected AAV6 vector that inserts a corrective copy of the IDUA (alpha-L- iduronidase) into the patient’s hepatocytes is in clinical trials (NCT02702115). Permanent liver-specific expression of IDUA is expected, and it is hoped that high blood levels of IDUA will cause passage through the BBB into brain. A 10-year follow-up for safety studies is also ongoing (NCTO4628871). Human treatment induced immunogenicity and “on-target” and “off-target” complications, including oncogenesis, is of particular importance in recent attempts at genome editing (Shim et al., 2017; Mills et al., 2006). Ex vivo and in vivo genome editing in MPS I and MPS II reached clinical trials testing, and concerns arose about immunogenic response to the bacterial protein in the Cas9 based on in vivo genome editing (Wang et al., 2015, 2020; Merkel et al., 2015; Poletto et al., 2020) and off-target effects with CRISPR/Cas9 in vivo editing (Lubroth et al., 2021).

Lafora Type Progressive Myoclonic Epilepsy: The Key Elements and Critical Genetic and Clinical Ingredients Necessary for a Quest to Cure Are Already Available

The critical genetic and clinical ingredients necessary for a quest to cure the ultra-rare Lafora disease (LD) or Lafora progressive myoclonus epilepsy (Lafora PME) ( de Haan et al., 2004 ) have been available. Two studies of the clinical natural history: (a) an international/global study of a 45-patient cohort led by UCLA and (b) a 32-patient cohort studied in Madrid, Bologna, Dallas, and Los Angeles. Genotyped LD patient cohorts that are essential to the quest for a cure in LD are now waiting for the clinical trials to start. These advances have been made possible by the Lafora Epilepsy Cure Initiative (LECI), a National Institutes of Health (NIH)-funded program project.

Curative Therapeutics: Preclinical Trials in epm2a and epm2b Knockout Models and Impending Human Clinical Trials in LD

Initially described, in 1911, by Gonzalo Lafora (Lafora & Glueck, 1911), scientific breakthroughs were made in 1998–2003, with the identification of EPM2A and EPM2b as the genetic basis for LD (Minassian et al., 1998; Serratosa et al., 1999; Chan et al., 2003). PTG depletion or downregulation of GS1 removes Lafora bodies and rescues the fatal epilepsy of Lafora disease (Turnbull et al., 2011; Tagliabracci et al., 2008). Using epm2a and epm2b KO models:

Gys1-ASO is an antisense oligonucleotide that targets the mRNA of the brain-expressed glycogen synthase 1 as synthesized by Ionis pharma (Ahonen et al., 2021; see Chapter 56, this volume). The preclinical trial, testing efficacy, and adverse effects of “every-3-months” intrathecal Gys1-ASO has been completed in epm2a and epm2b knockout mouse models. Gys1-ASO prevented Lafora body formation in young mice that had not yet formed them and inhibited further LB accumulation in older mice. Astrogliosis was prevented, and markers of neuroinflammation were reduced. Testing for safety and adverse effects has also been completed in nonhuman simians. The study design for a human clinical trial testing “every-3-months” intrathecal Gys1-ASO in presymptomatic individuals and LD patients with “seizures only” is projected for late 2022 to early 2023.

Canine safety testing pancreatic alpha-amylase bound to a cell penetrating monoclonal antibody (VAL-0417), an antibody-enzyme fusion delivered by intracerebral/intraventricular injection, has completed preclinical tests in epm2a KO models and progressed to canine safety testing (see Chapter, this volume; Austin et al., 2019, Brewer et al., 2019, Sun et al., 2021). The pharma company that has developed this therapeutic agent has obtained an IND number from the FDA and plans to proceed with the novel concept known as “N of 1 treatment.” The pancreatic alpha-amylase bound to a cell penetrating monoclonal antibody (VAL-0417) was developed to digest and degrade the Lafora inclusion bodies.

A single, once-in-a-lifetime intracerebral/intraventricular dose, a GYS1-targeting/ downregulation by microRNA packaged in an “scAAV9” vehicle, has also completed preclinical trials in epm2a and epm2b KO models. Safety concerns, adverse effects, and the effects of varying doses will be tested in a nonhuman simian prior to a clinical trial in humans (see Section VII by Kayani et al., 2022 Chapter 52, this volume).

A single, once-in-a-lifetime dose by intrathecal injection of a wild-type gene to replace a mutated gene or GRT is also being developed for LD. Two scAAV9 constructs, one containing human EPM2A and the other human EPM2B gene, are undergoing preclinical trials in epm2a and epm2b knockout models as a “single, one-time intrathecal injection” in Dallas, Madrid, and Lexington (Kentucky).

Oral small molecules or preclinical small molecules downregulating GYS1 conceived as a potential oral drug against LD were developed by Drs. Peter Roach and Anna DePauli-Roach, Tom Hurley, and David Watt (Tang et al., 2020). This preclinical success was built upon by MAZE pharma, which developed a small molecule that is administered as an oral pill. This small-molecule downregulating GYS1 is being tested now in a clinical trial in Pompe disease where clinical experience will be invaluable for LD clinical trials.

Novel Clinical Study Design to Maximize proof of halting progression, reversal of symptoms and cure based on Natural History of Lafora Disease.

A novel study design that maximizes likelihood of demonstrating proof for halting progression, reversal of symptoms, and a cure will consider the five clinical and EEG stages of LD. During stage zero when the individual is still clinically asymptomatic, 3.5 Hz-multispike-slow waves appear in fragments and sleep architecture is normal. During Stage I “Visual Seizures Only” (6–14 years) and Stage II “Beginning Mild Cognitive Decline” (12–14 years), overnight sleep video-EEGs record loss of REMs. The 3.5 Hz-multispike-slow wave index is 0.33% to 4% in awake, and 2% to 9% in N1 and N2 sleep stages, but is paradoxically suppressed in stage N3 delta sleep. During Stage III “Established Dementia and Status Epilepticus” (15 years), overnight sleep video-EEG show not only loss of REMs, but loss of EEG desynchrony and decrease to loss of N3 delta sleep stage. The 3.5 Hz-multispike-slow wave index rises to 3% to 30% to 80% as absence status and obtundation status dominate clinically. During Stage IV “Myoclonic Encephalopathy” (16–20 years) with no sleep spindles and vertex waves, EEG cannot differentiate wakefulness from sleep. High-voltage 16 Hz polyspikes index dominate with indices rising from 2.6% to 80% as myoclonic status and myoclonic-tonic-clonic status recur. During Stage V, progressive neurologic deterioration (>21 years) leads to exitus.

An open-label study design should treat the EPM2A and EPM2B clinically presymptomatic person and the LD patient when seizures are the only or main phenotype and prevent increases in clinical seizures, EEG 3.5 Hz-multispike-slow wave indices, and EEG high-voltage 16 Hz polyspikes indices and prevent cognitive decline, loss of REMs, and suppression of polyspike-slow waves during N3 delta sleep.

With more clinical trials in NCL disease assessing efficacy of the “single, one-time, intrathecal injection GRT,” research units addressing Lafora disease also tilted in favor of GRT, more recently. However, this was not always the case at first, when the excitement involving nusinersen (Spinraza) for spinal muscular atrophy (SMA) encouraged a similar program for ASOs in Lafora PME. Hence, ASOs were the first therapeutic agents tested in epm2a–/– and epm2b–/– mice.

In Lafora disease, pathogenic variants in EPM2A (glycogen dual specificity phosphatse) and EPM2B (E3 ubiquitin ligase) form longer-than-normal glucan chains and less branch points rendering the glycogen molecule insoluble and forming what has been defined as PAS+ Lafora inclusion bodies. ASO downregulation of GYS1 transcripts halts the formation of these PAS+ inclusion bodies, neuroinflammation, and progression of disease in epm2a–/– and epm2b–/– mice. An ASO will likely be the first therapeutic agent tested in a clinical trial for Lafora disease, projected for 2022/2023.

Lafora Disease: Replacing the Every-Three-Months Intrathecal ASO Injection with One-Time, Single Injection of an AAV9 Viral Package Containing microRNA That Downregulates GS1

Because intrathecal ASOs have to be delivered every 3 to 4 months and probably require lifelong treatment, a “one-time” intrathecal injection of a microRNA which targets and downregulates GYS1 and packaged in an “AAV9 vehicle” was developed and is ongoing as a preclinical trial in epm2a–/– and epm2b–/– mice (Gumusgoz et al., 2022).

“Every-Three-Months Intrathecal ASO” versus “a Single One-and-Done Intrathecal or Intravenous Injection of GRT”

“Which one should my child take?” The success of AVXS-101 or onasemnogene abeparvovec (brand name Zolgensma), a “one-and-done, single intravenous injection” of GRT in type 1 SMA, in three open-label, single-arm, single-dose phase 3 trials in 22 type 1 SMA infants younger than 6 months (Mendell et al., 2017, 2021; Al-Zaidy al., 2019a,b; Lowes et al., 2019) prompted the next study in presymptomatic individuals. Within 2 years of introduction, a one-time intravenous dosing of AVXS-101 in three presymptomatic infants (age up to 42 days) was started, realizing that a “one-and-done, single intravenous injection” of AVXS-101 will cross the BBB in infants. Results are not yet available. Newborn screening for SMN1 variants has also started in some countries in anticipation of intravenous dosing with AVXS-101.

Now, parents of patients with Lafora type progressive myoclonic epilepsies and parents of infants with Wednig Hoffman type 1 SMA ask, “Which treatment should we choose—ASO or GRT? Can I start with ASO and then also get GRT for my child? Or vice-versa?”

In 2019, Dabbous et al. (2019) compared results of AVXS-101 one-time intravenous infusion treatment (clinical trial NCT02122952) with “every-3-to-4-months” intrathecal injection of nusinersen (clinical trial NCT02193074) using “frequentist and Bayesian” approaches. Results suggest that AVXS-101 has an efficacy advantage over nusinersen concerning overall survival, independence from permanent assisted ventilation, motor functions, and milestones.

To evaluate the cost-benefit of AVXS-101 versus nusinersen, results from the clinical trials were also used to create a model that assessed probabilities of patient dying and quality of life. Probabilities showed that 50% of SMA infants treated with AVXS-101 will survive until the age of 3 years. SMA infants treated with nusinersen, only 50% survival to the age of 3 years. Quality-of-life scores also favored AVXS-101 at 15.65 versus 5.29 for nusinersen.

Perhaps these results, comparing these nusinersen versus AVXS-101 clinical trials, convinced the research teams on Lafora disease in Madrid and Lexington (Kentucky) to conduct their preclinical trials testing of intrathecal GRT consisting of EPM2A or EPM2B contained in scAAV9 in epm2a–/– and epm2b–/– mice.

Developmental and Epileptic Encephalopathies

The same five key genetic and clinical ingredients would need to be present for a quest to cure de novo loss-of-function, X-linked, and gain-of-function variants in the classic DEEs of infantile spasms, Lennox-Gastaut syndrome, and Dravet syndrome.  Epilepsy variants have been defined in some DEEs, but the molecular mechanisms of many of these epilepsy variants still need to be fully defined. In 2013, Epi4K and EPGP investigators searched for de novo mutations in two “classic” forms of EE: infantile spasms (IS) or West syndrome and Lennox-Gastaut syndrome (LGS). Exomes of 264 trios were sequenced and 439 putative de novo mutations were identified. Sanger sequencing confirmed 329 de novo mutations. In these 264 trios, nine genes with de novo SNV mutations were present in two or more probands (SCN1A, n = 7; STXBP1, n = 5; GABRB3, n = 4; CDKL5, n = 3; SCN8A, n = 2; SCN2A; n = 2; ALG13, n = 2; DNM1, n = 2; and HDAC4, n = 2). Among these nine genes, SCN1A, STXBP1, SCN8A, SCN2A, and CDKL5 have previously been associated with EE (Allen et al., 2013).

Investigators claim that they had implicated, for the first time, “GABRB as a single gene cause of EE and provide the strongest evidence yet available for any epilepsy” based on (1) four unique de novo mutations in GABRB3, (2) the absence of similar de novo mutations in 610 control exomes, and (3) a computational modeling of these genes associated with EE having low tolerance for protein-disrupting mutations and producing adverse phenotypic consequences. They used the same reasoning to implicate ALG13, an X-linked gene encoding a subunit of the uridine diphosphate-N-acetylglucosamine transferase, which had been reported in two earlier studies of severe intellectual disability and seizures (Allen et al., 2013).

The next year in 2014, the Epi4K consortium combined their 264 trios with 356 trios of the EuroEPINOMICS-RES consorium and analyzed their exome sequencing results. All trios had a diagnosis of epileptic spasms and Lennox Gastaut syndrome. With the expanded cohorts, they found 429 de novo mutations, including de novo variants in DNM1 in five persons and de novo variants in GABBR2, FASN, and RYR3 in two persons. The combined cohort provided a sigificant likelihood analysis (p = 8.2 × 104) (EuroEPINOMICS-RES, Epilepsy Phenome/Genome Project and Epi4K Consortium, 2014).

Hamdan et al. (2017) combined three strategies during a search for de novo mutations in persons with DEE: (1) they identified candidate variants with de novo mutations during whole genome sequencing of 197 individuals with both epilepsy and intellectual disability, (2) they identified additional individuals with the same candidate variants in another series, and (3) they mined several datasets of developmental and epileptic encephalopathy and intellectual disability. By combining these strategies, these authors suggested a causal link between DEE and NTRK2, GABRB2, CLTC, DHDDS, NUS1, RAB11A, GABBR2, and SNAP25.

Epileptic seizure types, a varying degree of intellectual disability and developmental delay, sometimes with dysmorphic features or nystagmus, have also been associated with variants in GABRA3 encoding the α3-subunit of the GABAA receptor on chromosome Xq28 (Niturad et al., 2017).

Aristaless-Related homeobox X-linked gene (ARX), which results in defective GABAergic interneuronal migration, has also been observed in infants with West syndrome epileptic encephalopathy (Katsarou et al., 2017).

More recently, an autosomal-recessive DEE was associated with biallelic variants in PTPN23, which encodes a tyrosine phosphatase (Sowada et al, 2017). Myers et al. (2017) identified six individuals with DEE harboring a de novo mutation in PPP3Ca, which encodes the alpha isoform of calcineurin (a calcium and calmodulin-dependent serine/threonine phosphatase).

A cohort of 23 patients with epileptic encephalopathy carrying loss- or gain-of-function KCNA2 mutations was reported by Masnada et al. (2017). The more severe epilepsy, with developmental problems and ataxia, and atrophy of the cerebellum or even the whole brain, was observed in about half of the patients with gain-of-function mutations.

Preclinical Trial in DNM1 (dynamin1) Mice Model of Epileptic Encephalopathy: Proof of Principle for Halting Disease Progression and Reversing Symptoms and Pathology

Individuals with pathogenic DNM1 variants suffer from infantile spasms and Lennox-Gastaut syndrome (EuroEPINOMICS-RES Consortium, Epilepsy Phrnomr/Genome Project and Epi4K Consortium, 2014). At leasy 20 heterozygous de novo variants have been reported in persons with drug-resistant seizures in infancy, profound intellectual disability, hypotonia of muscles, lack of speech, dystonia, and spasticity (von Spiczak et al., 2017; Li et al., 2019). DNM1 encodes a multimeric brain-specific GTPase, dynamin-1, which is located in the presynapse where it mediated endocytosis (Dhindsa et al., 2015). Aimiuwu et al. (2020) tested a Dnm1-targeted therapeutic microRNA contained in a self-complementaryAAV vector. The thrapeutic package is injected once into the cerebral-ventricular system of both cerebral hemispheres of the Dnm1Ftfl/Ftfl homozygotes at day 1 or at birth. The one-time injection reduced seizure severity, extended lifespan, improved growth, and abolished various developmental impairments. miDnm1, a type of treated mice, had significant reduction of Dnm1a mRNA 2 weeks postdelivery and did not show fibrillary gliosis and cell degeneration in CA1 of hippocampus. This preclinical study provided proof of principle that postnatal gene silencing by a microRNA targetted to Dmn1a can curb the fundamental symptomatology of DNM1 epileptic encephalopathy (Aimiuwu et al., 2020).

Unsilencing Paternal UBE3A by ASO in Human Angelman Syndrome

Meng et al. (2015) targeted the UBE3A-ATS with ASO treatment which led to RNase–H-mediated cleavage of the AS-RAN heteroduplex, thereby unsilencing the paternal UBE3A gene. Silva-Santos et al. (2015) and Sonzogni et al. (2020) then showed that unsilencing the paternal UBE3A gene around birth works best at improved behavioral phenotypes of Angelman syndrome. Two clinical trials, one by Hoffman-La Roche (NCTO442881) and the second by GeneTxBiotherapeutics (NCTO4259281), are currently active using the principle of unsilencing the paternal UBE3A gene (Elgersma, 2021).

Dravet Syndrome (Severe Myoclonic Epilepsy of Infancy)

The working hypothesis now is that febrile, afebrile generalized, unilateral, clonic and tonic clonic seizures in the first year of life, developmental delay evident in the second year of life, progressing cognitive decline, later followed by repeated bouts of status epilepticus, behavioral disorder, ataxia, and motor impairment in Dravet syndrome (Dravet, 1965, 1978, 1992, 2005a, 2005b; Delogu et al., 2011) are caused by loss-of-function variants in one allele of the SCN1A gene. This produces nonfunctional NaV1.1 units located in GaBAergic neurons, leading to epileptic seizures, the symptomatology of an encephalopathy, and risk of sudden death. ASOs are being explored as treatment in Dravet syndrome (Hill and Meisler, 2021; Han et al., 2020). Dravet syndrome, X-linked and found only in females is due to variants in protocadherin (Depienne et al., 2009).

Preclinical Studies Provide Proof of Principle That Symptomatology of Dravet Syndrome Can Be Reversed in a Conditional Knock-in Mouse Model

Valassina et al. (2022) generated an Scn1a conditional knock-in mouse model (Scn1astop/+), in which Scn1a can be reactivated on demand during the mouse lifetime. Here, Scn1a gene was disrupted by inserting a STOP cassette arrayed between loxP sites in the intron between exons 6 and 7. The STOP cassette was conditionally removed upon Cre recombinase expression to reconstitute the functional Scn1a allele. Valessina et al. (2022) show that Scn1a reactivation when symptoms were already manifest at P30 or at P90 led to complete rescue of both spontaneous and thermic induced seizures, marked amelioration of behavioraal abnormalities, and normalization of hippocampal fast-spiking interneuron firing.

Preclinical Studies Provide Proof of Concept That Genome Editing Exploiting dCas9-Based Gene Activation from a sgRNA Will Increase Scn1a Expression in Dravet Syndrome Pups and Suppress Seizures

Colasante et al. (2020) developed nuclease-dead Cas9 (dCas9) fused to effector domains such as the V16 transcriptor activator domians for transcription gene regulation. This platform manipulates transcription without cleaving the target. They then delivered the Scn1a-dCas9 activation system to Dravet syndrome pups using an adeno-associated virus vector. In the package they also included the Dlx5/6 gene activation system that exclusively tartgets forebrain interneurons. With this, Scn1a gene activation in cortical interneurons reduced and even totally suppressed temperature-induced epileptic seizures. A similar gene-editing approach is suggested for human Dravet syndrome.

The ASO Stoke Therapeutics Trial in Dravet Syndrome Patients (NCT04442295)

An ASO (STK-001) was developed to specifically target the SCN1A NMD or “poison exons” 20N and reduces nonproductive mRNA while increasing levels of productive mRNA and Nav1.1 sodium channel protein. (Hill & Meisler, 2021)The upregulated expression of wild-type SCN1A should compensatesfor the haploinsufficiency of SCN1A in Dravet syndrome. Preclinical studies had shown that a single intracerebral-ventricular injection at day 2 in the Scn1aTmKea heterozygous mice increased Scn1a mRNA and Nav1.1 protein expression, prevented SUDEP in 97% of DS mice, and suppressed seizure frequency and latency. (See Chapter 75, this volume.)

Early reports from an interim analysis hint that the intrathecal STK-001 medication may reduce seizure frequency among trial participants. As of November 23, 2021, it has so far raised no safety concerns among children taking part in the ongoing phase ½ MONARCH clinical trial. There is also an open-label extension study (NCT04740476), which will enroll 69 participants with an estimated study completion date of March 3, 2027.

Focal Epilepsies

Among the 40 to 60 million persons worldwide with epilepsies, focal epilepsies account for the greatest proportion. Even with optimal treatment with antiseizure medications, approximately 30% remain uncontrolled. Consequently, new treatment paradigms are being developed.

Epilepsy Gene Therapy Using an Engineered Potassium Channel Suppresses Focal Neocortical and Temporal Lobe Seizures in Rats

Snowball et al. (2019) codon-optimized the voltage-gated potassium channel Kv1.1 for human expression and mutated to accelerate recovery of the channel from inactivation. The engineered potassium channel gene was packaged in a nonintegratng lentiviral vector under the control of a cell-specific CAMK2A promoter. The potassium channel-lentiviral vector robustly reduced seizure frequency in a rat model of focal neocortical epilepsy during a blinded, randomized, placebo-controlled preclinical trial.

When packaged into an adeno-associated viral vector (AAV2/9), the engineered potassium channel gene was also effective at suppressing seizures in a male rat model of temporal lobe epilepsy. The authors conclude that the engineered potassium channel gene therapy was ready for clinical translation (Snowball et al., 2019).

Gene Therapy Vector Encoding Neuropeptide Y and Its Receptor Y2 Suppress Temporal Lobe Seizures: Preclinical Data in Rats

Szczygiel et al. (2020) used a vector that encodes the combination of neuropeptide Y and its antiepileptic receptor Y2 and administered the package unilaterally into the dorsal ventral hippocampus of adult male rats. Previous studies had shown that this package has antiepiletic effects in in vivo rodents. Combined overexpression of NPY and Y2 in the hippocampus had exerted a superior seizure-suppressant effect compared to single transgene expression (Cattaneo et al., 2020; Woldbye et al., 2005, 2010). In the present study, the expression of both transgenes was found to be elevated and remain without a decline at 6 months post-injection. The package mediated expression selectively in hippocampal neurons without expression in astrocytes and oligodendrocytes. No effects were observed on body weight, or on short-term or long-term memory as tested by the Y-maze SA test or the Morris water-maze test. Authors conclude that the combination of neuropeptide Y and Y2 receptor gene therapy is ready for future gene therapy in pharmaco-resistant temporal lobe epilepsy patients (Szczygiel et al., 2020). CombiGene in Lund, Sweden, reports their intention to start a clinical trial and treat focal epilepsies delivering NPY and Y2 receptors through an AAV approach (Binder et al., 2020).

Vigilance for Unexpected Adverse Effects of ASOs and GRT

Recently, the US FDA proclaimed that they expect to be approving 10 to 20 cell and gene therapies a year from 2025 onward (Jensen et al., 2021). It has been 32 years (September 14, 1990) since Anderson, Blaese, and Culver conducted the first clinical trial of human gene therapy at NIH on a 4-year-old girl with severe combined immune deficiency (SCID). The girl, reportedly, continues to do well, even after three decades. However, 9 years after that first gene therapy, the “Gelsinger case” occurred (Wilson, 2009). Jesse Gelsinger from Arizona suffered from ornithine transcarbamylase deficiency. He was treated with GRT that used a replication defective human adenovirus type 5 vector. The gene therapy package was injected into the Gelsinger patient’s hepatic artery, which then found its led to the liver. Four days later, Jesse Gelsinger died from multiple organ failures triggered by an immunogenic reaction from the adenovirus vector (Raper et al, 2003; Lehrman, 1999; Verma, 2000).

The research community has not forgotten this early decade (1990 to 2000) of the “stop and go” history of gene therapy, the stop periods causing careers of scientists to be derailed by NIH and FDA grant suspensions or stopped altogether (e.g., an institute director at Penn confined to animal research and no human studies) and even costing medical school deanships. Of course, this was complicated by the legal community’s exploitation of these unfortunate and unforeseen complications of gene therapy. In the research community, only the brave of heart have “soldiered on” in spite of the “harsh” lessons on adverse effects of gene therapy learned during its first decade, during the turn of the century (Thompson, 2000).

Aside from the notable unfortunate story of the 1999 Gelsinger case from Arizona, there was the 2002 LMO2 insertional mutagenesis causing “T-cell acute lymphoblastic leukemia case” from Paris. Eleven children in the Necker hospital in Paris were enrolled for GRT to treat X-linked severe combined immunodeficienct (X-SCID). A gamma-retroviral vector was used to package the wild-type gene. Among these 11 children, two of the youngest boys developed leukemia. The retroviral vector inserted the retrovirus genome close to the promoter of the LMO2 gene in the two boys’ genome. The promoter of LMO2 gene encodes a transcription factor, which caused the continous expression of LMO2 protein, which, in turn, led to the development of T-cell acute lymphoblastic leukemia (Hacein-Bey-Abina et al., 2003).

These GRT complications emphasized the short-term and long-term complications of GRT, genome integration issues, and concerns about long-term sustained safety. These adverse effects overshadowed the successful results of GRT in two patients with SCID in the United States (Thompson, 2000) and four patients in the United Kingdom (Gaspar et al, 2004), in patients with Hemophilia B (Factor IX deficiency) and in patients with Canavan disease (aspartoacylase deficiency) (Jiang et al., 2006; McPhee et al., 2006). Despite these setbacks, there has been continuing high hopes for GRT over the last decade because of safer AAV vectors, as discussed above, in NCL diseases. Vigilance remains for unexpected complications such as thrombotic microangiopathy with AVXS-101 and intrinsic uncertainties of GRT. Genome integration issues must be better understood. As an example, wild-type AAV integrates into a specific site in human chromosome 19 (Kotin et al., 1990, 1991). Unexpected sudden deaths from liver failure have been reported in clinical trials using systemically administered AAV vectors for X-linked myotubular myopathy where hepatic disease had not been previously recognized (Shieh et al., 2020), and hepatic dysfunctions have been observed even in intracerebral delivery of AAV (Merkel et al., 2015). The genome of AAV vectors when inserted into dog genes involved in cell growth may potentially lead to malignancies (Nguyen et al., 2021). These short-term and long-term complications of GRT, genome integration issues, the insertion, deletion, or replacement of nucleotides during genome editing, the immunogenicity of their genome editing tools, on- and off–target modifications, and concerns about long-term sustained safety should be part of the discussions during the consenting process of parents and affected persons (Gaspar et al., 2004; Jensen et al., 2021; Wang et al., 2007; Wang & Gao, 2014).

Key Social and Ethical Issues in the Quest to Cure the Epilepsies

General Concerns

The ascendance of therapeutic agents that can halt the progression of disease, reverse symptoms, and cure what was previously considered incurable fatal epilepsies raise a variety of issues on equity, ethics, and the consenting process with regard to the treating physician, the pharmaceutical industry, and the patients and their families (Krauss et al., 2000; Barns et al., 2000; Temkin, 1971; Jacoby, 2002, 2004; Shostak and Ottman, 2006; Ngugi et al., 2010; West, 1979).

Equity in Availability and Affordability

The expensive and novel therapeutic agents are a first concern (Gehlert et al., 2000). For example, will antisense oligonucleotides (e.g., intrathecal STK-001; IONIS intrathecal Gys1-ASO), GRT (e.g., intracerebral-ventricular cerliponase alfa [Brineura]; Amicus single lifetime intrathecal AT-GTX-502), and special molecules such as the pancreatic alpha-amylase bound to a cell-penetrating monoclonal antibody (VAL-0417) be available to patients with rare fatal epilepsies who are located around the world, including countries with low socioeconomic status? This is an important first concern.

Parents Rely on the Treating Physician

To make sense of the benefits and risks of the therapy being offered, parents will often rely on the treating doctor for advice. When making the final decision, parents often ask, “On- and off-targets . . . what is that?” “Do you mean I have to wait ten years to see if my child develops leukemia?” “What would you do if it were your child?” A significant addition to the burden of being diagnosed with a fatal form of epilepsy is the complexity of information on the therapeutic agent being offered as treatment. This can amplify the stress parents are undergoing, even though they feel relief that finally a curative agent has become available. Emerging information on adverse effects of the treatment offered is anxiety building enough, and the honest admission by the treating physician that unknown, previously unrecognized complications may be lurking somewhere during the long process of monitoring adverse effects (e.g., 10 years follow-up after a single lifetime intrathecal injection of GRT) can be too much to bear.

Difficulties for Parents Treating an Asymptomatic Offspring

The child may carry the epilepsy-causing variant, but the full array of the therapy’s complications is still unknown. Because there is a lack of social scientific literature, guidelines, or policies from government agencies such as the National Human Genome Research Institute (NHGRI), societies representing medical genetics professionals (e.g., American Society of Human Genetics/American College of Medical Genetics), or the Recombinant DNA Advisory Committee (a forum for genetic engineering debates), which addresses the particular social dimensions that confront families, parents, and their affected child, the treating physician often turns to workshops on the subject of ethics in gene therapy and the experience of human ethics committees and institutional review boards (IRBs) of universities for discussion and advise.

Importantly, it is not a perfect science. It is difficult to determine whether parents and affected persons have a clear and total understanding of the burden and benefits of treatment. Can they understand what a seemingly elevated ammonia level without other laboratory signs of liver dysfunction means, and what signals of adverse effects in preclinical trials of nonhuman simians should contraindicate the human trial?

In the often-quoted Gelsinger case (Thompson, 2000; Verma, 2000), the FDA, in its investigation, claims that one contribution to Gelsinger’s death was “incomplete disclosure of the potential risks of Jesse’s treatment to his family. . . .Gelsinger’s high ammonia levels at the time of treatment should have excluded him from the study. . . . Two patients who had experienced serious side effects from gene therapy were not immediately reported and the deaths of monkeys given the similar treatment were never included in the informed consent discussion.” In those early years of gene therapy, it is difficult to blame the investigators when the antenna for possible complications signaled by the sole sign of an elevated ammonia level and the “dots/signs” of possible complications appearing during the preclinical trials and the “dots/signs” of possible complications during the human trial were not being connected. These were all done, of course, “post hoc” after liver organ failure caused the death of Gelsinger.

It is also difficult to decide what they, as parents, would consider acceptable results of treatment in the context of the neurologically debilitating and invariably fatal nature of their epilepsy syndrome being treated.

Costs: Who Should Pay?

To incentivize biopharmaceutical companies to develop treatments for previously incurable rare diseases and neurodegenerative diseases such as Alzheimer disease, Parkinson disease, and amyotrophic lateral sclerosis, the FDA and EMA offer the prospect of fast-track designations and longer market exclusivity with their patents. For rare diseases and their small market, biopharmaceutical companies are forced to price their novel therapeutics with very high prices, many-fold higher than that offered for treatment modalities for common diseases. This necessarily limits the number of patients who can receive these novel therapeutics. Which brings us to the first concern discussed earlier, namely, equity (Jensen et al., 2021).

The COVID-19 pandemic has brought a new problem to the public and exacerbated the issue of equity. Some “start-up” pharmaceutical companies have gone bankrupt because of the pandemic. Even well-established large pharmaceutical companies have had to roll back or actually stop funds during preclinical experiments and clinical trials. The public asks, “Where should the responsibility lie?” Should it lie with the government agencies, like the NIH, or with the patient advocacy organizations and the nonprofit lay associations?

It will be important for the drug developers, pharma industry, investors, public agencies, and medical insurance companies, the advocacy organizations, and the families of patients with rare epilepsy diseaess and now the more frequent developmental epileptic encephalopathies to work together to mold new models of pricing and reimbursements to ensure that these novel curative therapeutics reach the patients in need (Jensen et al., 2021).

References

  1. Abdelnour E, Gallentine W, McDonald M, Sachdev M, Jiang YH, Mikati MA.  Does age affect response to quinidine in patients with KCNT1 mutations? Report of three new cases and reveiw of the literature.  Seizure. 2018 Feb;55:1–3. doi: 10.1016/j.seizure.2017.11.017 PMID: 29291456 [PubMed: 29291456]
  2. Ahonen S, Nitschke S, Grossman TR, Kordasiewicz H, Wang P, Zhao X, Guisso DR, Kasiri S, Nitschke F, Minassian BA.  Gys1 antisense therapy rescues neuropathological bases of murine Lafora disease. Brain. 2021 Oct; 144(10): 2985–2993. PMID: 33993268 doi :10.1093/brain/awab194 [PMC free article: PMC8634080] [PubMed: 33993268]
  3. Aimiuwu OV, Fowler AM, Sah M, Teoh JJ, Kanber A, Pyne NK, Petri S, Rosenthal-Weiss C, Yang M, Harper SQ, Frankel WN.  RNAi-based gene therapy rescues developmental and epileptic encephalopathy in a genetic mouse model.  Mol Ther. 2020 Jul 8;28(7):1706–1716. doi: 10.1016/j.ymthe.2020.04.007. PMID: 32353324 [PMC free article: PMC7335739] [PubMed: 32353324]
  4. Epi4K Consortium; Epilepsy Phenome/Genome Project, Allen AS, Berkovic SF, Cossette P, Delanty N, Dlugos D, Eichler EE, Epstein MP, Glauser T, Goldstein DB, Han Y, Heinzen EL, Hitomi Y, Howell KB, Johnson MR, Kuzniecky R, Lowenstein DH, Lu YF, Madou MR, Marson AG, Mefford HC, Esmaeeli Nieh S, O’Brien TJ, Ottman R, Petrovski S, Poduri A, Ruzzo EK, Scheffer IE, Sherr EH, Yuskaitis CJ, Abou-Khalil B, Alldredge BK, Bautista JF, Berkovic SF, Boro A, Cascino GD, Consalvo D, Crumrine P, Devinsky O, Dlugos D, Epstein MP, Fiol M, Fountain NB, French J, Friedman D, Geller EB, Glauser T, Glynn S, Haut SR, Hayward J, Helmers SL, Joshi S, Kanner A, Kirsch HE, Knowlton RC, Kossoff EH, Kuperman R, Kuzniecky R, Lowenstein DH, McGuire SM, Motika PV, Novotny EJ, Ottman R, Paolicchi JM, Parent JM, Park K, Poduri A, Scheffer IE, Shellhaas RA, Sherr EH, Shih JJ, Singh R, Sirven J, Smith MC, Sullivan J, Lin Thio L, Venkat A, Vining EP, Von Allmen GK, Weisenberg JL, Widdess-Walsh P, Winawer MR.  De novo mutations in epileptic encephalopathies.  Nature. 2013 Sep 12;501(7466):217–221. PMID: 23934111 PMCID: PMC3773011 DOI: 10.1038/nature12439 [PMC free article: PMC3773011] [PubMed: 23934111]
  5. Al-Zaidy S, Pickard AS, Kotha K, Alfano LN, Lowes L, Paul G, Church K, Lehman K, Sproule DM, Dabbous O, Maru B, Berry K, Arnold WD, Kissel JT, Mendell JR, Shell R.  Health outcomes in spinal muscular atrophy type 1 following AVXS-101 gene replacement therapy.  Pediatr Pulmonol.  2019a Feb; 54(2):179–185. PMID: 30548438 PMCID: PMC6590370 DOI: 10.1002/ppul.2420 [PMC free article: PMC6590370] [PubMed: 30548438]
  6. Al-Zaidy SA, Kolb SJ, Lowes L, Alfano LN, Church KR, Nagendran S, Sproule DM, Feltner DE, Wells C, Ogrinc F, Menier M, L’Italien J, Arnold WD, Kissel JT, Kaspar BK, Mendell JR.  AVXS-101 (onasemnogene abeparvovec) for SMA1: comparative study with a prospective natural history cohort.  J Neuromuscul Dis.  2019b;6(3):307–317. PMID: 31381526 doi: 10.3233/JND-190403 [PubMed: 31381526]
  7. Aragon-Gawinska K, Seferian AM, Daron A, Gargaun E, Vuillerot C, Cances C, Ropars J, Chouchane M, Cuppen I, Hughes I, Illingworth M, Marini-Bettolo C, Rambaud J, Taytard J, Annoussamy M, Scoto M, Gidaro T, Servais L.  Nusinersen in spinal muscular atrophy type 1 patients older than 7 months : a cohort study.  Neurology. 2018 Oct 2;91(14):e1312–e1318. doi: 10.1212/WNL.0000000000006281 PMID: 30158155 [PubMed: 30158155]
  8. Bailey JN, Patterson C, de Nijs L, Durón RM, Nguyen VH, Tanaka M, Medina MT, Jara-Prado A, Martínez-Juárez IE, Ochoa A, Molina Y, Suzuki T, Alonso ME, Wight JE, Lin YC, Guilhoto L, Targas Yacubian EM, Machado-Salas J, Daga A, Yamakawa K, Grisar TM, Lakaye B, Delgado-Escueta AV.  EFHC1 variants in juvenile myoclonic epilepsy: reanalysis according to NHGRI and ACMG guidelines for assigning disease causality,  Genet Med. 2017 Feb; 19(2):144–156. doi: 10.1038/gim.2016.86 PMID: 27467453 [PubMed: 27467453]
  9. Bailey JN, de Nijs L, Bai D, Suzuki T, Miyamoto H, Tanaka M, Patterson C, Lin YC, Medina MT, Alonso ME, Serratosa JM, Durón RM, Nguyen VH, Wight JE, Martínez-Juárez IE, Ochoa A, Jara-Prado A, Guilhoto L, Molina Y, Yacubian EM, López-Ruiz M, Inoue Y, Kaneko S, Hirose S, Osawa M, Oguni H, Fujimoto S, Grisar TM, Stern JM, Yamakawa K, Lakaye B, Delgado-Escueta AV.  Variant intestinal-cell kinase in juvenile myoclonic epilepsy.  N Engl J Med. 2018 Mar 15; 378(11):1018–1028. doi: 10.1056/NEJMoa1700175 PMID: 29539279 [PubMed: 29539279]
  10. Barns I, Schibecci R, Davison A, Shaw R.  “What do you think about genetic medicine?” Facilitating sociable public discourse on developments in the new genetics.  Science, Technol Human Values.  2000; 25:283–308. doi: 10.1177/016224390002500302. PMID: 15362239 [PubMed: 15362239]
  11. Baulac S, Huberfeld G, Gourfinkel-An I, Mitropoulou G, Beranger A, Prud’Homme JF, Baulac M, Brice A, Bruzzone R, LeGuern E.  First genetic evidence of GABA(A) receptor dysfunction in epilepsy: a mutation in the gamma2-subunit gene.  Nat Genet. 2001 May; 28(1):46–48. PMID: 11326274, DOI: 10.1038/ng0501-46 [PubMed: 11326274]
  12. Bertini E, Hwu WL, Reyna SP et al. Efficacy and safety of nusinersen in infants with presymptomatic spinal muscular atrophy (SMA): interim results from the NURTURE study,  Eur J Pediatri Neurol.  2017: 21(S1): e14.
  13. Biervert C, Steinlein OK.  Structural and mutational analysis of KCNQ2, the major gene locus for benign familial neonatal convulsions.  Hum Genet. 1999 Mar;104(3):234–240. doi: 10.1007/pl00008713 PMID: 10323247 [PubMed: 10323247]
  14. Biervert C, Schroeder BC, Kubisch C, Berkovic SF, Propping P, Jentsch TJ, Steinlein OK. A potassium channel mutation in neonatal human epilepsy. Science. 1998 Jan 16;279(5349):403–406. PMID: 9430594 DOI: 10.1126/science.279.5349.403 [PubMed: 9430594]
  15. Binder DK, Boison D, Eid T, Frankel WN, Mingorance A, Smith BN, Dacks PA, Whittemore V, Poduri A, AES/NINDS epilepsy Brenchmarks Stewards. Epilepsy Benchmarks Area II: Prevent Epilepsy and Its Progression.  Epilepsy Curr. 2020 Jan-Feb; 20(S1):14S–22S. PMID: 31937124 PMCID: PMC7031802 DOI: 10.1177/1535759719895274 [PMC free article: PMC7031802] [PubMed: 31937124]
  16. Biffi A, Montini E, Lorioli L, Cesani M, Fumagalli F, Plati T, Baldoli C, Martino S, Calabria A, Canale S, Benedicenti F, Vallanti G, Biasco L, Leo S, Kabbara N, Zanetti G, Rizzo WB, Mehta NA, Cicalese MP, Casiraghi M, Boelens JJ, Del Carro U, Dow DJ, Schmidt M, Assanelli A, Neduva V, Di Serio C, Stupka E, Gardner J, von Kalle C, Bordignon C, Ciceri F, Rovelli A, Roncarolo MG, Aiuti A, Sessa M, Naldini L.  Lentiviral hematopoietic stem cell gene therapy benefits metachromatic leukodystrophy.  Science. 2013 Aug 23;341(6148):1233158. doi: 10.1126/science.1233158 PMID: 23845948 [PubMed: 23845948]
  17. Bingham PM, Lynch D, McDonald-McGinn D, Zackai E.  Polymicrogyria in chromosome 22 deletion syndrome.  Neurology. 1998 Nov;51(5):1500–1502. doi: 10.1212/wnl.51.5.1500 PMID: 9818897 [PubMed: 9818897]
  18. Cain JT, Likhite S, White KA, Timm DJ, Davis SS, Johnson TB, Dennys-Rivers CN, Rinaldi F, Motti D, Corcoran S, Morales P, Pierson C, Hughes SM, Lee SY, Kaspar BK, Meyer K, Weimer J.  Gene therapy corrects brain and behavioral pathologies in CLN6-Batten disease.  Mol Ther. 2019 Oct 2;27(10):1836–1847. doi: 10.1016/j.ymthe.2019.06.015 PMID: 31331814 [PMC free article: PMC6822284] [PubMed: 31331814]
  19. Calbi V, Fumagalli F, Sessa M, Zambon A, Baldoli C, Cugnata F, et al. Lentiviral hematopoietic stem and progenitor cell gene therapy (HSPC- GT) for metachromatic leukodystrophy (MLD): clinical outcomes from 33 patients. The 46th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians Oral Session (O010–O173).  Bone Marrow Transplant.  2020; 55, 22–174. doi: 10.1038/s41409-020-01119-3 [PMC free article: PMC7729707] [PubMed: 33311610]
  20. Carvill GL, Heavin SB, Yendle SC, McMahon JM, O’Roak BJ, Cook J, Khan A, Dorschner MO, Weaver M, Calvert S, Malone S, Wallace G, Stanley T, Bye AM, Bleasel A, Howell KB, Kivity S, Mackay MT, Rodriguez-Casero V, Webster R, Korczyn A, Afawi Z, Zelnick N, Lerman-Sagie T, Lev D, Møller RS, Gill D, Andrade DM, Freeman JL, Sadleir LG, Shendure J, Berkovic SF, Scheffer IE, Mefford HC.  Targeted resequencing in epileptic encephalopathies identifies de novo mutations in CHD2 and SYNGAP1.  Nat Genet 2013 Jul.45(7):825–830. doi: 10.1038/ng.2646 PMID: 23708187 [PMC free article: PMC3704157] [PubMed: 23708187]
  21. Cattaneo S, Verlengia G, Marino P, Simonato M, Bettegazzi B.  NPY and gene therapy for epilepsy: how, when,… and Y. Front.  Mol. Neurosci. 2020 Jan 22;13:608001. PMID: 33551745 doi: 10.3389/fnmol.2020.608001 [PMC free article: PMC7862707] [PubMed: 33551745]
  22. Cavalleri GL, Walley NM, Soranzo N, Mulley J, Doherty CP, Kapoor A, Depondt C, Lynch JM, Scheffer IE, Heils A, Gehrmann A, Kinirons P, Gandhi S, Satishchandra P, Wood NW, Anand A, Sander T, Berkovic SF, Delanty N, Goldstein DB, Sisodiya SM.  A multicenter study of BRD2 as a risk factor for juvenile myoclonic epilepsy.  Epilepsia.  2007a April;48(4):706–712. PMID: 17437413 DOI: 10.1111/j.1528-1167.2007.00977.x [PubMed: 17437413]
  23. Cavalleri GL, Weale ME, Shianna KV, Singh R, Lynch JM, Grinton B, Szoeke C, Murphy K, Kinirons P, O’Rourke D, Ge D, Depondt C, Claeys KG, Pandolfo M, Gumbs C, Walley N, McNamara J, Mulley JC, Linney KN, Sheffield LJ, Radtke RA, Tate SK, Chissoe SL, Gibson RA, Hosford D, Stanton A, Graves TD, Hanna MG, Eriksson K, Kantanen AM, Kalviainen R, O’Brien TJ, Sander JW, Duncan JS, Scheffer IE, Berkovic SF, Wood NW, Doherty CP, Delanty N, Sisodiya SM, Goldstein DB.  Multicenter search for genetic susceptibility loci in sporadic epilepsy syndrome and seizure types: a case-control study.  Lancet Neurol.  2007b Nov;6(11):970–980. doi: 10.1016/S1474-4422(07)70247-8. PMID: 17913586 [PubMed: 17913586]
  24. Chan EM, Young EJ, Ianzano L, Munteanu I, Zhao X, Christopoulos CC, Avanzini G, Elia M, Ackerley CA, Jovic NJ, Bohlega S, Andermann E, Rouleau GA, Delgado-Escueta AV, Minassian BA, Scherer SW.  Mutations in NHLRC1 cause progressive myoclonus epilepsy.  Nat Genet. 2003 Oct;35(2):125–127. doi: 10.1038/ng1238. PMID: 12958597 [PubMed: 12958597]
  25. Charlier C, Singh NA, Ryan SG, Lewis TB, Reus BE, Leach RJ, Leppert M.  A pore mutation in a novel KQT-like potassium channel gene in an idiopathic epilepsy family.  Nat Genet. 1998 Jan;18(1):53–55. PMID: 9425900 DOI: 10.1038/ng0198-53 [PubMed: 9425900]
  26. Chen TH.  New and developing therapies in spinal muscular atrophy : From genotype, penotype to treatment and where do we stand?  Int J Mol Sci.  2020;21:3297. PMID: 32392694 doi: 10.3390/ijms21093297 [PMC free article: PMC7246502] [PubMed: 32392694]
  27. Chen Y, Lu J, Pan H, Zhang Y, Wu H, Xu K, Liu X, Jiang Y, Bao X, Yao Z, Ding K, Lo WHY, Qiang B, Chan P, Shen Y, Wu X.  Association between genetic variation of CACNA1H and childhood absence epilepsy.  Ann Neurol. 2003 Aug;54(2):239–243. PMID: 12891677 DOI: 10.1002/ana.10607 [PubMed: 12891677]
  28. Chiriboga CA, Swoboda KJ, Darras BT, Iannaccone ST, Montes J, De Vivo DC, Norris DA, Bennett CF, Bishop KM.  Results from a phase 1 study of nusinersen (ISIS-SMN(Rx)) in children with spinal muscular atrophy.  Neurology. 2016 Mar 8;86(10):890–897. doi: 10.1212/WNL.0000000000002445. PMID: 26865511 [PMC free article: PMC4782111] [PubMed: 26865511]
  29. Chu Y, Bartus RT, Manfredsson FP, Olanow CW, Kordower JH.  Long-term post-mortem studies following neurturin gene therapy in patients with advanced Parkinson’s disease.  Brain.  2020;143:960–975. doi: 10.1093/brain/awaa020 [PMC free article: PMC7089653] [PubMed: 32203581]
  30. Claes L, Del-Favero J, Ceulemans B, Lagae L, Van Broeckhoven C, De Jonghe P.  De Novo mutations in the sodium-channel gene SCN1A cause severe myoclonic epilepsy of infancy.  Am J Hum Genet. 2001 Jun;68(6):1327–1332. doi: 10.1086/320609 PMID: 11359211 [PMC free article: PMC1226119] [PubMed: 11359211]
  31. Colasante G, Lignani G, Brusco S, Berardino CD, Carpenter J, Giannelli S, Valassina N, Bido S, Ricci R, Castoldi V, Marenna S, Church T, Massimino L, Morabito G, Benfenati F, Schorge S, Leocani L, Kullmann DM, Broccoli V.  dCas9-based Scn1a gene activation restores inhibitory interneuron excitability and attenuates seizures in Dravet syndrome mice.  Mol Ther. 2020 Jan 8;28(1):235–253. doi: 10.1016/j.ymthe.2019.08.018 PMID: 31607539 [PMC free article: PMC6952031] [PubMed: 31607539]
  32. Corbett MA, Bahlo M, Jolly L, Afawi Z, Gardner AE, Oliver KL, Tan S, Coffey A, Mulley JC, Dibbens LM, Simri W, Shalata A, Kivity S  Jackson GD, Berkovic SF, Gecz J.  A focal epilepsy and intellectual disability syndrome is due to a mutation in TBC1D24.  Am J Hum Genet. 2010 Sep 10; 87(3):371–375. doi: 10.1016/j.ajhg.2010.08.001. PMID: 20797691 [PMC free article: PMC2933342] [PubMed: 20797691]
  33. Cossette P, Liu L, Brisebois K, Dong H, Lortie A, Vanasse M, Saint-Hilaire J-M, Carmant L, Verner A, Lu W-Y, Wang YT, Rouleau GA.  Mutation of GABRA1 in an autosomal dominant form of juvenile myoclonic epilepsy.  Nat Genet. 2002 Jun;31(2):184–189. PMID: 11992121 DOI: 10.1038/ng885 [PubMed: 11992121]
  34. Cristescu ME.  The concept of genome after one century of usage,  Genome.  2019; 62 : iii–v. NRC Research Press, dx.doi.org/10.1139/gen-2019-0129 [PubMed: 31565997]
  35. Dabbous O, Maru B, Jansen JP, Lorenzi M, Cloutier M, Guérin A, Pivneva I, Wu EQ, Arjunji R, Feltner D, Sproule DM.  Survival motor function, and motor milestones: comparison of AVXS-101 relative to nusinersen for the treatment of infants with spinal muscular atrophy Type 1.  Adv Ther. 2019 May;36(5):1164–1176. doi: 10.1007/s12325-019-00923-8 PMID: 30879249 [PMC free article: PMC6824368] [PubMed: 30879249]
  36. Day JW, Finkel RS, Chiriboga CA, Connolly AM, Crawford TO, Darras BR  Iannaccone ST, Kuntz NL, Peña LDM, Shieh PB, Smith EC, Kwon JM, Zaidman CM, Schultz M, Feltner DE, Tauscher-Wisniewski S, Ouyang H, Chand DH, Sproule DM, Macek TA, Mendell JR.  Onasemnogene abeparvovec gene therapy for symptomatic infantile-onset spinal muscular atrophy in patients with two copies of SMN2 (STR1VE): an open label, single arm, multicentre, phase 3 trial.  Lancet Neurol. 2021 Apr;20(4):284–293. doi: 10.1016/S1474-4422(21)00001-6. PMID: 33743238 [PubMed: 33743238]
  37. De Fusco M, Becchetti A, Patrignani A, Annesi G, Gambardella A, Quattrone A, Ballabio A, Wanke E, Casari G.  The nicotinic receptor beta 2 subunit is mutant in nocturnal frontal lobe epilepsy.  Nat Genet. 2000 Nov;26(3):275–276. doi: 10.1038/81566. PMID: 11062464 [PubMed: 11062464]
  38. de Haan GJ, Halley DJ, Doelman JC, Geesink HH, Augustijn PB, Jager-Jongkind AD, Majoie M, Bader AJ, Leliefeld-Ten Doeschate LAWM, Deelen WH, Bertram E, Lehesjoki AE, Lindhout D.  Unverricht-Lundborg disease: underdiagnosed in the Netherlands.  Epilepsia. 2004 Sep;45(9):1061–1063. PMID: 15329070 DOI: 10.1111/j.0013-9580.2004.43703.x [PubMed: 15329070]
  39. de Nijs L, Léon C, Nguyen L, Loturco JJ, Delgado-Escueta AV, Grisar T, Lakaye B.  EFHC1 interacts with microtubules to regulate cell division and cortical development.  Nat Neurosci. 2009 Oct;12(10):1266–1274. doi: 10.1038/nn.2390 PMID: 19734894 [PubMed: 19734894]
  40. Delogu AB, Spinelli A, Battaglia D, Dravet C, De Nisco A, Saracina A, Romagnoli C, Lanza GA, Crea F.  Electrical and autonomic cardiac function in patients with Dravet syndrome.  Epilepsia. 2011 Apr;52(S2):55–58. PMID: 21463281 DOI: 10.1111/j.1528-1167.2011.03003.x [PubMed: 21463281]
  41. Depienne C, Bouteiller D, Keren B, Cheuret E, Poirier K, Trouillard O, Benyahia B, Quelin C, Carpentier W, Julia S, Afenjar A, Gautier A, Rivier F, Meyer S, Berquin P, Hélias M, Py I, Rivera S, Bahi-Buisson N, Gourfinkel-An I, Cazeneuve C, Ruberg M, Brice A, Nabbout R, Leguern E.  Sporadic infantile epileptic encephalopathy caused by mutations in PCDH19 resembles Dravet syndrome but mainly affects females.  PLoS Genet. 2009 Feb;5(2):e1000381. doi: 10.1371/journal.pgen.1000381 PMID: 19214208 [PMC free article: PMC2633044] [PubMed: 19214208]
  42. Deprez L, Weckhuysen S, Holmgren P, Suls A, Van Dyck T, Goossens D, Del-Favero J, Jansen A, Verhaert K, Lagae L, Jordanova A, Van Coster R, Yendle S, Berkovic SF, Scheffer I, Ceulemans B, De Jonghe P.  Clinical spectrum of early-onset epileptic encephalopathies associated with STXBP1 mutations.  Neurology. 2010 Sep 28;75(13):1159–1165. doi: 10.1212/WNL.0b013e3181f4d7bf PMID: 20876469 [PubMed: 20876469]
  43. Dhindsa RS, Bradrick SS, Yao X, Heinzen EL, Petrovski S, Krueger BJ, Johnson MR, Frankel WN, Petrou S, Boumil RM, and Goldstein DB.  Epileptic encephalopathy-causing mutations in DNM1 impair synaptic vesicle endocytosis.  Neurol. Genet. 2015 Apr 17;1(1):e4. PMID: 27066543 PMCID: PMC4821085 DOI: 10.1212/01.NXG.0000464295.65736.da [PMC free article: PMC4821085] [PubMed: 27066543]
  44. Dibbens LM, Feng HJ, Richards MC, Harkin LA, Hodgson BL, Scott D, Jenkins M, Petrou S, Sutherland GR, Scheffer IE, Berkovic SF, Macdonald RL, Mulley JC.  GABRD encoding a protein for extra- or peri-synaptic GABAA receptors is a susceptibility locus for generalized epilepsies.  Hum Mol Genet. 2004 Jul 1;13(13):1315–1319. PMID: 15115768 DOI: 10.1093/hmg/ddh146 [PubMed: 15115768]
  45. Dibbens LM, Michellucci R, Gambardella A, Andermann F, Rubboli G, Bayly MA, Joensuu T, Vears DF, Franceschetti S, Canafoglia L, Wallace R, Bassuk AG, Power DA, Tassinari CA, Andermann E, Lehesjoki AE, Berkovic SF.  SCARB2 mutations in progressive myoclonus epilepsy (PME) without renal failure.  Ann Neurol. 2009 Oct;66(4):532–536. doi: 10.1002/ana.21765. PMID: 19847901 [PubMed: 19847901]
  46. Dibbens LM, Mullen S, Helbig I, Mefford HC, Bayly MA, Bellows S, Leu C, Trucks H, Obermeier T, Wittig M, Franke A, Caglayan H, Yapici Z; EPICURE Consortium, Sander T, Eichler EE, Scheffer IE, Mulley JC, Berkovic SF.  Familial and sporadic 15q13.3 microdeletions in idiopathic generalized epilepsy: precedent for disorders with complex inheritance.  Hum Mol Genet. 2009 Oct 1;18(19):3626–3631. doi: 10.1093/hmg/ddp311 PMID: 19592580 [PMC free article: PMC3465696] [PubMed: 19592580]
  47. Dibbens LM, Tarpey PS, Hynes K, Bayly MA, Scheffer IE, Smith R, Bomar J, Sutton E, Vandeleur L, Shoubridge C, Edkins S, Turner SJ, Stevens C, O’Meara S, Tofts C, Barthorpe S, Buck G, Cole J, Halliday K, Jones D, Lee R, Madison M, Mironenko T, Varian J, West S, Widaa S, Wray P, Teague J, Dicks E, Butler A, Menzies A, Jenkinson A, Shepherd R, Gusella JF, Afawi Z, Mazarib A, Neufeld MY, Kivity S, Lev D, Lerman-Sagie T, Korczyn AD, Derry CP, Sutherland GR, Friend K, Shaw M, Corbett M, Kim HG, Geschwind DH, Thomas P, Haan E, Ryan S, McKee S, Berkovic SF, Futreal PA, Stratton MR, Mulley JC, Gécz J.  X-linked protocadherin 19 mutations cause female-limited epilepsy and cognitive impairment.  Nat Genet. 2008 Jun;40(6):776–781. doi: 10.1038/ng.149 PMID: 18469813 [PMC free article: PMC2756413] [PubMed: 18469813]
  48. Dobyns WB, Reiner O, Carrozzo R, Ledbetter DH.  Lissencephaly: A human brain malformation associated with deletion of the LIS1 gene located at chromosome 17p13.  JAMA. 1993 Dec 15;270(23):2838–2842. doi: 10.1001/jama.270.23.2838 PMID: 7907669 [PubMed: 7907669]
  49. Dravet C.  Encéphalopathie épileptique de l’enfant avec Pointe-Onde lente diffuse (Petit Mal Variant). Thesis, 1965, Marseille. [PubMed: 4958167]
  50. Dravet C.  Les épilepsies graves de l’enfant.  Vie Med.  1978; 8: 543–548.
  51. Dravet C, Bureau M, Guerrini R, Giraud N, Roger J.  Severe myoclonic epilepsy in infants. In: Roger J, Bureau M, Dravet C, Dreifuss FE, Perret A, Wolf P (eds) Epileptic Syndromes in Infancy, Childhood and Adolescence, 2nd ed, 1992; pp. 75–88. London: John Libbey & Company Ltd.
  52. Dravet C, Bureau M, Oguni H, Fukuyama Y, Cokar O.  Severe myoclonic epilepsy in infancy: Dravet syndrome.  Adv Neurol.  2005;95:71–102. PMID: 15508915 [PubMed: 15508915]
  53. Dravet C, Bureau M, Oguni H, Fukuyama Y, Cokar O.  Severe myoclonic epilepsy in infancy. In: Roger J, Bureau M, Dravet C, Genton P, Tassinari CA, Wolf P (eds) Epileptic Syndromes in Infancy, Childhood and Adolescence, 4th ed, 2005a; pp. 89–113. Paris: John Libbey Eurotext.
  54. Elgersma Y, Sonzogni M.  UBE3A reinstatement as a disease- modifying therapy for Angelman syndrome.  Dev. Med. Child Neurol. 2021 Jul; 63(7), 802–807. doi: 10.1111/dmcn.14831 PMCID: PMC8248324 PMID: 33543479 [PMC free article: PMC8248324] [PubMed: 33543479]
  55. Escayg A, MacDonald BT, Meisler MH, Baulac S, Huberfeld G, An-Gourfinkel I, Brice A, LeGuern E, Moulard B, Chaigne D, Buresi C, Malafosse A.  Mutations of SCN1A, encoding a neuronal sodium channel in two families with GEFS+2.  Nat Genet. 2000 Apr;24(4):343–345. doi: 10.1038/74159. PMID: 10742094 [PubMed: 10742094]
  56. Escayg A, Heils A, MacDonald BT, Haug K, Sander T, Meisler MH.  A novel SCN1A mutation associated with generalized epilepsy with febrile seizures plus -- and prevalence of variants in patients with epilepsy.  Am J Hum Genet. 2001 Apr;68(4):866–873. doi: 10.1086/319524. PMID: 11254445 [PMC free article: PMC1275640] [PubMed: 11254445]
  57. EuroEPINOMICS-RES Consortium, Epilepsy Phrnomr/Genome Project and Epi4K Consortium. De Novo Mutations in Synaptic transmission Genes including DNM1 Cause Epileptic Encephalopathies.  American Journal of Human Genetics. 2014 Oct 2;95:360–370. [PMC free article: PMC4185114] [PubMed: 25262651]
  58. Falace A, Filipello F, La Padula V, Vanni N, Madia F, De Pietri Tonelli D, e Falco FA, Striano P, Bricarelli FD, Minetti C, Benfenati F, Fassio A, Zara F.  TBC1D24, an ARF6-Interacting protein, is mutated in familial infantile myoclonic epilepsy.  Am J Hum Genet. 2010 Sep 10;87(3):365–370. PMID: 20727515 PMCID: PMC2933335 DOI: 10.1016/j.ajhg.2010.07.020 [PMC free article: PMC2933335] [PubMed: 20727515]
  59. Finkel RS, Chiriboga CA, Vajsar J, Day JW, Montes J, De Vivo DC, Yamashita M, Rigo F, Hung G, Schneider E, Norris DA, Xia S, Bennett CF, Bishop KM.  Treatment of infantfile-onset spinal muscular atrophy with nusinersen : a phase 2, open-label, dose-escalation study.  Lancet. 2016 Dec 17; 388(10063):3017–3026. doi: 10.1016/S0140-6736(16)31408-8. PMID: 27939059 [PubMed: 27939059]
  60. Finkel RS, Day JW, Darras BR, Kuntz NL, Connolly AM, Crawford RJ [more authors] Spector SA, Feltner DE, Mendell JR.  Intrathecal administration of AVXS-101 gene-replacement therapy (GRT) for spinal muscular atrophy type 2 (SMA1): phase 1/2A study (strong).  J Neurol Sci. 2019 Oct 15;405(S57):103984. doi: 10.1016/j.jns.2019.10.322
  61. Finkel RS, Day JW, Darras BT, Kuntz NL, Connoly AM, Crawford T et al. One-time intrathecal (IT) administration of AVXS-101 IT gene-replacement therapy for spinal muscular atrophy: phase 1 study (STRONG).  Neurology.  2020; 94(S15): available online https://n​.neurology.org​/content/94/15_Supplement/2493
  62. Finkel RS, Mercuri E, Darras BT, Connolly AM, Kuntz NL, Kirschner J  Chiriboga CA, Saito K, Servais L, Tizzano E, Topaloglu H, Tulinius M, Montes J, Glanzman AM, Bishop K, Zhong ZJ, Gheuens S, Bennett CF, Schneider E, Farwell W, De Vivo DC; ENDEAR Study Group. Nusinersen versus sham control in infantile-onset spinal muscular atrophy.  N Engl J Med. 2017 Nov 2;377(18):1723–1732. doi: 10.1056/NEJMoa1702752 PMID: 29091570 [PubMed: 29091570]
  63. Fumagalli F, Calbi V, Natali Sora MG, Sessa M, Baldoli C, Rancoita PMV, Ciotti F, Sarzana M, Fraschini M, Zambon AA, Acquati S, Redaelli D, Attanasio V, Miglietta S, De Mattia F, Barzaghi F, Ferrua F, Migliavacca M, Tucci F, Gallo V, Del Carro U, Canale S, Spiga I, Lorioli L, Recupero S, Fratini ES, Morena F, Silvani P, Calvi MR, Facchini M, Locatelli S, Corti A, Zancan S, Antonioli G, Farinelli G, Gabaldo M, Garcia-Segovia J, Schwab LC, Downey GF, Filippi M, Cicalese MP, Martino S, Di Serio C, Ciceri F, Bernardo ME, Naldini L, Biffi A, Aiuti A.  Lentiviral haematopoietic stem-cell gene therapy for early-onset metachromatic leukodystrophy: long term results from a non-randomised, open-label, phase ½ trial and expanded access.  Lancet. 2022 Jan 22; 399(10322:372–383. doi: 10.1016/S0140-6736(21)02017-1. PMID: 35065785 [PMC free article: PMC8795071] [PubMed: 35065785]
  64. Gaspar HB, Parsley KL, Howe S, King D, Gilmur KC, Sinclair J, Brouncs G, Schmidt M, Von Halle C, Barington T, Jakobsen MA, Christensen HO, Ghonaium AA, White HN, Smith JL, Levinsky RJ, Ali RR, Kinnon C, Thrasher AJ.  Gene therapy of X-linked severe combined immunodeficiency by use of a pseudotyped gammaretroviral vector.  Lancet. 2004 Dec;364(9452):2181–2187. doi: 10.1016/S0140-6736(40)17590-9 [PubMed: 15610804]
  65. Gazzerro E, Bonetto A, Minetti C.  Caveolinopathies: Translational implications of caveolin-3 in skeletal and cardiac muscle disorders.  Handb Clin Neurol.  2011;101:135–142. doi: 10.1016/B978-0-08-045031-5.00010-4 PMID: 21496630 [PubMed: 21496630]
  66. Gehlert S, Difrancesco A, Chang CH.  Black-white differences in the psychosocial outcomes of epilepsy.  Epilepsy Res. 2000 Nov;42(1):63–73. PubMed: 10996507 doi: 10.1016/s0920-1211(00 )00161-3 [PubMed: 10996507]
  67. Gidaro T, Servais L.  Nusinersen treatment of spinal muscular astrophy: Current knowledge and existing gaps,  Dev Med Child Neurol. 2019 Jan; 61(1):19–24. doi: 10.1111/dmcn.14027 PMID: 30221755 [PubMed: 30221755]
  68. Greenberg DA, Cayanis E, Strug L, Marathe S, Durner M, Pal DK, Alvin GB, Klotz I, Dicker E, Shinnar S, Bromfield EB, Resor S, Cohen J, Moshe SL, Harden C, Kang H.  Malic enzyme 2 may underlie susceptibility to adolescent-onset idiopathic generalized epilepsy.  Am J Hum Genet. 2005 Jan;76(1):139–146. PMID: 15532013 PMCID: PMC1196416 DOI: 10.1086/426735 [PMC free article: PMC1196416] [PubMed: 15532013]
  69. Guerrini R, Marini C.  Genetic malformations of cortical development.  Exp Brain Res. 2006 Aug;173(2):322–333. doi: 10.1007/s00221-006-0501-z PMID: 16724181 [PubMed: 16724181]
  70. Gumusgoz E, Kasiri S, Guisso DR, Wu J, Dear M, Verhalen B, Minassian BA.; AAV-Mediated artificial miRNA reduces pathogenic polyglucosan bodies and neuroinflamation in adult polyglucosan body and Lagoroa diseae mouse models, Neurotherapeutics. 2022 Apr;19(3):982-993. doi: 10.1007/s13311-022-01218-7. PMID: 35347645. [PMC free article: PMC9294094] [PubMed: 35347645]
  71. Hacein-Bey-Abina S, Von Kalle C, Schmidt M, McCormack MP, Wulffraat N, Leboulch P, Lim A, Osborne CS, Pawliuk R, Morillon E, Sorensen R, Forster A, Fraser P, Cohen JI, de Saint Basile G, Alexander I, Wintergerst U, Frebourg T, Aurias A, Stoppa-Lyonnet D, Romana S, Radford-Weiss I, Gross F, Valensi F, Delabesse E, Macintyre E, Sigaux F, Soulier J, Leiva LE, Wissler M, Prinz C, Rabbitts TH, Le Deist F, Fischer A, Cavazzana-Calvo M.  LMO2-associated clonal T cell proliferation in two patients after gene therapy for SCID-X1.  Science. 2003 Oct 17;302(5644):415–419. doi: 10.1126/science.1088547 PMID: 14564000 [PubMed: 14564000]
  72. Hamdan FF, Myers CT, Cossette P, Lemay P, Spiegelman D, Laporte AD et al. Michaud JL.  High rate of recurrent de novo mutations in developmental and epileptic encephalopathies.  Am J Hum Genet.  2017 ; 101: 664–685. PMID: 29100083 PMCID: PMC5673604 DOI: 10.1016/j.ajhg.2017.09.008 [PMC free article: PMC5673604] [PubMed: 29100083]
  73. Han Z, Chen C, Christiansen A, Ji S, Lin Q, Anumonwo C, Liu C, Leiser SC, Meena, Aznarez I, Liau G, Isom LL.  Antisense oligonucleotides increase Scn1a expression and reduce seizures and SUDEP incidence in a mouse model of Dravet syndrome,  Sci Transl Med. 2020 Aug 26;12(558):eaaz6100. doi: 10.1126/scitranslmed.aaz6100 PMID: 32848094 [PubMed: 32848094]
  74. Harkin LA, McMahon JM, Iona X, Dibbens L, Pelekanos JT, Zuberi SM, Sadleir LG, Andermann E, Gill D, Farrell K, Connolly M, Stanley T, Harbord M, Andermann F, Wang J, Batish SD, Jones JG, Seltzer WK, Gardner A; Infantile Epileptic Encephalopathy Referral Consortium, Sutherland G, Berkovic SF, Mulley JC, Scheffer IE.  The spectrum of SCN1A-related infantile epileptic encephalopathies.  Brain. 2007 Mar;130(Pt 3):843–852. doi: 10.1093/brain/awm002 PMID: 17347258 [PubMed: 17347258]
  75. Helbig I, Mefford HC, Sharp AJ, Guipponi M, Fichera M, Francke A, Muhle H, de Kovel C, Baker C, von Spiczak S, Kron KL, Steinich I, Kleefuss-Lie AA, Leu C, Gaus V, Schmitz B, Klein KM, Reif PS, Rosenow F, Weber Y, Lerche H, Zimprich F, Urah L, Fuchs K, Feucht M, Genton P, Thomas P, Visscher F, de Haan G-J, Moller RS, Hjalgrim H, Luciano D, Wittig M, Nothnagel M, Elger CE, Nurnberg P, Romano C, Malafosse A, Koeleman BPC, Lindhout D, Stephani U, Schreiber S, Eichler EE, Sander T.  15q13.3 microdeletions increase risk of idiopathic generalized epilepsy.  Nat Genet. 2009 Feb;41(2):160–162. PMID: 19136953 PMCID: PMC3026630 DOI: 10.1038/ng.292 [PMC free article: PMC3026630] [PubMed: 19136953]
  76. Hill SF, Meisler MH.  Antisense Oligonucleotide therapy for neurodevelopmental disorders,  Developmental Neurosci,  2021 ; 43(3-4) : 247–252. doi: 10.1159/000517686 PMID: 34412058 PMCID: PMC8440367 [PMC free article: PMC8440367] [PubMed: 34412058]
  77. Hirose S, Iwata H, Akiyoshi H, Kobayashi K, Ito M, Wada K, Kaneko S, Mitsudome A.  A novel mutation of CHRNA4 responsible for autosomal dominant nocturnal frontal lobe epilepsy.  Neurology. 1999 Nov 10;53(8):1749–1753. doi: 10.1212/wnl.53.8.1749 PMID: 10563623 [PubMed: 10563623]
  78. Hirose S, Zenri F, Akiyoshi H, Fukuma G, Iwata H, Inoue T, Yonetani M, Tsutsumi M, Muranaka H, Kurokawa T, Hanai T, Wada K, Kaneko S, Mitsudome A.  A novel mutation of KCNQ3 (c.925T-->C) in a Japanese family with benign familial neonatal convulsions.  Ann Neurol. 2000 Jun;47(6):822–826. PMID: 10852552 [PubMed: 10852552]
  79. Iffland H, Carson V, Bordey A, Crino PB.  GATORopathies: The role of amino acid reguatory gene mutations in epilepsy and cortical malformations.  Epilepsia 2019 Nov;60(11):2163–2173. PMID: 31625153 PMCID: PMC7155771 DOI: 10.1111/epi.16370 [PMC free article: PMC7155771] [PubMed: 31625153]
  80. Imbrici P, Jaffe SL, Eunson LH, Davies NP, Herd C, Robertson R, Robertson R, Kullmann DM, Hanna MG.  Dysfunction of the brain calcium channel CaV2.1 in absence epilepsy and episodic ataxia.  Brain. 2004 Dec;127(Pt 12):2682–2692. PMID: 15483044 DOI: 10.1093/brain/awh301 [PubMed: 15483044]
  81. Jacoby A.  Stigma, epilepsy, and quality of life.  Epilepsy Behavior.  2002;3 :10–20. PubMed 12609301 [PubMed: 12609301]
  82. Jacoby A, Gorry J, Gamble C, Baker GA.  Public knowledge, private grief : a study of public attitudes to epilepsy in the United Kingdom and implications for stigma.  Epilepsia.  2004;45:1405–1415. PubMed 15509242 [PubMed: 15509242]
  83. Jensen TL, Gotzsche CR  Woldbye DPD.  Current and Future Prospects for Gene Therapy for Rare Genetic Diseases Affecting the Brain and Spinal Cord,  Front Molecular Neurosci. 2021 Oct 6;14:695937. PMID: 34690692 PMCID: PMC8527017 DOI: 10.3389/fnmol.2021.695937 [PMC free article: PMC8527017] [PubMed: 34690692]
  84. Jiang H, Pierce GF, Ozelo MC, de Paula EV, Vargas JA, Smith P, Sommer J, Luk A, Manno CS, High KA, Arruda VR.  Evidence of Multiyear Factor IX Expression iby AAV-mediated Gene Transfer to Skeletal Muscle in an Individual with Severe Hemophilia B.  Mol Ther. 2006 Sep;14(3):452–455. doi: 10.1016/j.ymthe.2006.05.004 PMID: 16822719 [PubMed: 16822719]
  85. Kalachikov S, Evgrafov O, Ross B, Winawer M, Barker-Cummings C, Boneschi FM, Choi C  Morozov P, Das K, Teplitskaya E, Yu A, Cayanis E, Penchaszadeh G, Kottman AH, Pedley TA, Hauser WA, Ottman R, Gilliam TC.  Mutations in LGI1 cause autosomal-dominant partial epilepsy with auditory features.  Nat Genet. 2002 Mar;30(3):335–341. PMID: 11810107 PMCID: PMC2606053 DOI: 10.1038/ng832 [PMC free article: PMC2606053] [PubMed: 11810107]
  86. Kalscheuer VM, Tao J, Donnelly A, Hollway G, Schwinger E, Kübart S, Menzel C, Hoeltzenbein M, Tommerup N, Eyre H, Harbord M, Haan E, Sutherland GR, Ropers HH, Gécz J.  Disruption of the serine/threonine kinase 9 gene causes severe X-linked infantile spasms and mental retardation.  Am J Hum Genet. 2003 Jun;72(6):1401–1411. doi: 10.1086/375538 PMID: 12736870 [PMC free article: PMC1180301] [PubMed: 12736870]
  87. Kananura C, Haug K, Sander T, Runge U, Gu W, Hallmann K, Rebstock J, Heils A, Steinlein OK.  A splice-site mutation in GABRG2 associated with childhood absence epilepsy and febrile convulsions.  Arch Neurol. 2002 Jul;59(7):1137–1141. PMID: 12117362 DOI: 10.1001/archneur.59.7.1137 [PubMed: 12117362]
  88. Kato M, Saitoh S, Kamei A, Shiraishi H, Ueda Y, Akasaka M, Tohyama J, Akasaka N, Hayasaka K.  A longer polyalanine expansion mutation in the ARX gene causes early infantile epileptic encephalopathy with suppression-burst pattern (Ohtahara syndrome).  Am J Hum Genet. 2007 Aug;81(2):361–366. PMID: 17668384 PMCID: PMC1950814 DOI: 10.1086/518903 [PMC free article: PMC1950814] [PubMed: 17668384]
  89. Katsarou A-M, Galanopoulou AS, Moshe SL.  Epileptogenesis in neonatal brain, Semin Fetal Neonatal Med. 2018 Jun ; 23(3) :159–167. PMID: 29467100 PMCID: PMC6015538 DOI: 10.1016/j.siny.2017.12.004 [PMC free article: PMC6015538] [PubMed: 29467100]
  90. Kim J, Hu C, Moufawad El Achkar C, Black LE, Douville J, Larson A, Pendergast MK, Goldkind SF, Lee EA, Kuniholm A, Soucy A, Vaze J, Belur NR, Fredriksen K, Stojkovska I, Tsytsykova A, Armant M, DiDonato RL, Choi J, Cornelissen L, Pereira LM, Augustine EF, Genetti CA, Dies K, Barton B, Williams L, Goodlett BD, Riley BL, Pasternak A, Berry ER, Pflock KA, Chu S, Reed C, Tyndall K, Agrawal PB, Beggs AH, Grant PE, Urion DK, Snyder RO, Waisbren SE, Poduri A, Park PJ, Patterson A, Biffi A, Mazzulli JR, Bodamer O, Berde CB, Yu TW.  Patient-customized oligonucleotide therapy for a rare genetic disease.  N Engl J Med 2019 Oct 24; 381:1644–1652. Doi: 10.1056/NEJMoal813279 PMID: 31597037 PMCID : PMC6961983 [PMC free article: PMC6961983] [PubMed: 31597037]
  91. Kitamura K, Yanazawa M, Sugiyama N, Miura H, Iizuka-Kogo A, Kusaka M, Omichi K, Suzuki R, Kato-Fukui Y, Kamiirisa K, Matsuo M, Kamijo S-I, Kasahara M, Yoshioka H, Ogata T, Fukuda T, Kondo I, Kato M, Dobyns WB, Yokoyama M, Morohashi K-i.  Mutation of ARX causes abnormal development of forebrain and testes in mice and X-linked lissencephaly with abnormal genitalia in humans.  Nat Genet. 2002 Nov;32(3):359–369. PMID: 12379852 DOI: 10.1038/ng1009 [PubMed: 12379852]
  92. Kleine Holthaus SM, Aristorena M, Maswood R, Semenyuk O, Hoke J, Hare A, Smith AJ, Mole SE, Ali RR.  Gene therapy targeting the inner retina rescues the retinal phenotype in a mouse model of CLN3 Batten Disease.  Hum Gene Ther. 2020 Jul;31(13-14):709–718. doi: 10.1089/hum.2020.038. PMID: 32578444 [PMC free article: PMC7404834] [PubMed: 32578444]
  93. Ko A, Youn SE, Kim SH, Lee JS, Kim S, Choi JR, Kim HD, Lee ST, Kang HC.  Targeted gene panel and genotype-phenotype correlation in children with developmental and epileptic encephalopathy.  Epilepsy Res. 2018 Mar; 141:48–55. PMID: 29455050 doi: 10.1016/j.epilepsyres.2018.02.003. [PubMed: 29455050]
  94. Kodera H, Nakamura K, Osaka H, Maegaki Y, Haginoya K, Mizumoto S, Kato M, Okamoto N, Iai M, Kondo Y, Nishiyama K, Tsurusaki Y, Nakashima M, Miyake N, Hayasaka K, Sugahara K, Yuasa I, Wada Y, Matsumoto N, Saitsu H.  De novo mutations in SLC35A2 encoding a UDP-galactose transporer cause early-onset epileptic encephalopathy.  Hum Mutat. 2013 Dec;34(12):1708–1714. doi: 10.1002/humu.22446 PMID: 24115232 [PubMed: 24115232]
  95. Kotin RM, Menninger JC, Ward DC, Berns KI.  Mapping and direct visualization of a region specific viral DNA integration site on chromosome 19q13-qter.  Genomics. 1991 Jul; 10(3):831–834. PMID: 1653762 DOI: 10.1016/0888-7543(91)90470-y [PubMed: 1653762]
  96. Kotin RM, Siniscalco M, Samulski RJ, Zhu XD, Hunter L, Laughlin CA, McLaughlin S, Muzyczka N, Rocchi M, Berns KI.  Site-specific integration by adeno-associated virus.  Proc Natl Acad Sci USA. 1990 Mar;87(6):2211–2215. PMID: 2156265 PMCID: PMC53656 DOI: 10.1073/pnas.87.6.2211 [PMC free article: PMC53656] [PubMed: 2156265]
  97. Krauss GL, Gondek S, Krumholz A, Paul S, Shen F.  “The scarlet E”: the presentation of epilepsy in the English language print media.  Neurology.  2000;54:1894–1898. PubMed 10822424 [PubMed: 10822424]
  98. Lafora GR & Glueck B. Beitrag zur Histopathologie der myoklo-nischen Epilepsie.  Zeitschrift fur die gesamte Neurologie and Psychiatrie  1911; 6(Suppl 1): 1–14.
  99. Leen WG, Klepper J, Verbeek MM, Leferink M, Hofste T, van Engelen BG, Wevers RA, Arthur T, Bahi-Buisson N, Ballhausen D, Bekhof J, van Bogaert P, Carrilho I, Chabrol B, Champion MP, Coldwell J, Clayton P, Donner E, Evangeliou A, Ebinger F, Farrell K, Forsyth RJ, de Goede CG, Gross S, Grunewald S, Holthausen H, Jayawant S, Lachlan K, Laugel V, Leppig K, Lim MJ, Mancini G, Marina AD, Martorell L, McMenamin J, Meuwissen ME, Mundy H, Nilsson NO, Panzer A, Poll-The BT, Rauscher C, Rouselle CM, Sandvig I, Scheffner T, Sheridan E, Simpson N, Sykora P, Tomlinson R, Trounce J, Webb D, Weschke B, Scheffer H, Willemsen MA.  Glucose transporter-1 deficiency syndrome: the expanding clinical and genetic spectrum of a treatable disorder.  Brain. 2010 Mar;133(Pt 3): 655–670. doi: 10.1093/brain/awp336 PMID: 20129935 [PubMed: 20129935]
  100. Lehrman S.  Virus treatment questioned after gene therapy death.  Nature. 1999 Oct;401(6753):517–518. doi: 10.1038/43977 PMID: 10524611 [PubMed: 10524611]
  101. Leppert M, Anderson VE, Quattlebaum T  Stauffer D, O’Connell P, Nakamura Y, Lalouel JM, White R.  Benign familial neonatal convulsions linked to genetic markers on chromosome 20,  Nature. 1989 Feb 16;337 (6208):647–648. doi: 10.1038/337647a0 PMID: 2918897 [PubMed: 2918897]
  102. Lesca G, Rudolf G, Bruneau N, Lozovaya N, Labalme A, Boutry-Kryza N, Salmi M, Tsintsadze T, Addis L, Motte J, Wright S, Tsintsadze V, Michel A, Doummar D, Lascelles K, Strug L, Waters P, de Bellescize J, Vrielynck P, de Saint Martin A, Ville D, Ryvlin P, Arzimanoglou A, Hirsch E, Vincent A, Pal D, Burnashev N, Sanlaville D, Szepetowski P.  GRIN2A mutations in acquired epileptic aphasia and related childhood focal epilepsies and encephalopathies with speech and language dysfunction.  Nat Genet. 2013 Sep;45(9):1061–1066. doi: 10.1038/ng.2726. PMID: 23933820 [PubMed: 23933820]
  103. Li H, Fang F, Xu M, Liu Z, Zhou J, Wang X, Wang X, Han T.  Clinical Assessments and EEG Analyses of Encephalopathies Associated With Dynamin-1 Mutation.  Front Pharmacol. 2019 Dec 4;10:1454. PMID: 31920647 PMCID: PMC6915043 DOI: 10.3389/fphar.2019.01454 [PMC free article: PMC6915043] [PubMed: 31920647]
  104. Lorenz S, Taylor KP, Gehrmann A, Becker T, Muhle H, Gresch M, Tauer U, Sander T, Stephani U.  Association of BRD2 polymorphisms with photoparoxysmal response.  Neurosci Lett. 2006 May 29;400(1-2):135–139. doi: 10.1016/j.neulet.2006.02.026 PMID: 16516380 [PubMed: 16516380]
  105. Lowes LP, Alfano LN, Arnold WD, Shell R, Prior TW, McColly M, Lehman KJ, Church K, Sproule DM, Nagendran S, Menier M, Feltner DE, Wells C, Kissel JT, Al-Zaidy S, Mendell J.  Impact of age and motor function in a phase 1/2A study of infants with SMA type 1 receiving single-dose gene replacement therapy.  Pediatr Neurol. 2019 Sep; 98:39–45. PMID: 31277975 DOI: 10.1016/j.pediatrneurol.2019.05.005 [PubMed: 31277975]
  106. Lubroth P, Colasante G, Lignani G.  In vivo genome editing therapeutic approaches for neurological disorders: where are we in the translational pipeline? Front. Neurosci. 2021 Feb 18;15:632522. PMID: 33679313 PMCID: PMC7930815 DOI: 10.3389/fnins.2021.63252 [PMC free article: PMC7930815] [PubMed: 33679313]
  107. Marini C, Mei D, Parmeggiani L, Norci V, Calado E, Ferrari A, Moreira A, Pisano T, Specchio N, Vigevano F, Battaglia D, Guerrini R.  Protocardherin 19 mutatons in girls with infantile-onset epilepsy.  Neurology. 2010 Aug 17;75(7):646–653. doi: 10.1212/WNL.0b013e3181ed9e67 PMID: 20713952 [PubMed: 20713952]
  108. Marini C, Mei D, Temudo T, Ferrari AR, Buti D, Dravet C, Dias AI, Moreira A, Calado E, Seri S, Neville B, Narbona J, Reid E, Michelucci R, Sicca F, Cross HJ, Guerrini R.  Idiopathic epilepsies with seizures precipitated by fever and SCN1A abnormalities.  Epilepsia. 2007 Sep;48(9):1678–1685. PMID: 17561957 DOI: 10.1111/j.1528-1167.2007.01122.x [PubMed: 17561957]
  109. Marini C, Scheffer IE, Nabbout R, Mei D, Cox K, Dibbens LM, McMahon JM, Iona X, Carpintero RS, Elia M, Cilio MR, Specchio N, Giordano L, Striano P, Gennaro E, Cross JH, Kivity S, Neufeld MY, Afawi Z, Andermann E, Keene D, Dulac O, Zara F, Berkovic SF, Guerrini R, Mulley JC.  SCN1A duplications and deletions detected in Dravet syndrome: implications for molecular diagnosis.  Epilepsia. 2009 Jul;50(7):1670–1678. doi: 10.1111/j.1528-1167.2009.02013.x. PMID: 19400878 [PubMed: 19400878]
  110. Marshall CR, Young EJ, Pani AM, Freckmann ML, Lacassie Y, Howald C, Fitzgerald KK, Peippo M, Morris CA, Shane K, Priolo M, Morimoto M, Kondo I, Manguoglu E, Berker-Karauzum S, Edery P, Hobart HH, Mervis CB, Zuffardi O, Reymond A, Kaplan P, Tassabehji M, Gregg RG, Scherer SW, Osborne LR.  Infantile spasms is associated with deletion of the MAG12 gene on chromosome 7q11.23-q21.11.  Am J Hum Genet. 2008 Jul;83(1):106–111. doi: 10.1016/j.ajhg.2008.06.001. PMID: 18565486 [PMC free article: PMC2443840] [PubMed: 18565486]
  111. Masnada S, Hedrich UBS, Gardella E, Schubert J, Kaiwar C, Klee EW, Lanpher BC, Gavrilova RH, Synofzik M, Bast T, Gorman K, King MD, Allen NM, Conroy J, Ben Zeev B, Tzadok M, Korff C, Dubois F, Ramsey K, Narayanan V, Serratosa JM, Giraldez BG, Helbig I, Marsh E, O’Brien M, Bergqvist CA, Binelli A, Porter B, Zaeyen E, Horovitz DD, Wolff M, Marjanovic D, Caglayan HS, Arslan M, Pena SDJ, Sisodiya SM, Balestrini S, Syrbe S, Veggiotti P, Lemke JR, Møller RS, Lerche H, Rubboli G.  Clinical spectrum and genotype-phenotype associations of KCNA2-related encephalopathies.  Brain. 2017 Sep 1;140(9):2337–2354. doi: 10.1093/brain/awx184 PMID: 29050392 [PubMed: 29050392]
  112. McPhee SW, Janson CG, Li C, Samulski RJ, Camp AS, Francis J, Shera D, Lioutermann L, Feely M, Freese A, Leone P.  Immune responses to AAV in a phase I study for Canavan disease.  J Gene Med. 2006 May;8(5):577–588. doi: 10.1002/jgm.885 PMID: 16532510 [PubMed: 16532510]
  113. Mei D, Marini C, Novara F, Bernardina BD, Granata T, Fontana E, Parrini E, Ferrari AR, Murgia A, Zuffardi O, Guerrini R.  Xp22.3 genomic deletions involving the CDKL5 gene in girls with early onset epileptic encephalopathy.  Epilepsia. 2010 Apr;51(4):647–654. PMID: 19780792 DOI: 10.1111/j.1528-1167.2009.02308.x [PubMed: 19780792]
  114. Mendell JR, Al-Zaidy S, Shell R, Arnold WD, Rodino-Klapac LR, Prior TW  Lowes L, Alfano L, Berry K, Church K, Kissel JT, Nagendran S, L’Italien J, Sproule DM, Wells C, Cardenas JA, Heitzer MD, Kaspar A, Corcoran S, Braun L, Likhite S, Miranda C, Meyer K, Foust KD, Burghes AHM, Kaspar BK.  Single-dose gene-replacement therapy for spinal muscular atrophy.  N Engl J Med. 2017 Nov 2;377(18):1713–1722. doi: 10.1056/NEJMoa1706198 PMID: 29091557 [PubMed: 29091557]
  115. Mendell JR, Al-Zaidy SA, Rodino-Klapac LR, Goodspeed K, Gray SJ, Kay CN, Boye SL, Boye SE, George LA, Salabarria S, Corti M, Byrne BJ, Tremblay JP.  Current clinical applications of in vivo gene therapy with AAVs.  Mol Ther. 2021 Feb 3;29(2):464–488. doi: 10.1016/j.ymthe.2020.12.007 PMID: 33309881 [PMC free article: PMC7854298] [PubMed: 33309881]
  116. Meng L, Ward AJ, Chun S, Bennett CF, Beaudet AL, Rigo F.  Towards a therapy for Angelman syndrome by targeting a long non-coding RNA.  Nature. 2015 Feb 19;518(7539):409–412. PMID: 25470045 [PMC free article: PMC4351819] [PubMed: 25470045]
  117. Mercuri E, Darras BT, Chiriboga CA  Day JW, Campbell C, Connolly AM, Iannaccone ST, Kirschner J, Kuntz NL, Saito K, Shieh PB, Tulinius M, Mazzone ES, Montes J, Bishop KM, Yang Q, Foster R, Gheuens S, Bennett CF, Farwell W, Schneider E, De Vivo DC, Finkel RS; CHERISH Study Group. Nusinersen versus sham control in later-onset spinal muscular atrophy.  N Engl J Med. 2018 Feb15;378(7):625–635. doi: 10.1056/NEJMoa1710504 PMID: 29443664 [PubMed: 29443664]
  118. Merkel O, Kenner L, Turner SD.  Stem cell hunt in NLH, Editorial.  Oncoscience.  2015;2(10):809–810. [PMC free article: PMC4671931] [PubMed: 26682256]
  119. Merkle FT, Neuhausser WM, Santos D, Valen E, Gagnon JA, Maas K, Sandoe J, Schier AF, Eggan K.  Efficient CRISPR-Cas9-mediated generation of knockin human pluripotent stem cells lacking undesired mutations at the targeted locus.  Cell Rep.  2015 May 12;11:875–883. doi: 10.1016/j.celrep.2015.04.007 PMID: 25937281 PMCID: PMC5533178 [PMC free article: PMC5533178] [PubMed: 25937281]
  120. Michelucci R, Poza JJ, Sofia V, de Feo MR, Binelli S, Bisulli F, Scudellaro E, Simionati B, Zimbello R, D’Orsi G, Passarelli D, Avoni P, Avanzini G, Tinuper P, Biondi R, Valle G, Mautner VF, Stephani U, Tassinari CA, Moschonas NK, Siebert R, de Munain AL, Perez-Tur J, Nobile C.  Autosomal dominant lateral lobe epilepsy: Clinical spectrum, new epitempin mutations, and genetic heterogeneity in seven European families.  Epilepsia. 2003 Oct;44(10):1289–1297. PMID: 14510822 DOI: 10.1046/j.1528-1157.2003.20003.x [PubMed: 14510822]
  121. Mills PB, Struys E, Jakobs C, Plecko B, Baxter P, Baumgartner M, Willemsen MA, Omran H, Tacke U, Uhlenberg B, Weschke B, Clayton PT.  Mutations in antiquitin in individuals with pyridoxine-dependent seizures.  Nat Med. 2006 Mar;12(3):307–309. doi: 10.1038/nm1366 PMID: 16491085 [PubMed: 16491085]
  122. Minassian BA, Lee JR, Herbrick JA, Huizenga J, Soder S, Mungall AJ, Dunham I, Gardner R, Fong CY, Carpenter S, Jardim L, Satishchandra P, Andermann E, Snead 3rd OC, Lopez-Cendes I, Tsui LC, Delgado-Escueta AV, Rouleau GA, Scherer SW.  Mutations in a gene encoding a novel protein tyrosine phosphatase cause progressive myoclonus epilepsy.  Nat Genet. 1998 Oct;20(2):171–174. PMID: 9771710 DOI: 10.1038/2470 [PubMed: 9771710]
  123. Nabbout R, Gennaro E, Dalla Bernardina B, Dulac O, Madia F, Bertini E, Capovilla G, Chiron C, Cristofori G, Elia M, Fontana E, Gaggero R, Granata T, Guerrini R, Loi M, La Selva L, Lispi ML, Matricardi A, Romeo A, Tzolas V, Valseriati D, Veggiotti P, Vigevano F, Vallée L, Dagna Bricarelli F, Bianchi A, Zara F.  Spectrum of SCN1A mutations in severe myoclonic epilepsy of infancy.  Neurology. 2003 Jun 24;60(12):1961–1967. doi: 10.1212/01.wnl.0000069463.41870.2f. PMID: 12821740 [PubMed: 12821740]
  124. Nakagama Y, Isojima T, Mizuno Y, Takahashi N, Kitanaka S, Igarashi T.  Growth hormone deficiency: a possible complication of glucose transporter 1 deficiency?  Acta Paediatr. 2012 Jun;101(6):e259–262. PMID: 22251140 DOI: 10.1111/j.1651-2227.2012.02606.x [PubMed: 22251140]
  125. Nganou G, Silva CG, Gladwyn-Ng I, Engel D, Coumans B, Delgado-Escueta AV, Tanaka M, Nguyen L, Grisar T, de Nijs L, Lakaye B.  Importin-8 modulates division of apical progenitors, dendritogenesis and tangential migration during development of mouse cortex.  Front Mol Neurosci. 2018 Jul 10; 11:234. PMID: 30042658 PMCID: PMC6048241 DOI: 10.3389/fnmol.2018.00234 [PMC free article: PMC6048241] [PubMed: 30042658]
  126. Ngugi AK, Bottomley C, Kleinschmidt I, Sander JW, Newton CR.  Estimation of the burden of active and life-time epilepsy : a meta-analytic approach.  Epilepsia. 2010 May;51(5) : 883–890. doi: 10.1111/j.1528-1167.2009.02481.x PMID: 20067507 [PMC free article: PMC3410521] [PubMed: 20067507]
  127. Nguyen GN, Everett JK, Kafle S, Roche AM, Raymond HE, Leiby J, Wood C, Assenmacher CA, Merricks EP, Long CT, Kazazian HH, Nichols TC, Bushman FD, Sabatino DE.  A long-term study of AAV gene therapy in dogs with hemophilia A identifies clonal expansions of transduced liver cells.  Nat Biotechnol. 2021 Jan; 39(1):47–55. doi: 10.1038/s41587-020-0741-7 PMID: 33199875 [PMC free article: PMC7855056] [PubMed: 33199875]
  128. Niturad CE, Lev D, Kalscheuer VM, Charzewska A, Schubert J, Lerman-Sagie T, Kroes HY, Oegema R, Traverso M, Specchio N, Lassota M, Chelly J, Bennett-Back O, Carmi N, Koffler-Brill T, Iacomino M, Trivisano M, Capovilla G, Striano P, Nawara M, Rzonca S, Fischer U, Bienek M, Jensen C, Hu H, Thiele H, Altmüller J, Krause R, May P, Becker F; EuroEPINOMICS Consortium, Balling R, Biskup S, Haas SA, Nürnberg P, van Gassen KLI, Lerche H, Zara F, Maljevic S, Leshinsky-Silver E.  Rare GABRA3 variatns are associated with epileptic seizures, encephalopathy and dysmorphic features.  Brain. 2017 Nov 1; 140(11):2879–2894. doi: 10.1093/brain/awx236 PMID: 29053855 [PubMed: 29053855]
  129. Oguni H, Hayashi K, Awaya Y, Fukuyama Y, Osawa M.  Severe myoclonic epilepsy in infants - A review based on the Tokyo Women’s Medical University series of 84 cases.  Brain Dev. 2001 Nov;23(7):736–748. doi: 10.1016/s0387-7604(01)00276-5. PMID: 11701288 [PubMed: 11701288]
  130. Oguni H, Hayashi K, Osawa M, Awaya Y, Fukuyama Y, Fukuma G, Hirose S, Mitsudome A, Kaneko S.  Severe myoclonic epilepsy in infancy: clinical analysis and relation to SCN1A mutations in a Japanese cohort.  Adv Neurol.  2005;95:103–117. PMID: 15508916 [PubMed: 15508916]
  131. Ottman R, Hirose S, Jain S, Lerche H, Lopes-Cendes I, Noebels JL, Serratosa J, Zara F, Scheffer I.  Genetic testing in the epilepsies - Report of the ILAE genetics commission.  Epilepsia. 2010 Apr;51(4):655–670. PMID: 20100225 PMCID: PMC2855784 DOI: 10.1111/j.1528-1167.2009.02429.x [PMC free article: PMC2855784] [PubMed: 20100225]
  132. Ou L, Przybilla MJ, Ahlat O, Kim S, Overn P, Jarnes J, O’Sullivan MG, Whitley CB.  A highly efficacious PS gene editing system corrects metabolic and neurological complications of mucopolysaccharidosis type I.  Mol Ther. 2020 Jun 3;28(6):1442–1454. doi: 10.1016/j.ymthe.2020.03.018 PMID: 32278382 PMCID: PMC7264433 [PMC free article: PMC7264433] [PubMed: 32278382]
  133. Pal DK, Evgrafov OV, Tabares P, Zhang F, Durner M, Greenberg DA.  BRD2 (RING3) is a probable major susceptibility gene for common juvenile myoclonic epilepsy.  Am J Hum Genet. 2003 Aug;73(2):261–270. PMID: 12830434 PMCID: PMC1180366 DOI: 10.1086/377006 [PMC free article: PMC1180366] [PubMed: 12830434]
  134. Pechmann A, Langer T, Schorling D, Stein S, Vogt S, Schara U, Kölbel H, Schwartz O, Hahn A, Giese K, Johannsen J, Denecke J, Weiß C, Theophil M, Kirschner J.  Evaluation of children with SMA type 1 under treatment with Nusinersen within the expanded access program in Germany.  J Neuromuscul Dis.  2018;5(2):135–143. doi: 10.3233/JND-180315 PMID: 29689734 [PMC free article: PMC6004898] [PubMed: 29689734]
  135. Pennachio LA, Lehesjoki AE, Stone NE, Willour VL, Virtaneva K, Miao J, D’Amato E, Ramirez L, Faham M, Koskiniemi M, Warrington JA, Norior R, de la Chapelle A, Cox DR, Myers RM. (1996): Mutations in the gene encoding cystatin B in progressive myoclonus epilepsy (EPM1).  Science. 1996 Mar 22;271(5256): 1731–1734. PMID: 8596935 DOI: 10.1126/science.271.5256.1731 [PubMed: 8596935]
  136. Poletto E, Baldo G, Gomez-Ospina N.  Genome editing for mucopolysaccharidoses.  Int J Mol Sci. 2020 Jan 13;21(2):500. doi: 10.3390/ijms21020500 PMID: 31941077 [PMC free article: PMC7014411] [PubMed: 31941077]
  137. Rao SN, Maity R, Sharma J, Dey P, Shankar SK, Satischandra P, Jana NR.  Sequestration of chaperones and proteasome into Lafora bodies and proteasomal dysfunction induced by Lafora disease-associated mutations of malin.  Hum Mol Genet. 2010 Dec 1;19(23):4726–4734. doi: 10.1093/hmg/ddq407 PMID: 20858601 [PubMed: 20858601]
  138. Raper SE, Chirmule N, Lee FS, Wivel NA, Bagg A, Gao GP, Wilson JM, Batshaw ML.  Fatal systemic inflammatory syndrome in a ornithine transcarbarmylase deficient patient following adenoviral gene transfer.  Mol. Genet. Metab. 2003 Sep-Oct;80(1-2):148–158. doi: 10.1016/j.ymgme.2003.08.016 PMID: 14567964 [PubMed: 14567964]
  139. Rodrigues-Pinguet N, Jia L, Li M, Fiji A, Klaassen A, Troung A, Lester HA, Cohen BN.  Five ADNFLE mutations reduce the Ca2+ dependence of the mammalian alpha4beta2 acetylcholine response.  J Physiol (Lond). 2003 Jul 1;550(Pt 1):11–26. PMID: 12754307 PMCID: PMC2343021 DOI: 10.1113/jphysiol.2003.036681 [PMC free article: PMC2343021] [PubMed: 12754307]
  140. Roger J, Dravet C, Bureau M, Dreifuss FE, Wolf P. Epileptic Syndromes in Infancy, Childhood and Adolescence, 1st ed.  1985; London: John Libbey.
  141. Roll P, Rudolf G, Pereira S, Royer B, Scheffer IE, Massacrier A, Valenti MP, Roeckel-Trevisiol N, Jamali S, Beclin C, Seegmuller C, Metz-Lutz MN, Lemainque A, Delepine M, Caloustian C, de Saint Martin A, Bruneau N, Depétris D, Mattéi MG, Flori E, Robaglia-Schlupp A, Lévy N, Neubauer BA, Ravid R, Marescaux C, Berkovic SF, Hirsch E, Lathrop M, Cau P, Szepetowski P.  SRPX2 mutations in disorders of language cortex and cognition.  Hum Mol Genet. 2006 Apr 1;15(7):1195–1207. doi: 10.1093/hmg/ddl035. PMID: 16497722 [PubMed: 16497722]
  142. Saitsu H, Kato M, Mizuguchi T, Hamada K, Osaka H, Tohyama J, Uruno K, Kumada S, Nishiyama K, Nishimura A, Okada I, Yoshimura Y, Hirai S, Kumada T, Hayasaka K, Fukuda A, Ogata K, Matsumoto N.  De novo mutations in the gene encoding STXBP1 (MUNC18-1) cause early infantile epileptic encephalopathy.  Nat Genet. 2008 Jun;40(6):782–788. doi: 10.1038/ng.150 PMID: 18469812 [PubMed: 18469812]
  143. Salganik M, Hirsch ML, and Samulski RJ.  Adeno-associated virus as a mammalian DNA vector. Microbiol. Spectr.  2015; 3. doi: 10.1128/microbiolspec.MDNA3-0052-2014 [PMC free article: PMC4677393] [PubMed: 26350320]
  144. Sapio MR, Vessaz M, Thomas P, Genton P, Fricker LD, Salzmann A.  Novel carboxypeptidase A6 (CPA6) mutations identified in patients with juvenile myoclonic and generalized epilepsy.  PloS One. 2015 Apr 13;10(4):e0123180 PMID: 25875328 PMCID: PMC4395397 DOI: 10.1371/journal.pone.0123180 [PMC free article: PMC4395397] [PubMed: 25875328]
  145. Schaefers J, van der Giessen LJ, Klees C, Jacobs EH, Sieverdink S, Dremmen MHG, Spoor JKH, van der Ploeg AT, van den Hout JMP, Huidekoper HH.  Presymptomatic treatment of classic late-infantile neuronal ceroid lipofuscinosis with cerliponase alfa.  Orphanet J Rare Dis.  2021;16:221. doi : 10.1186/s13023-021-01858-6 PMCID: PMC8120778 PMID : 33990214 DOI : 10.1186/s13023-021-01858-6 [PMC free article: PMC8120778] [PubMed: 33990214]
  146. Scheffer IE, Berkovic SF.  Generalized epilepsy with febrile seizures plus. A genetic disorder with heterogeneous clinical phenotypes.  Brain. 1997 Mar;120(Pt 3):479–490. doi: 10.1093/brain/120.3.479 PMID: 9126059 [PubMed: 9126059]
  147. Schinzel A, Niedrist D.  Chromosome imbalances associated with epilepsy.  Am J Med Genet.  2001 Summer;106 (2):119–124. doi: 10.1002/ajmg.1576 PMID: 11579431 [PubMed: 11579431]
  148. Schulz A, Ajayi T, Specchio N, de Los Reyes E, Gissen P, Ballon D, Dyke JP, Cahan H, Slasor P, Jacoby D, Kohlschutter A, CLN2 Study Group. Study of intraventricular ceroid alfa for CLN2 disease.  N Engl J Med. 2018 May 17;378(20):1898–1907. PMID: 29688815 Doi: 10.1056/NEJMoal1712649 [PubMed: 29688815]
  149. Sehara Y, Fujimoto K, Ikeguchi K, Katakai Y, Ono F, Takino N, et al. Persistent expression of dopamine-synthesizing enzymes 15 years after gene transfer in a primate model of Parkinson’s disease.  Hum Gene Ther Clin Dev.  2017;28,74–79. doi: 10.1089/humc.2017.010 PMID: 28279081 [PubMed: 28279081]
  150. Serratosa JM, Gómez-Garre P, Gallardo ME, Anta B, de Bernabé DB, Lindhout D, Augustijn PB, Tassinari CA, Malafosse RM, Topcu M, Grid D, Dravet C, Berkovic SF, de Córdoba SR.  A novel protein tyrosine phosphatase gene is mutated in progressive myoclonus epilepsy of the Lafora type (EPM2).  Hum Mol Genet. 1999 Feb;8(2):345–352. doi: 10.1093/hmg/8.2.345 PMID: 9931343 [PubMed: 9931343]
  151. Sessa M, Lorioli L, Fumagalli F, Acquati S, Redaelli D, Baldoli C, Canale S, Lopez ID, Morena F, Calabria A, Fiori R, Silvani P, Rancoita PM, Gabaldo M, Benedicenti F, Antonioli G, Assanelli A, Cicalese MP, Del Carro U, Sora MG, Martino S, Quattrini A, Montini E, Di Serio C, Ciceri F, Roncarolo MG, Aiuti A, Naldini L, Biffi A.  Lentiviral haemopoietic stem-cell gene therapy in early-onset metachromatic leukodystrophy: an ad-hoc analysis of a non-randomised, open-label, phase 1/2 trial.  Lancet. 2016 Jul 30;388(10043):476–487. doi: 10.1016/S0140-6736(16)30374-9 PMID: 27289174 [PubMed: 27289174]
  152. Shibata A, Kasai M, Terashima H, Hoshino A, Miyagawa T, Kikuchi K, Ishii A, Matsumoto H, Kubota M, Hirose S, Oka A, Mizuguchi M.  Case-control association study of rare nonsynonymous variants of SCN1A and KCNQ2 in acute encephalopathy with biphasic seizures and late reduced diffusion.  J Neurol Sci. 2020 Jul 15;414:116808. doi: 10.1016/j.jns.2020.116808 PMID: 32276107 [PubMed: 32276107]
  153. Shieh PB, Bonnemann CG, Muller-Felber W, Blaschek A, Dowling JJ, Kuntz NL, Seferian AM.  Re : ‘Moving forward after two deaths in a gene therapy trial of myotubular myopathy’ by Wilson and Flotte, Letter to the Editor,  Hum Gene Ther.  2020;31(15 and 16): 787. PMID: 32777938 PMCID: PMC7462017 DOI: 10.1089/hum.2020.217 [PMC free article: PMC7462017] [PubMed: 32777938]
  154. Shim G, Kim D, Park GT, Jin H, Suh S-K, Oh Y-K. Therapeutic gene editing: delivery and regulatory perspectives.  Acta Pharmacol Sin. 2017 Jun;38(6):738–753. doi: 10.1038/aps.2017.2 PMID: 28392568 PMCID: PMC5520188 [PMC free article: PMC5520188] [PubMed: 28392568]
  155. Shostak S, Ottman R.  Ethical, legal, and social dimensions of epilepsy genetics,  Epilepsia. 2006 Oct;47(10) : 1595–1602. doi: 10.1111/j.1528-1167.2006.00632.x PMID: 17054679 [PMC free article: PMC1802101] [PubMed: 17054679]
  156. Silva-Santos S, van Woerden GM, Bruinsma CF, Mientjes E, Jolfaei MA, Distel B, Kushner SA, Elgersma Y.  Ube3a reinstatement identifies distinct developmental windows in a murine Angelman syndrome model.  J Clin Invest. 2015 May;125(5):2069–2076. PMID: 25866966 PMCID: PMC4463212 DOI: 10.1172/JCI80554 [PMC free article: PMC4463212] [PubMed: 25866966]
  157. Singh NA, Charlier C, Stauffer D, DuPont BR, Leach RJ, Melis R, Ronen GM, Bjerre I, Quattlebaum T, Murphy JV, McHarg ML, Gagnon D, Rosales TO, Peiffer A, Anderson VE, Leppert M.  A novel potassium channel gene, KCNQ2, is mutated in an inherited epilepsy of newborns.  Nat Genet. 1998 Jan;18(1):25–29. PMID: 9425895 DOI: 10.1038/ng0198-25 [PubMed: 9425895]
  158. Singh NA, Pappas C, Dahle EJ, Claes LR, Pruess TH, De Jonghe P, Thompson J, Dixon M, Gurnett C, Peiffer A, White HS, Filloux F, Leppert MF.  A role of SCN9A in human epilepsies, as a cause of febrile seizures and as a potential modifier of Dravet syndrome.  PLoS Genet. 2009 Sep;5(9):e10000649. doi: 10.1371/journal.pgen.1000649 PMID: 19763161 [PMC free article: PMC2730533] [PubMed: 19763161]
  159. Snowball A, Chabrol E, Wykes RC, Shekh-Ahmad T, Cornford JH, Lieb A, Hughes MP, Massaro G, Rahim AA, Hashemi KS, Kullman DM, Walker MC, Schorge S.  Epilepsy gene therapy using an engineered potassium channel.  J Neurosci. 2019 Apr 17;39(16),3159–3169. PMID: 30755487 PMCID: PMC6468110 doi: 10.1523/JNEUROSCI.1143-18.2019 [PMC free article: PMC6468110] [PubMed: 30755487]
  160. Sondhi D, Hackett NR, Peterson DA, Stratton J, Baad M, Travis KM, Wilson JM, Crystal RG.  Enhanced survival of the LINCL mouse following CLN2 gene transfer using the rh.10 rhesus macaque-derived adeno-associated virus vector.  Mol Ther. 2007 Mar;15(3):481–491. doi: 10.1038/sj.mt.6300049 PMID: 17180118 [PubMed: 17180118]
  161. Sonzogni M, Zhai P, Mientjes EJ, van Woerden GM, Elgersma Y.  Assessing the requirements of prenatal UBE3A expression for rescue of behavioral phenotypes in a mouse model for Angelman syndrome.  Mol Autism. 2020 Sep 18;11(1):70. PMID: 32948244 PMCID: PMC7501605 DOI: 10.1186/s13229-020-00376-9 [PMC free article: PMC7501605] [PubMed: 32948244]
  162. Sowada N, Hashem MO, Yilmaz R  Hamad M, Kakar N, Thiele H, Arold ST, Bode H, Alkuraya FS, Borck G.  Mutations of PTPN23 in developmental and epileptic encephalopathy,  Hum Genet. 2017 Oct 31 ; 136 : 1455–1461. doi : 10.1007/s00439-017-1850-3 PMID : 29090338 [PubMed: 29090338]
  163. Specchio N, Di Micco V, Trivisano M, Ferretti A, Curatolo P. The epilepsy-autism spectrum disorder phenotype in the era of molecular genetics and precision therapy. Epilepsia. 2022 Jan;63(1):6–21. PMID: 34741464 DOI: 10.1111/epi.17115 [PubMed: 34741464]
  164. Steinlein O, Mulley JC, Propping P, Wallace RH, Phillips HA, Sutherland GR, Scheffer IE, Berkovic SF.  A missense mutation in the neuronal nicotinic acetylcholine receptor alpha 4 subunit is associated with autosomal dominant nocturnal frontal lobe epilepsy.  Nat Genet. 1995 Oct;11(2):201–203. PMID: 7550350 DOI: 10.1038/ng1095-201 [PubMed: 7550350]
  165. Steinlein OK, Mgnusson A, Stoodt J, Bertrand S, Weiland S, Berkovic SF, Nakken KO, Propping P, Bertrand D.  An insertion mutation of the CHRNA4 gene in a family with autosomal dominant nocturnal frontal lobe epilepsy.  Hum Mol Genet. 1997 Jun;6(6):943–947. doi: 10.1093/hmg/6.6.943 PMID: 9175743 [PubMed: 9175743]
  166. Stromme P, Mangelsdorf ME, Shaw MA, Lower KM, Lewis SM, Bruyere H, Lutcherath V, Gedeon AK, Wallace RH, Scheffer IE, Turner G, Partington M, Frints SGM, Fryns J-P, Sutherland GR, Mulley JC, Gecz J.  Mutations in the human ortholog of Aristaless cause X-linked mental retardation and epilepsy.  Nat Genet. 2002 Apr;30(4):441–445. PMID: 11889467 DOI: 10.1038/ng862 [PubMed: 11889467]
  167. Strug LJ, Clarke T, Chiang T, Chien M, Baskurt Z, Li W, Dorfman R, Bali B, Wirrell E, Kugler SL, Mandelbaum DE, Wolf SM, McGoldrick P, Hardison H, Novotny EJ, Ju J, Greenberg DA, Russo JJ, Pal DK.  Centrotemporal sharp wave EEG trait in Rolandic epilepsy maps to Elongator Protein Complex 4 (ELP4).  Eur J Hum Genet. 2009 Sep;17(9):1171–1181. PMID: 19172991 PMCID: PMC2729813 DOI: 10.1038/ejhg.2008.267 [PMC free article: PMC2729813] [PubMed: 19172991]
  168. Suls A, Kedeken P, Goffin K, Van Esch H, Dupont P, Cassiman D, Kempfle J, Wuttke TV, Weber Y, Lerche H, Afawi Z, Vandenberghe W, Korczyn AD, Berkovic SF, Ekstein D, Kivity S, Ryvlin P, Claes LRF, Deprez L, Maljevic S, Vargas A, Van DYck T, Goossens K, Del-Favero J, Van Laere KV, De Jonghe P, Van Paesschen W.  Paroxysmal exercise-induced dyskinesia and epilepsy is due to mutations in SLC2A1, encoding the glucose transporter GLUT1.  Brain. 2008 Jul;131(Pt 7):1831–1844. PMID: 18577546 PMCID: PMC2442425 DOI: 10.1093/brain/awn113 [PMC free article: PMC2442425] [PubMed: 18577546]
  169. Suls A, Mullen SA, Weber YG, Verhaert K, Ceulemans B, Guerrini R, Wuttke TV, Salvo-Vargas A, Deprez L, Claes LRF, Jordanova A, Berkovic SF, Lerche H, De Jonghe P, Scheffer IE.  Early-onset absence epilepsy caused by mutations in the glucose transporter GLUT1.  Ann Neurol. 2009 Sep;66(3):415–419. PMID: 19798636 DOI: 10.1002/ana.21724 [PubMed: 19798636]
  170. Suzuki T, Delgado-Escueta AV, Aguan K, Alonso ME, Shi J, Hara Y, Nishida M, Numata T, Medina MT, Takeuchi T, Morita R, Bai D, Ganesh S, Sugimoto Y, Inazawa J, Bailey JN, Ochoa A, Jara-Prado A, Rasmussen A, Ramos-Peek J, Cordova S, Rubio-Donnadieu F, Inoue Y, Osawa M, Kaneko S, Oguni H, Mori Y, Yamakawa K.  Mutations in EFHC1 cause juvenile myoclonic epilepsy.  Nat Genet. 2004 Aug;36(8):842–849. doi: 10.1038/ng1393. PMID: 15258581 [PubMed: 15258581]
  171. Szczygiel JA, Danielsen KI, Melin E, Rosenkranz SH, Pankratova S, Ericsson A, Agerman K, Kokaia M, Woldbye DPD.  Gene therapy vector encoding neuropeptide Y and its receptor Y2 for future treatment of epilepsy: Preclinical data in rats,  Front Mol Neurosci, 2020 Dec 4,13:232. PMID: 33343295 PMCID: PMC7746806 DOI: 10.3389/fnmol.2020.603409 [PMC free article: PMC7746806] [PubMed: 33343295]
  172. Tagliabracci VS, Girard JM, Segvich D, Meyer C, Turnbull J, Zhao X, Minassian BA, Depaoli-Roach AA, Roach PJ.  Abnormal metabolism of glycogen phosphate as a cause for Lafora disease.  J Biol Chem. 2008 Dec 5;283(49):33816–33825. doi: 10.1074/jbc.M807428200 PMID: 18852261 [PMC free article: PMC2590708] [PubMed: 18852261]
  173. Tanaka M, Olsen RW, Medina MT, Schwartz E, Alonso ME, Duron RM, Castsro-Ortega R, Martinez-Juarez IE, Pascual-Castroviejo I, Machado-Salas J, Silva R, Bailey JN, Bai D, Ochoa A, Jara-Prado A, Pineda G, Macdonald RL, Delgado-Escueta AV.  Hyperglycosylation and reduced GABA currents of mutated GABRB3 polypeptide in remitting childhood absence epilepsy.  Am J Hum Genet. 2008 Jun;82(6):1249–1261. PMID: 18514161 PMCID: PMC2427288 DOI: 10.1016/j.ajhg.2008.04.020 [PMC free article: PMC2427288] [PubMed: 18514161]
  174. Tang B, Frasinyuk MS, Chikwana VM, Hahalingan KK, Morgan CA, Segvich DM, Bondarenko S, Mrug GP, Wyrebek P, Watt DS, DePaoli-Roach AA, Roach PJ, Hurley TD.  Discovery and development of small-molecule inhibitors of glycogen synthase,  J Med Chem. 2020 Apr 9; 63(7): 3538–3551. Doi: 10.1021/acs.jmedchem.9b01851. [PMC free article: PMC7233370] [PubMed: 32134266]
  175. Temkin O.  The falling sickness: a history of epilepsy from the Greeks to the beginnings of modern neurology. Johns Hopkins University Press, Baltimore, 1971.
  176. Tester DJ, Will ML, Haglund CM, Ackerman MJ.  Compendium of cardiac channel mutations in 541 consecutive unrelated patients referred for long QT syndrome genetic testing.  Heart Rhythm. 2005 May;2(5):507–517. doi: 10.1016/j.hrthm.2005.01.020 PMID: 15840476 [PubMed: 15840476]
  177. Thompson L.  Human gene therapy. Harsh lessons, high hopes.  FDA Consum. 2000 Sep-Oct;34(5):19–24. PMID: 11590787 [PubMed: 11590787]
  178. Turnbull J, DePaoli-Roach AA, Zhao X, Cortez MA, Pencea N, Tiberia E, Piliguian M, Roach PJ, Wang P, Ackerley CA, Minassian BA.  PTG depletion removes Lafora bodies and rescues the fatal epilepsy of Lafora disease.  PLos Genet. 2011 Apr; 7(4):e1002037. doi: 10.1371/journal.pgen.1002037 PMID: 21552327 [PMC free article: PMC3084203] [PubMed: 21552327]
  179. Valassina N, Brusco S, Salamone A, Serra L, Luoni M, Giannelli S, Bido S, Massimino L, Ungaro F, Mazzara PG, D’Adamo P, Lignani G, Broccoli V, Colasante G.  Scn1a gene reactivation after symptom onset rescues pathological phenotypes in a mouse model of Dravet syndrome.  Nat Commun. 2022, Jan 10;13(1):161. PMID: 35013317 PMCID: PMC8748984 DOI: 10.1038/s41467-021-27837-w [PMC free article: PMC8748984] [PubMed: 35013317]
  180. Verma IM.  A tumultuous year for gene therapy.  Mol Ther. 2000 Nov; 2(5):415–416. doi: 10.1006/mthe.2000.0213. PMID: 11082313 [PubMed: 11082313]
  181. von Spiczak S, Helbig KL, Shinde DN, Huether R, Pendziwiat M, Lourenço C, Nunes ME, Sarco DP, Kaplan RA, Dlugos DJ, Kirsch H, Slavotinek A, Cilio MR, Cervenka MC, Cohen JS, McClellan R, Fatemi A, Yuen A, Sagawa Y, Littlejohn R, McLean SD, Hernandez-Hernandez L, Maher B, Møller RS, Palmer E, Lawson JA, Campbell CA, Joshi CN, Kolbe DL, Hollingsworth G, Neubauer BA, Muhle H, Stephani U, Scheffer IE, Pena SDJ, Sisodiya SM, Helbig I; Epi4K Consortium; EuroEPINOMICS-RES NLES Working Group. DNM1 encephalopathy: A new disease of vesicle fission. Neurology. 2017; 89, 385–394. doi: 10.1212/WNL.0000000000004152 PMID: 28667181 [PMC free article: PMC5574673] [PubMed: 28667181]
  182. von Stülpnagel C, Ensslen M, Møller RS, Pal DK, Masnada S, Veggiotti P, Piazza E, Dreesmann M, Hartlieb T, Herberhold T, Hughes E, Koch M, Kutzer C, Hoertnagel K, Nitanda J, Pohl M, Rostásy K, Haack TB, Stöhr K, Kluger G, Borggraefe I.  Epilepsy in patients with GRIN2A alterations : Genetics, neurodevelopment, epileptic phenotype and response to anticonvulsive drugs.  Eur J Paediatr Neurol. 2017 May; 21(3): 530–541. doi: 10.1016/j.ejpn.2017.01.001 PMID: 28109652 [PubMed: 28109652]
  183. Wallace RH, Hodgson BL, Grinton BE, Gardiner RM, Robinson R, Rodriguez-Casero V, Sadleir L, Morgan J, Harkin LA, Dibbens LM, Yamamoto T, Andermann E, Mulley JC, Berkovic SF, Scheffer IE.  Sodium channel alpha1-subunit mutations in severe myoclonic epilepsy of infancy and infantile spasms.  Neurology. 2003 Sep 23;61(6):765–679. PMID: 14504318 DOI: 10.1212/01.wnl.0000086379.71183.78 [PubMed: 14504318]
  184. Wallace RH, Marini C, Petrou S, Harkin LA, Bowser DN, Panchal RG, Williams DA, Sutherland GR, Mulley JC, Scheffer IE, Berkovic SF.  Mutant GABA(A) receptor gamma2-subunit in childhood absence epilepsy and febrile seizures.  Nat Genet.  2001a May;28(1):49–52. doi: 10.1038/ng0501-49. PMID: 11326275 [PubMed: 11326275]
  185. Wallace RH, Scheffer IE, Barnett S, Richards M, Dibbens L, Desai RR, Lerman-Sagie T, Lev D, Mazarib A, Brand N, Ben-Zeev B, Goikhman I, Singh R, Kremmidiotis G, Gardner A, Sutherland GR, George AL Jr, Mulley JC, Berkovic SF.  Neuronal sodium-channel alpha1-subunit mutations in generalized epilepsy with febrile seizures plus.  Am J Hum Genet.  2001b; 68: 859–865. doi: 10.1086/319516 PMID: 11254444 [PMC free article: PMC1275639] [PubMed: 11254444]
  186. Wallace RH, Wang DW, Singh R, Scheffer IE, George AL Jr, Phillips HA, Saar K, Reis A, Johnson EW, Sutherland GR, Berkovic SF, Mulley JC.  Febrile seizures and generalized epilepsy associated with a mutation in the Na+-channel beta1 subunit gene SCN1B.  Nat Genet. 1998 Aug;19(4):366–370. PMID: 9697698 DOI: 10.1038/1252 [PubMed: 9697698]
  187. Wang D, Pascual JM, De Vivo D.  Glucose Transporter Type 1 Deficiency Syndrome. 2002 Jul 30 [updated 2018 Mar 1]. In: Adam MP, Ardinger HH, Pagon RA, Wallace SE, Bean LJH, Gripp KW, Mirzaa GM, Amemiya A, editors. GeneReviews® [Internet]. Seattle (WA): University of Washington, Seattle; 1993–2022. PMID: 20301603
  188. Wang CH, Finkel RS, Bertini E, Schroth M, Simonds A, Wong B, Aloysius A, Morrison L, Main M, Crawford TO, Trela A, Participants of the International Conference on SMA Standard of Care. Consensus statement for standard of care in spinal muscular atrophy.  J Child Neurol. 2007 Aug;22(8):1027–1049. PMID: 17761659 DOI: 10.1177/0883073807305788 [PubMed: 17761659]
  189. Wang D and Gao G.  State-of-the-art human gene therapy: Part II. Gene therapy strategies and clinical applications.  Discov Med.  2014 Sep;18(98):151–161. PMID: 25227756 [PMC free article: PMC4440458] [PubMed: 25227756]
  190. Wang D, Mou H, Li S, Li Y, Hough S, Tran K, Li J, Yin H, Anderson DG, Sontheimer EJ, Weng Z, Gao G, Xue W.  Adenovirus-mediated somatic genome editing of Pten by CRISPR/Cas9 in mouse liver in spite of Cas9-specific immune responses.  Hum Gene Ther. 2015 Jul;26(7),432–442. PMID: 26086867 PMCID: PMC4509492 DOI: 10.1089/hum.2015.087 [PMC free article: PMC4509492] [PubMed: 26086867]
  191. Wang D, Tai PWL, Gao G.  Adeno-associated virus vector as a platform for gene therapy delivery.  Nat Rev Drug Discov. 2019 May;18(5):358–378. doi: 10.1038/s41573-019-0012-9 PMID: 30710128 [PMC free article: PMC6927556] [PubMed: 30710128]
  192. Wang D, Zhang F, Gao G.  CRISPR-based therapeutic genome editing: strategies and in vivo delivery by AAV vectors.  Cell. 2020 Apr 2;181(1):136–150. doi: 10.1016/j.cell.2020.03.023 PMID: 32243786 [PMC free article: PMC7236621] [PubMed: 32243786]
  193. Weaving LS, Christodoulou J, Williamson SL, Friend KL, McKenzie OL, Archer H, Evans J, Clarke A, Pelka GJ, Tam PP, Watson C, Lahooti H, Ellaway CJ, Bennetts B, Leonard H, Gécz J.  Mutations of CDKL5 cause a severe neurodevelopmental disorder with infantile spasms and mental retardation.  Am J Hum Genet. 2004 Dec;75(6):1079–1093. doi: 10.1086/426462 PMID: 15492925 [PMC free article: PMC1182143] [PubMed: 15492925]
  194. Weckhuysen S, Mandelstam S, Suls A, Audenaert D, Deconinck T, Claes LRF, Deprez L, Smets K, Hristova D, Yordanova I, Jordanova A, Ceulemans B, Jansen A, Hasaerts D, Roelens F, Lagae L, Yendle S, Stanley T, Heron SE, Mulley JC, Berkovic SF, Scheffer IE, de Jonghe P.  KNCQ2 encephalopathy: emerging phenotype of a neonatal epileptic encephalopathy.  Ann Neurol. 2012 Jan; 71(1):15–25. doi: 10.1002/ana.22644 PMID: 22275249 [PubMed: 22275249]
  195. West P.  An investigation into the social construction and consequences of the label epilepsy.  Sociology Review. 1979 Nov;27(4):719–741. doi: 10.1111/j.1467-954x.1979.tb00357.x. PMID: 531594 [PubMed: 531594]
  196. Wilson JM.  Lessons learned from the gene therapy trial for ornithine transcarbamylase deficiency.  Mol Genet Metab. 2009 Apr;96(4):151–175. doi: 10.1016/j.ymgme.2008.12.016 PMID: 19211285 [PubMed: 19211285]
  197. Woldbye DPD, Angehagen M, Gøtzsche CR, Elbrønd-Bek H, Sørensen AT, Christiansen SH, Olesen MV, Nikitidou L, Hansen TV, Kanter-Schlifke I, Kokaia M.  Adeno-associated viral vector-induced overexpression of neuropeptide Y Y2 receptors in the hippocampus suppresses seizures.  Brain. 2010 Sep;133(9):2778–2788. doi: 10.1093/brain/awq219 PMID: 20688813 [PubMed: 20688813]
  198. Woldbye DPD, Nanobashvili A, Sørensen AT, Husum H, Bolwig TG, Sørensen G, Ernfors P, Kokaia M.  Differential suppression of seizures via Y2 and Y5 neuropeptide Y receptors.  Neurobiol Dis. 2005 Dec;20(3):760–772. doi: 10.1016/j.nbd.2005.05.010 PMID: 15979311 [PubMed: 15979311]
  199. Winkler H.  Verbreitung und Ursache der Parthenogenesis im Pflanzen-und Tierreiche.  1920. Verlag Fischer, Jena.
  200. Wong M.  Mammalian target of rapamycin (mTOR) inhibition as a potential antiepileptogenic therapy: from tuberous sclerosis to common acquired epilepsies.  Epilepsia. 2010 Jan;51(1):27–36. PMID: 19817806 doi: 10.1111/j.1528-1167.2009.02341.x [PMC free article: PMC3022513] [PubMed: 19817806]
  201. Yang H, Wang D, Engelstad K, Bagay L, Wei Y, Rotstein M, Aggarwal V, Levy B, Ma L, Chung WK, De Vivo DC.  Glut1 deficiency syndrome and erythrocyte glucose uptake assay.  Ann Neurol. 2011 Dec;70(6): 996–1005. doi: 10.1002/ana.22640 PMID: 22190371 [PubMed: 22190371]
Copyright Notice

This is an open access publication, available online and distributed under the terms of a Creative Commons Attribution-Non Commercial-No Derivatives 4.0 International licence (CC BY-NC-ND 4.0), a copy of which is available at https://creativecommons.org/licenses/by-nc-nd/4.0/. Subject to this license, all rights are reserved.

Bookshelf ID: NBK609863PMID: 39637213DOI: 10.1093/med/9780197549469.003.0005

Views

  • PubReader
  • Print View
  • Cite this Page
  • PDF version of this title (283M)

Related information

  • PMC
    PubMed Central citations
  • PubMed
    Links to PubMed

Similar articles in PubMed

See reviews...See all...

Recent Activity

Your browsing activity is empty.

Activity recording is turned off.

Turn recording back on

See more...