1
|
Tsai CW, Ho SY, Chen IC, Chang KC, Chen HJ, Tsai FC, Liou HH. Abnormal increased mTOR signaling regulates seizure threshold in Dravet syndrome. Neuropharmacology 2025; 262:110166. [PMID: 39374769 DOI: 10.1016/j.neuropharm.2024.110166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 09/10/2024] [Accepted: 09/16/2024] [Indexed: 10/09/2024]
Abstract
Excessive activation of mTOR has been observed in the brains of mouse models for Dravet syndrome. We aim to confirm whether that the overactivation of mTOR contributes to the neuropathological changes leading to epileptogenesis and neurobehavior deficits to support a novel pharmacological therapeutic approach for Dravet syndrome. The mTOR inhibitor everolimus, as a clinical antiseizure medication, was utilized to investigate whether mTOR is involved in hyperthermia-induced seizures, anxiety-like, and autism-like behaviors, as well as to explore potential pathogenic mechanisms in Scn1aE1099X/+ mice, a model of Dravet syndrome. First, we found that mTOR signaling was upregulated in hippocampus tissues and neural cultures derived from Scn1aE1099X/+ mice prior to seizure onset. Behaviorally, everolimus increased the seizure threshold and improved anxiety-like and autism-like behaviors in Scn1aE1099X/+ mice. Electrophysiologically, everolimus reduced the frequency of spontaneous excitatory postsynaptic currents in dentate granule neurons from Scn1aE1099X/+ mice. Biochemically, everolimus prevented hyperthermia-induced phosphorylation of hippocampal S6 ribosome in hippocampus, and it delayed hyperthermia-induced increase of cytosolic Ca2+ level in primary neuronal cultures derived from Scn1aE1099X/+ mice. Our results provide the evidence that overactivated mTOR as an important neuropathological change which regulates seizure threshold, impairments of neurobehavior, neuronal glutamatergic transmission and intracellular Ca2+ levels in Scn1aE1099X/+ mice. Inhibition of mTOR is a potential pharmacological therapeutic approach.
Collapse
Affiliation(s)
- Che-Wen Tsai
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei City, Taiwan
| | - Shih-Yin Ho
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei City, Taiwan; Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University College of Medicine, Taipei City, Taiwan; Graduate Institute of Biomedical and Pharmaceutical Science, College of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan; Department of Neurology, Fu Jen Catholic University Hospital, Fu Jen Catholic University, New Taipei City, Taiwan
| | - I Chun Chen
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei City, Taiwan
| | - Kai-Chieh Chang
- Department of Neurology, National Taiwan University Hospital Yunlin Branch, Douliu, Taiwan
| | - Hou-Jen Chen
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei City, Taiwan
| | - Feng-Chiao Tsai
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei City, Taiwan; Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei City, Taiwan
| | - Horng-Huei Liou
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei City, Taiwan; Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University College of Medicine, Taipei City, Taiwan; Graduate Institute of Biomedical and Pharmaceutical Science, College of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan; Department of Neurology, Fu Jen Catholic University Hospital, Fu Jen Catholic University, New Taipei City, Taiwan.
| |
Collapse
|
2
|
Neal ES, Xu W, Borges K. Metabolic aspects of genetic ion channel epilepsies. J Neurochem 2024; 168:3911-3935. [PMID: 37594756 PMCID: PMC11591411 DOI: 10.1111/jnc.15938] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 07/17/2023] [Accepted: 08/01/2023] [Indexed: 08/19/2023]
Abstract
Nowadays, particularly in countries with high incomes, individual mutations in people affected by genetic epilepsies are identified, and genetic therapies are being developed. In addition, drugs are being screened to directly target specific mutations, and personalised medicine is possible. However, people with epilepsy do not yet benefit from these advances, and many types of epilepsies are medication-resistant, including Dravet syndrome. Thus, in the meantime, alternative and effective treatment options are needed. There is increasing evidence that metabolic deficits contribute to epileptic seizures and that such metabolic impairments may be amenable to treatment, with metabolic treatment options like the ketogenic diet being employed with some success. However, the brain metabolic alterations that occur in ion channel epilepsies are not well-understood, nor how these may differ from epilepsies that are of acquired and unknown origins. Here, we provide an overview of studies investigating metabolic alterations in epilepsies caused by mutations in the SCN1A and KCNA1 genes, which are currently the most studied ion channel epilepsies in animal models. The metabolic changes found in these models are likely to contribute to seizures. A metabolic basis of these ion channel epilepsies is supported by human and/or animal studies that show beneficial effects of the ketogenic diet, which may be mediated by the provision of auxiliary brain fuel in the form of ketone bodies. Other potentially more preferred dietary therapies including medium-chain triglycerides and triheptanoin have also been tested in a limited number of studies, but their efficacies remain to be clearly established. The extent to which brain metabolism is affected in people with Dravet syndrome, KCNA1 epilepsy and the models thereof still requires clarification. This requires more experiments that yield functional insight into metabolism.
Collapse
Affiliation(s)
- Elliott S. Neal
- School of Biomedical SciencesThe University of QueenslandSt LuciaQueenslandAustralia
| | - Weizhi Xu
- School of Biomedical SciencesThe University of QueenslandSt LuciaQueenslandAustralia
| | - Karin Borges
- School of Biomedical SciencesThe University of QueenslandSt LuciaQueenslandAustralia
| |
Collapse
|
3
|
Wirrell EC, Lagae L, Scheffer IE, Cross JH, Specchio N, Strzelczyk A. Practical considerations for the use of fenfluramine to manage patients with Dravet syndrome or Lennox-Gastaut syndrome in clinical practice. Epilepsia Open 2024; 9:1643-1657. [PMID: 38962968 PMCID: PMC11450599 DOI: 10.1002/epi4.12998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 05/20/2024] [Accepted: 06/04/2024] [Indexed: 07/05/2024] Open
Abstract
Fenfluramine (FFA), an antiseizure medication (ASM) with serotonergic and sigma-1 receptor activity, is used to manage patients with developmental and epileptic encephalopathies (DEEs). It is approved in the US for treating seizures associated with Dravet syndrome (DS) and Lennox-Gastaut syndrome (LGS) in patients ≥2 years old and as add-on therapy for seizures associated with DS and LGS in the EU, UK, and Japan in similarly aged patients. Consensus guidelines for treatment of DS have recommended FFA to be an early-line ASM, and it has also shown efficacy in managing seizures associated with LGS. DS and LGS are DEEs associated with a range of seizure types, developmental impairments, and multiple comorbidities. Here we provide case vignettes describing 4 patients (3 DS and 1 LGS) aged 4-29 years old in whom up to 14 ASMs had previously failed, to illustrate real-world practice issues encountered by neurologists. This review provides guidance on the use of FFA in the context of ASM polytherapy and drug-drug interactions (DDIs), behavioral issues, dose titration, and adverse events. Along with data from the clinical trial program, these case vignettes emphasize the low risk of DDIs, a generally well-tolerated safety profile, and other seizure and nonseizure benefits (eg, improved cognition and sleep) associated with the use of FFA in DS or LGS. PLAIN LANGUAGE SUMMARY: Fenfluramine is used to treat seizures in individuals with Dravet syndrome and Lennox-Gastaut syndrome, but there are a range of issues that clinicians may face when treating patients. This review highlights four patients from the authors' everyday clinical work and offers guidance and practical considerations by neurologists with expertise in managing these complex conditions related to drug interactions, dosing, and side effects associated with fenfluramine.
Collapse
Affiliation(s)
- Elaine C. Wirrell
- Divisions of Child and Adolescent Neurology and Epilepsy, Department of NeurologyMayo ClinicRochesterMinnesotaUSA
| | - Lieven Lagae
- Member of the European Reference Network EpiCARE, Department of Pediatric NeurologyUniversity of LeuvenLeuvenBelgium
| | - Ingrid E. Scheffer
- Austin Hospital and Royal Children’'s Hospital, Florey and Murdoch Children's Research InstitutesUniversity of MelbourneMelbourneVictoriaAustralia
| | - J. Helen Cross
- Developmental Neurosciences Research & Teaching DepartmentUCL NIHR BRC Great Ormond Street Institute of Child HealthLondonUK
- Department of NeurologyGreat Ormond Street HospitalLondonUK
| | - Nicola Specchio
- Neurology, Epilepsy and Movement Disorders UnitBambino Gesù Children's Hospital, IRCCS, Full Member of European Reference Network EpiCARERomeItaly
| | - Adam Strzelczyk
- Goethe‐University Frankfurt, Epilepsy Center Frankfurt Rhine‐Main and Department of NeurologyUniversity Hospital FrankfurtFrankfurt am MainGermany
| |
Collapse
|
4
|
Sriretnakumar V, Harripaul R, Kennedy JL, So J. When rare meets common: Treatable genetic diseases are enriched in the general psychiatric population. Am J Med Genet A 2024; 194:e63609. [PMID: 38532509 DOI: 10.1002/ajmg.a.63609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 03/06/2024] [Accepted: 03/13/2024] [Indexed: 03/28/2024]
Abstract
Mental illnesses are one of the biggest contributors to the global disease burden. Despite the increased recognition, diagnosis and ongoing research of mental health disorders, the etiology and underlying molecular mechanisms of these disorders are yet to be fully elucidated. Moreover, despite many treatment options available, a large subset of the psychiatric patient population is nonresponsive to standard medications and therapies. There has not been a comprehensive study to date examining the burden and impact of treatable genetic disorders (TGDs) that can present with neuropsychiatric features in psychiatric patient populations. In this study, we test the hypothesis that TGDs that present with psychiatric symptoms are more prevalent within psychiatric patient populations compared to the general population by performing targeted next-generation sequencing of 129 genes associated with 108 TGDs in a cohort of 2301 psychiatric patients. In total, 48 putative affected and 180 putative carriers for TGDs were identified, with known or likely pathogenic variants in 79 genes. Despite screening for only 108 genetic disorders, this study showed a two-fold (2.09%) enrichment for genetic disorders within the psychiatric population relative to the estimated 1% cumulative prevalence of all single gene disorders globally. This strongly suggests that the prevalence of these, and most likely all, genetic diseases is greatly underestimated in psychiatric populations. Increasing awareness and ensuring accurate diagnosis of TGDs will open new avenues to targeted treatment for a subset of psychiatric patients.
Collapse
Affiliation(s)
- Venuja Sriretnakumar
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Ricardo Harripaul
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Canada
- Institute of Medical Science, University of Toronto, Toronto, Canada
| | - James L Kennedy
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Canada
- Department of Psychiatry, University of Toronto, Toronto, Canada
| | - Joyce So
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
- Department of Psychiatry, University of Toronto, Toronto, Canada
- Department of Medicine, University of Toronto, Toronto, Canada
- Division of Medical Genetics, Departments of Medicine and Pediatrics, University of California, San Francisco, California, USA
| |
Collapse
|
5
|
Ng ACH, Chahine M, Scantlebury MH, Appendino JP. Channelopathies in epilepsy: an overview of clinical presentations, pathogenic mechanisms, and therapeutic insights. J Neurol 2024; 271:3063-3094. [PMID: 38607431 DOI: 10.1007/s00415-024-12352-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 03/24/2024] [Accepted: 03/25/2024] [Indexed: 04/13/2024]
Abstract
Pathogenic variants in genes encoding ion channels are causal for various pediatric and adult neurological conditions. In particular, several epilepsy syndromes have been identified to be caused by specific channelopathies. These encompass a spectrum from self-limited epilepsies to developmental and epileptic encephalopathies spanning genetic and acquired causes. Several of these channelopathies have exquisite responses to specific antiseizure medications (ASMs), while others ASMs may prove ineffective or even worsen seizures. Some channelopathies demonstrate phenotypic pleiotropy and can cause other neurological conditions outside of epilepsy. This review aims to provide a comprehensive exploration of the pathophysiology of seizure generation, ion channels implicated in epilepsy, and several genetic epilepsies due to ion channel dysfunction. We outline the clinical presentation, pathogenesis, and the current state of basic science and clinical research for these channelopathies. In addition, we briefly look at potential precision therapy approaches emerging for these disorders.
Collapse
Affiliation(s)
- Andy Cheuk-Him Ng
- Clinical Neuroscience and Pediatric Neurology, Department of Pediatrics, Cumming School of Medicine, Alberta Children's Hospital, University of Calgary, 28 Oki Drive NW, Calgary, AB, T3B 6A8, Canada
- Division of Neurology, Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta and Stollery Children's Hospital, Edmonton, AB, Canada
| | - Mohamed Chahine
- Department of Medicine, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
- CERVO, Brain Research Centre, Quebec City, Canada
| | - Morris H Scantlebury
- Clinical Neuroscience and Pediatric Neurology, Department of Pediatrics, Cumming School of Medicine, Alberta Children's Hospital, University of Calgary, 28 Oki Drive NW, Calgary, AB, T3B 6A8, Canada
- Hotchkiss Brain Institute and Alberta Children's Hospital Research Institute, Calgary, Canada
| | - Juan P Appendino
- Clinical Neuroscience and Pediatric Neurology, Department of Pediatrics, Cumming School of Medicine, Alberta Children's Hospital, University of Calgary, 28 Oki Drive NW, Calgary, AB, T3B 6A8, Canada.
| |
Collapse
|
6
|
Ballesteros-Sayas C, Muñoz-Montero A, Giorgi S, Cardenal-Muñoz E, Turón-Viñas E, Pallardó F, Aibar JÁ. Non-pharmacological therapeutic needs in people with Dravet syndrome. Epilepsy Behav 2024; 150:109553. [PMID: 38035538 DOI: 10.1016/j.yebeh.2023.109553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 11/16/2023] [Accepted: 11/17/2023] [Indexed: 12/02/2023]
Abstract
Dravet syndrome (DS) is a genetic rare disease, which is usually caused by a mutation in the SCN1A gene. DS is characterised by a drug-resistant epilepsy and by cognitive and behavioural disturbances. Thus, DS patients require both pharmacological and non-pharmacological treatments. However, there is a paucity of studies on non-pharmacological therapies and their potential benefits. The main aim of this study was to describe the non-pharmacological therapy modalities received by DS patients and their socio-economic impact on the family. Thus, we designed an online survey addressed to caregivers of DS patients. Our results indicated that up to 91.9% of the surveyed patients required non-pharmacological therapies, which were mainly directed to treat cognitive, sensory and motor impairments. In many cases, the economic costs of these therapies were borne entirely by the families. Nevertheless, patients required a deployment of resources not only at a health care level, but also at an educational level.
Collapse
Affiliation(s)
- Carla Ballesteros-Sayas
- Early Infant Care Center, Hospital San Rafael, C/ San Juan de Dios, 19, 18001 Granada, Spain.
| | | | - Simona Giorgi
- Dravet Syndrome Foundation Spain, C/ Toledo, 46, 1º, 28005 Madrid, Spain.
| | | | - Eulalia Turón-Viñas
- Child Neurology Unit, Hospital de la Santa Creu i Sant Pau, C/ Sant Quintí 89, 08001 Barcelona, Spain.
| | - Federico Pallardó
- University of Valencia, Cellular and Organ Physiopathology of Oxidative Stress Research Group (INCLIVA), Av/ Vicente Blasco Ibáñez, 15, 46010 Valencia, Spain.
| | - José Ángel Aibar
- Dravet Syndrome Foundation Spain, C/ Toledo, 46, 1º, 28005 Madrid, Spain.
| |
Collapse
|
7
|
Tian Y, Shi Z, Cai J, Hou C, Wang X, Zhu H, Peng B, Shi K, Li X, Gong S, Chen WX. Levetiracetam may be an unsuitable choice for patients with PRRT2-associated self-limited infantile epilepsy. BMC Pediatr 2023; 23:529. [PMID: 37880614 PMCID: PMC10601096 DOI: 10.1186/s12887-023-04212-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Accepted: 07/26/2023] [Indexed: 10/27/2023] Open
Abstract
INTRODUCTION Self-limited infantile epilepsy (SeLIE) is a benign epilepsy. Previous studies have shown that monotherapy with most antiseizure medications can effectively relieve seizures in patients with SeLIE, but the efficacy of levetiracetam has not been investigated. OBJECTIVE This study aimed to investigate the efficacy of levetiracetam in the treatment of SeLIE patients with PRRT2 mutations. METHODS The clinical data of 39 SeLIE patients (21 males and 18 females, aged 4.79 ± 1.60 months) with pathogenic variants in PRRT2 or 16p11.2 microdeletion were retrospectively analyzed. Based on the use of initial antiseizure medication (ASM), the patients were classified into two groups: Levetiracetam group (LEG) and Other ASMs group (OAG). The difference of efficacy between the two groups was compared. RESULTS Among the 39 SeLIE patients, 16 were LEG (10 males and 6 females, aged 5.25 ± 2.07 months), with whom two obtained a seizure-free status (12.50%) and 14 ineffective or even deteriorated (87.50%). Among the 14 ineffective or deteriorated cases, 13 were seizure-controlled after replacing levetiracetam with other ASMs including topiramate, oxcarbazepine, lamotrigine, and valproate, and the remaining one finally achieved remission at age 3. Of the 39 patients, 23 were OAG (11 males and 12 females; aged 4.48 ± 1.12 months), of whom 22 achieved seizure remission, except for one patient who was ineffective with topiramate initially and relieved by oxcarbazepine instead. Although there were no significant differences in gender and age of onset between the two groups, the effective rate was significantly different (12.50% in LEG vs. 95.65% in OAG) (P < 0.01). CONCLUSION The findings showed that patients with SeLIE caused by the PRRT2 mutations did not benefit from the use of levetiracetam, but could benefit from other ASMs.
Collapse
Affiliation(s)
- Yang Tian
- The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
- Department of Neurology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, 510623, China
| | - Zhen Shi
- Department of Neurology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, 510623, China
| | - Jiahao Cai
- Department of Neurology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, 510623, China
| | - Chi Hou
- Department of Neurology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, 510623, China
| | - Xiuying Wang
- Department of Neurology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, 510623, China
| | - Haixia Zhu
- Department of Neurology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, 510623, China
| | - Binwei Peng
- Department of Neurology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, 510623, China
| | - Kaili Shi
- Department of Neurology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, 510623, China
| | - Xiaojing Li
- Department of Neurology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, 510623, China
| | - Sitang Gong
- The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China.
- Department of Neurology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, 510623, China.
- Department of Pediartic, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9# Jin Sui Road, Guangzhou, 510623, China.
| | - Wen-Xiong Chen
- Department of Neurology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, 510623, China.
- Department of Neurology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9# Jin Sui Road, Guangzhou, 510623, China.
| |
Collapse
|
8
|
The emergence of genotypic divergence and future precision medicine applications. HANDBOOK OF CLINICAL NEUROLOGY 2023; 192:87-99. [PMID: 36796950 DOI: 10.1016/b978-0-323-85538-9.00013-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Genotypic divergence is a term adapted from population genetics and intimately linked to evolution. We use divergence here to emphasize the differences that set individuals apart in any cohort. The history of genetics is filled with descriptions of genotypic differences, but causal inference of interindividual biological variation has been scarce. We suggest that the practice of precision medicine requires a divergent approach, an approach dependent on the causal interpretation of previous convergent (and preliminary) knowledge in the field. This knowledge has relied on convergent descriptive syndromology (lumping), which has overemphasized a reductionistic gene determinism on the quest of seeking associations without causal understanding. Regulatory variants with small effect and somatic mutations are some of the modifying factors that lead to incomplete penetrance and intrafamilial variable expressivity often observed in apparently monogenic clinical disorders. A truly divergent approach to precision medicine requires splitting, that is, the consideration of different layers of genetic phenomena that interact causally in a nonlinear fashion. This chapter reviews convergences and divergences in genetics and genomics, aiming to discuss what can be causally understood to approximate the as-yet utopian lands of Precision Medicine for patients with neurodegenerative disorders.
Collapse
|
9
|
Haviland I, Daniels CI, Greene CA, Drew J, Love-Nichols JA, Swanson LC, Smith L, Nie DA, Benke T, Sheidley BR, Zhang B, Poduri A, Olson HE. Genetic Diagnosis Impacts Medical Management for Pediatric Epilepsies. Pediatr Neurol 2023; 138:71-80. [PMID: 36403551 PMCID: PMC10099530 DOI: 10.1016/j.pediatrneurol.2022.10.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 10/05/2022] [Accepted: 10/19/2022] [Indexed: 11/18/2022]
Abstract
BACKGROUND Evidence of the impact of genetic diagnosis on medical management in individuals with previously unexplained epilepsy is lacking in the literature. Our goal was to determine the impact of genetic diagnosis on medical management in a cohort of individuals with early-onset epilepsy. METHODS We performed detailed phenotyping of individuals with epilepsy who underwent clinical genetic testing with an epilepsy panel and/or exome sequencing at Boston Children's Hospital between 2012 and 2019. We assessed the impact of genetic diagnosis on medical management. RESULTS We identified a genetic etiology in 152 of 602 (25%) individuals with infantile- or childhood-onset epilepsy who underwent next-generation sequencing. Diagnosis impacted medical management in at least one category for 72% of patients (110 of 152) and in more than one category in 34%. Treatment was impacted in 45% of individuals, including 36% with impact on antiseizure medication choice, 7% on use of disease-specific vitamin or metabolic treatments, 3% on pathway-driven off-label use of medications, and 10% on discussion of gene-specific clinical trials. Care coordination was impacted in 48% of individuals. Counseling on a change in prognosis was reported in 28% of individuals, and 1% of individuals had a correction of diagnosis. Impact was documented in 13 of 13 individuals with neurotypical development and in 55% of those with epilepsy onset after age two years. CONCLUSION We demonstrated meaningful impact of genetic diagnosis on medical care and prognosis in over 70% of individuals, including those with neurotypical development and age of epilepsy onset after age two years.
Collapse
Affiliation(s)
- Isabel Haviland
- Department of Neurology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Carolyn I Daniels
- Department of Neurology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Caitlin A Greene
- Department of Neurology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Jacqueline Drew
- Department of Neurology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts; Boston University Clinical Investigation Master's Program, Boston, Massachusetts
| | - Jamie A Love-Nichols
- Department of Neurology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts; Department of Genetics, Seattle Children's Hospital, Seattle, Washington
| | - Lindsay C Swanson
- Department of Neurology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Lacey Smith
- Division of Epilepsy and Clinical Neurophysiology and Epilepsy Genetics Program, Department of Neurology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Duyu A Nie
- Department of Neurology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts; Departments of Pediatrics, Neurology and Neurosurgery, Warren Alpert Medical School of Brown University, Providence, Rhode Island; Division of Pediatric Neurology and the Children's Neurodevelopment Center (CNDC), Hasbro Children's Hospital, Providence, Rhode Island
| | - Timothy Benke
- Departments of Pediatrics, Neurology, Pharmacology, and Otolaryngology, Children's Hospital Colorado, University of Colorado School of Medicine, Aurora, Colorado
| | - Beth R Sheidley
- Division of Epilepsy and Clinical Neurophysiology and Epilepsy Genetics Program, Department of Neurology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Bo Zhang
- Department of Neurology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts; Biostatistics and Research Design Center, Institutional Centers for Clinical and Translational Research, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Annapurna Poduri
- Department of Neurology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts; Division of Epilepsy and Clinical Neurophysiology and Epilepsy Genetics Program, Department of Neurology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Heather E Olson
- Department of Neurology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts; Division of Epilepsy and Clinical Neurophysiology and Epilepsy Genetics Program, Department of Neurology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
10
|
Strzelczyk A, Schubert-Bast S. Psychobehavioural and Cognitive Adverse Events of Anti-Seizure Medications for the Treatment of Developmental and Epileptic Encephalopathies. CNS Drugs 2022; 36:1079-1111. [PMID: 36194365 PMCID: PMC9531646 DOI: 10.1007/s40263-022-00955-9] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/07/2022] [Indexed: 02/06/2023]
Abstract
The developmental and epileptic encephalopathies encompass a group of rare syndromes characterised by severe drug-resistant epilepsy with onset in childhood and significant neurodevelopmental comorbidities. The latter include intellectual disability, developmental delay, behavioural problems including attention-deficit hyperactivity disorder and autism spectrum disorder, psychiatric problems including anxiety and depression, speech impairment and sleep problems. Classical examples of developmental and epileptic encephalopathies include Dravet syndrome, Lennox-Gastaut syndrome and tuberous sclerosis complex. The mainstay of treatment is with multiple anti-seizure medications (ASMs); however, the ASMs themselves can be associated with psychobehavioural adverse events, and effects (negative or positive) on cognition and sleep. We have performed a targeted literature review of ASMs commonly used in the treatment of developmental and epileptic encephalopathies to discuss the latest evidence on their effects on behaviour, mood, cognition, sedation and sleep. The ASMs include valproate (VPA), clobazam, topiramate (TPM), cannabidiol (CBD), fenfluramine (FFA), levetiracetam (LEV), brivaracetam (BRV), zonisamide (ZNS), perampanel (PER), ethosuximide, stiripentol, lamotrigine (LTG), rufinamide, vigabatrin, lacosamide (LCM) and everolimus. Bromide, felbamate and other sodium channel ASMs are discussed briefly. Overall, the current evidence suggest that LEV, PER and to a lesser extent BRV are associated with psychobehavioural adverse events including aggressiveness and irritability; TPM and to a lesser extent ZNS are associated with language impairment and cognitive dulling/memory problems. Patients with a history of behavioural and psychiatric comorbidities may be more at risk of developing psychobehavioural adverse events. Topiramate and ZNS may be associated with negative effects in some aspects of cognition; CBD, FFA, LEV, BRV and LTG may have some positive effects, while the remaining ASMs do not appear to have a detrimental effect. All the ASMs are associated with sedation to a certain extent, which is pronounced during uptitration. Cannabidiol, PER and pregabalin may be associated with improvements in sleep, LTG is associated with insomnia, while VPA, TPM, LEV, ZNS and LCM do not appear to have detrimental effects. There was variability in the extent of evidence for each ASM: for many first-generation and some second-generation ASMs, there is scant documented evidence; however, their extensive use suggests favourable tolerability and safety (e.g. VPA); second-generation and some third-generation ASMs tend to have the most robust evidence documented over several years of use (TPM, LEV, PER, ZNS, BRV), while evidence is still being generated for newer ASMs such as CBD and FFA. Finally, we discuss how a variety of factors can affect mood, behaviour and cognition, and untangling the associations between the effects of the underlying syndrome and those of the ASMs can be challenging. In particular, there is enormous heterogeneity in cognitive, behavioural and developmental impairments that is complex and can change naturally over time; there is a lack of standardised instruments for evaluating these outcomes in developmental and epileptic encephalopathies, with a reliance on subjective evaluations by proxy (caregivers); and treatment regimes are complex involving multiple ASMs as well as other drugs.
Collapse
Affiliation(s)
- Adam Strzelczyk
- Epilepsy Center Frankfurt Rhine-Main, Center of Neurology and Neurosurgery, Goethe-University and University Hospital Frankfurt, Schleusenweg 2-16, 60528, Frankfurt am Main, Germany. .,LOEWE Center for Personalized and Translational Epilepsy Research (CePTER), Goethe-University Frankfurt, Frankfurt am Main, Germany.
| | - Susanne Schubert-Bast
- Epilepsy Center Frankfurt Rhine-Main, Center of Neurology and Neurosurgery, Goethe-University and University Hospital Frankfurt, Schleusenweg 2-16, 60528, Frankfurt am Main, Germany.,LOEWE Center for Personalized and Translational Epilepsy Research (CePTER), Goethe-University Frankfurt, Frankfurt am Main, Germany.,Department of Neuropediatrics, Goethe-University and University Hospital Frankfurt, Frankfurt am Main, Germany
| |
Collapse
|
11
|
Hahn CD, Jiang Y, Villanueva V, Zolnowska M, Arkilo D, Hsiao S, Asgharnejad M, Dlugos D. A phase 2, randomized, double-blind, placebo-controlled study to evaluate the efficacy and safety of soticlestat as adjunctive therapy in pediatric patients with Dravet syndrome or Lennox-Gastaut syndrome (ELEKTRA). Epilepsia 2022; 63:2671-2683. [PMID: 35841234 PMCID: PMC9804149 DOI: 10.1111/epi.17367] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 07/11/2022] [Accepted: 07/13/2022] [Indexed: 01/05/2023]
Abstract
OBJECTIVE Dravet syndrome (DS) and Lennox-Gastaut syndrome (LGS) are rare treatment-resistant childhood epilepsies classed as developmental and epileptic encephalopathies. ELEKTRA investigated the efficacy and safety of soticlestat (TAK-935) as adjunctive therapy in children with DS or LGS (NCT03650452). METHODS ELEKTRA was a phase 2, randomized, double-blind, placebo-controlled study of soticlestat (≤300 mg twice daily, weight-adjusted) in children (aged 2-17 years) with DS, demonstrating three or more convulsive seizures/month, or with LGS, demonstrating four or more drop seizures/month at baseline. The 20-week treatment period comprised an 8-week dose-optimization period and a 12-week maintenance period. Efficacy endpoints included change from baseline in seizure frequency versus placebo. Safety assessments included incidence of treatment-emergent adverse events (TEAEs). RESULTS ELEKTRA enrolled 141 participants; 126 (89%) completed the study. The modified intent-to-treat population included 139 participants who received one or more doses of study drug and had one or more efficacy assessments (DS, n = 51; LGS, n = 88). ELEKTRA achieved its primary endpoint: the combined soticlestat-treated population demonstrated a placebo-adjusted median reduction in seizure frequency of 30.21% during the maintenance period (p = .0008, n = 139). During this period, placebo-adjusted median reductions in convulsive and drop seizure frequencies of 50.00% (p = .0002; patients with DS) and 17.08% (p = .1160; patients with LGS), respectively, were observed. TEAE incidences were similar between the soticlestat (80.3%) and placebo (74.3%) groups and were mostly mild or moderate in severity. Serious TEAEs were reported by 15.5% and 18.6% of participants receiving soticlestat and placebo, respectively. TEAEs reported in soticlestat-treated patients with ≥5% difference from placebo were lethargy and constipation. No deaths were reported. SIGNIFICANCE Soticlestat treatment resulted in statistically significant, clinically meaningful reductions from baseline in median seizure frequency (combined patient population) and in convulsive seizure frequency (DS cohort). Drop seizure frequency showed a nonstatistically significant numerical reduction in children with LGS. Soticlestat had a safety profile consistent with previous studies.
Collapse
Affiliation(s)
- Cecil D. Hahn
- Division of Neurology, Hospital for Sick Children and Department of PaediatricsUniversity of TorontoTorontoOntarioCanada
| | - Yuwu Jiang
- Department of PediatricsPeking University First HospitalBeijingChina
| | - Vicente Villanueva
- Refractory Epilepsy UnitLa Fe University and Polytechnic HospitalValenciaSpain
| | | | | | - Samuel Hsiao
- Takeda Pharmaceutical Company LimitedCambridgeMassachusettsUSA
| | | | - Dennis Dlugos
- Division of Neurology, Children's Hospital of PhiladelphiaUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
12
|
Suvekbala V, Ramachandran H, Veluchamy A, Mascarenhas MAB, Ramprasath T, Nair MKC, Garikipati VNS, Gundamaraju R, Subbiah R. The Promising Epigenetic Regulators for Refractory Epilepsy: An Adventurous Road Ahead. Neuromolecular Med 2022:10.1007/s12017-022-08723-0. [DOI: 10.1007/s12017-022-08723-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 07/13/2022] [Indexed: 10/14/2022]
|
13
|
Lopez JC, Pare JR, Blackmer AB, Orth LE. Treatment-Refractory Dravet Syndrome: Considerations for Novel Medications. J Pediatr Health Care 2022; 36:479-488. [PMID: 35987555 DOI: 10.1016/j.pedhc.2022.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 03/22/2022] [Accepted: 05/02/2022] [Indexed: 10/15/2022]
Abstract
Before 2018, there were no U.S. Food and Drug Administration-approved medications for managing seizures in Dravet syndrome (DS). Common agents used in the antiepileptic drug regimens of patients with DS included clobazam, valproic acid, topiramate, and levetiracetam, among others; however, these agents alone rarely provide adequate seizure control. Management of seizures in DS changed in recent years with the approval of cannabidiol and stiripentol in 2018 and fenfluramine in 2020. This continuing education article summarizes available efficacy and safety data involving cannabidiol, stiripentol, and fenfluramine and provides a practical review of dosing strategies, pharmacokinetics, and monitoring interventions relevant to their use.
Collapse
Affiliation(s)
- Juan Carlos Lopez
- Juan Carlos Lopez, PGY-1 Pharmacy Resident, Department of Pharmacy, Boston Medical Center, Boston, MA
| | - Jasmine R Pare
- Jasmine R. Pare, PGY-2 Pharmacy Resident, Department of Pharmacy, UCSF Benioff Children's Hospital, San Francisco, CA
| | - Allison B Blackmer
- Allison B. Blackmer, Adjunct Associate Professor of Pharmacy, Department of Clinical Pharmacy, University of Colorado Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, CO
| | - Lucas E Orth
- Lucas E. Orth, Assistant Professor of Pharmacy, Department of Pharmacy, Children's Hospital Colorado, Aurora, CO.
| |
Collapse
|
14
|
Gil-Nagel A, Falip M, Sánchez-Carpintero R, Abad-Sazatornil MR, Poveda JL, Aibar JÁ, Cardenal-Muñoz E, Aras LM, Sánchez R, Sancho-López A, Trillo-Mata JL, Torrejón M, Gil A. The contribution of fenfluramine to the treatment of Dravet syndrome in Spain through Multi-Criteria Decision Analysis. Epilepsy Behav 2022; 132:108711. [PMID: 35588562 DOI: 10.1016/j.yebeh.2022.108711] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 04/07/2022] [Accepted: 04/16/2022] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Dravet Syndrome (DS) is a severe, developmental epileptic encephalopathy (DEE) that begins in infancy and is characterized by pharmaco-resistant epilepsy and neurodevelopmental delay. Despite available antiseizure medications (ASMs), there is a need for new therapeutic options with greater efficacy in reducing seizure frequency and with adequate safety and tolerability profiles. Fenfluramine is a new ASM for the treatment of seizures associated with DS as add-on therapy to other ASMs for patients aged 2 years and older. Fenfluramine decreases seizure frequency, prolongs periods of seizure freedom potentially helping to reduce risk of Sudden Unexpected Death in Epilepsy (SUDEP) and improves patient cognitive abilities positively impacting on patients' Quality of Life (QoL). Reflective Multi-Criteria Decision Analysis (MCDA) methodology allows to determine what represents value in a given indication considering all relevant criteria for healthcare decision-making in a transparent and systematic manner from the perspective of relevant stakeholders. The aim of this study was to determine the relative value contribution of fenfluramine for the treatment of DS in Spain using MCDA. METHOD A literature review was performed to populate an adapted a MCDA framework for orphan-drug evaluation in Spain. A panel of ten Spanish experts, including neurologists, hospital pharmacists, patient representatives and decision-makers, scored four comparative evidence matrices. Results were analyzed and discussed in a group meeting through reflective MCDA discussion methodology. RESULTS Dravet syndrome is considered a severe, rare disease with significant unmet needs. Fenfluramine is perceived to have a higher efficacy profile than all available alternatives, with a better safety profile than stiripentol and topiramate and to provide improved QoL versus studied alternatives. Fenfluramine results in lower other medical costs in comparison with stiripentol and clobazam. Participants perceived that fenfluramine could lead to indirect costs savings compared to available alternatives due to its efficacy in controlling seizures. Overall, fenfluramine's therapeutic impact on patients with DS is considered high and supported by high-quality evidence. CONCLUSIONS Based on reflective MCDA, fenfluramine is considered to add greater benefit in terms of efficacy, safety and QoL when compared with available ASMs.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Alicia Gil
- Omakase Consulting S.L., Barcelona, Spain.
| |
Collapse
|
15
|
Bai YF, Zeng C, Jia M, Xiao B. Molecular mechanisms of topiramate and its clinical value in epilepsy. Seizure 2022; 98:51-56. [DOI: 10.1016/j.seizure.2022.03.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 03/29/2022] [Accepted: 03/31/2022] [Indexed: 10/18/2022] Open
|
16
|
Andrade DM, Berg AT, Hood V, Knupp KG, Koh S, Laux L, Meskis MA, Miller I, Perry MS, Scheffer IE, Sullivan J, Villas N, Wirrell E. Dravet syndrome: A quick transition guide for the adult neurologist. Epilepsy Res 2021; 177:106743. [PMID: 34624600 DOI: 10.1016/j.eplepsyres.2021.106743] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/02/2021] [Accepted: 08/13/2021] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Dravet syndrome (DS) is still seen as a "pediatric disease", where patients receive excellent care in pediatric centers, but care is less than optimal in adult health care systems (HCS). This creates a barrier when young adults need to leave the family-centered pediatric system and enter the adult, patient-centered HCS. Here we create a guide to help with the transition from pediatric to adult for patients with DS. METHODS Experts in Dravet syndrome flagged the main barriers in caring for adults with DS and created a 2-page transition summary guide based on their expertise and a literature review. RESULTS The 2-page guide addresses: DS diagnosis in children and adults; clinical manifestations, including the differences in seizures types and frequencies between children and adults with DS; the natural history of intellectual disability, behavior, gait, motor disorders and dysautonomia; a review of optimal treatments (including medications not commonly used in adult epilepsy settings such as stiripentol and fenfluramine), as well as emergency seizure management; avoidance of triggers, preventive measures, and vaccine administration in adults with DS. CONCLUSION Several young adults with DS are still followed by their child neurologist. This 2-page transition guide should help facilitate the transition of patients with DS to the adult HCS and should be given to families as well as adult health care providers that may not be familiar with DS.
Collapse
Affiliation(s)
- Danielle M Andrade
- Adult Epilepsy Genetics Program, Division of Neurology, Krembil Brain Institute, Toronto Western Hospital, University of Toronto, Toronto, Canada.
| | - Anne T Berg
- Ann & Robert H. Lurie Children's Hospital of Chicago, Departments of Pediatrics and Neurological Surgery, Northwestern Feinberg School of Medicine, Chicago, USA
| | | | - Kelly G Knupp
- Department of Pediatrics and Neurology, University of Colorado Anschutz Campus, Aurora, CO, USA
| | - Sookyong Koh
- Department of Pediatric Neurology at University of Nebraska Medical Center, Omaha, NE, USA
| | - Linda Laux
- Epilepsy Center, Department of Pediatrics, Ann & Robert H. Lurie Children's Hospital, Chicago, IL, USA
| | | | - Ian Miller
- Department of Neurology, Nicklaus Children's Hospital, Miami, FL, USA
| | - M Scott Perry
- Jane and John Justin Neurosciences Center, Cook Children's Medical Center, Fort Worth, TX, USA
| | - Ingrid E Scheffer
- Epilepsy Research Centre, The University of Melbourne, Austin Health, Royal Children's Hospital, Florey Institute, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Joseph Sullivan
- Department of Neurology & Pediatrics, University of California, San Francisco, CA, USA
| | | | - Elaine Wirrell
- Child and Adolescent Neurology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
17
|
Next-generation sequencing in childhood-onset epilepsies: Diagnostic yield and impact on neuronal ceroid lipofuscinosis type 2 (CLN2) disease diagnosis. PLoS One 2021; 16:e0255933. [PMID: 34469436 PMCID: PMC8409681 DOI: 10.1371/journal.pone.0255933] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 07/27/2021] [Indexed: 11/19/2022] Open
Abstract
Epilepsy is one of the most common childhood-onset neurological conditions with a genetic etiology. Genetic diagnosis provides potential for etiologically-based management and treatment. Existing research has focused on early-onset (<24 months) epilepsies; data regarding later-onset epilepsies is limited. The goal of this study was to determine the diagnostic yield of a clinically available epilepsy panel in a selected pediatric epilepsy cohort with epilepsy onset between 24-60 months of life and evaluate whether this approach decreases the age of diagnosis of neuronal ceroid lipofuscinosis type 2 (CLN2). Next-generation sequencing (NGS)-based epilepsy panels, including genes associated with epileptic encephalopathies and inborn errors of metabolism (IEMs) that present with epilepsy, were used. Copy-number variant (CNV) detection from NGS data was included. Variant interpretation was performed per American College of Medical Genetics and Genomics (ACMG) guidelines. Results are reported from 211 consecutive patients with the following inclusion criteria: 24-60 months of age at the time of enrollment, first unprovoked seizure at/after 24 months, and at least one additional finding such as EEG/MRI abnormalities, speech delay, or motor symptoms. Median age was 42 months at testing and 30 months at first seizure onset; the mean delay from first seizure to comprehensive genetic testing was 10.3 months. A genetic diagnosis was established in 43 patients (20.4%). CNVs were reported in 25.6% diagnosed patients; 27.3% of CNVs identified were intragenic. Within the diagnosed cohort, 11 (25.6%) patients were diagnosed with an IEM. The predominant molecular diagnosis was CLN2 (14% of diagnosed patients). For these patients, diagnosis was achieved 12-24 months earlier than reported by natural history of the disease. This study supports comprehensive genetic testing for patients whose first seizure occurs ≥ 24 months of age. It also supports early application of testing in this age group, as the identified diagnoses can have significant impact on patient management and outcome.
Collapse
|
18
|
Devi N, Madaan P, Asrar MM, Sahu JK, Bansal D. Comparative short-term efficacy and safety of add-on anti-seizure medications in Dravet syndrome: An indirect treatment comparison. Seizure 2021; 91:316-324. [PMID: 34274891 DOI: 10.1016/j.seizure.2021.06.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 06/15/2021] [Accepted: 06/18/2021] [Indexed: 10/21/2022] Open
Abstract
PURPOSE Although cannabidiol and fenfluramine have been recently approved by the US Food and Drug Administration (FDA) for seizures in children with Dravet syndrome (DS), the comparative efficacy and safety of these and stiripentol as an add-on therapy for DS has not been evaluated in head-to-head trials. The current study aimed to assess the comparative efficacy and safety of add-on anti-seizure medications in DS. METHODS PubMed and EMBASE database search and a manual search was done using keywords; "antiepileptic", "Dravet syndrome" and "antiseizure". The primary efficacy outcome was ≥50% reduction in convulsive seizure frequency from baseline while the safety outcome was treatment-emergent adverse events (TEAEs). Frequentist approach were used for combining direct and indirect evidence and network plots prepared. The drugs were ranked based on p-scores obtained using the surface under the cumulative ranking (SUCRA). Heterogeneity across studies was calculated by I2 statistic and Q test. RESULTS Five randomized controlled trials (RCTs) with 565 patients with DS (2-20 years) who received placebo or any of the three active interventions (stiripentol, cannabidiol, and fenfluramine) were included. Compared with placebo, all the three drugs were associated with a significant reduction in convulsive seizure frequency from baseline. Stiripentol had the highest probability ranking for ≥50% reduction in convulsive seizure frequency from baseline [OR: 20.2; 95% CI: 2.1-198.0] and for occurrence of any treatment emergent adverse events (TEAEs) [OR:53.9; 95% CI: 1.4 to 2079.8] followed by fenfluramine and cannabidiol. However, for serious TEAEs, the ranking order was stiripentol followed by cannabidiol and fenfluramine. The trial on stiripentol had limited sample size explaining the wide confidence intervals for the comparative outcomes. CONCLUSION In this indirect comparison, fenfluramine and stiripentol hadd comparable efficacy but fenfluramine appeareded to be safer in terms of less frequent serious TEAEs. Cannabidiol had relatively lower efficacy and was associated with serious TEAEs. A head-to-head trial between stiripentol, cannabidiol and fenfluramine is the need of the hour.
Collapse
Affiliation(s)
- Nagita Devi
- Department of Pharmacy Practice, National Institute of Pharmaceutical Education and Research, SAS Nagar, Punjab 160062, India
| | - Priyanka Madaan
- Pediatric Neurology Unit, Department of Pediatrics, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Mir Mahmood Asrar
- Department of Pharmacy Practice, National Institute of Pharmaceutical Education and Research, SAS Nagar, Punjab 160062, India
| | - Jitendra Kumar Sahu
- Pediatric Neurology Unit, Department of Pediatrics, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Dipika Bansal
- Department of Pharmacy Practice, National Institute of Pharmaceutical Education and Research, SAS Nagar, Punjab 160062, India.
| |
Collapse
|
19
|
Miziak B, Czuczwar S. Advances in the design and discovery of novel small molecule drugs for the treatment of Dravet Syndrome. Expert Opin Drug Discov 2020; 16:579-593. [PMID: 33275464 DOI: 10.1080/17460441.2021.1857722] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Introduction: Dravet syndrome (severe myoclonic epilepsy in infancy) begins in the first year of life characterized by generalized or unilateral clonic seizures that are frequently triggered by high fever. A subsequent worsening stage occurs (in years 1-4 of life) and seizure activity is accompanied by disturbed psychomotor development. The third stage of the disease, known as the 'stabilization phase,' is associated with seizures and intellectual impairment. Of note, a mutation in the voltage-gated sodium-channel gene α 1 subunit (SCN1A) has been found in around 85% of patients with Dravet syndrome.Areas covered: The authors review the current treatment strategies as well as potential drugs in the initial stages of clinical evaluation. The authors also review drugs with protective activity in mice models of Dravet syndrome.Expert opinion: Experimental data and results from initial clinical studies have brought attention to several drugs with various mechanisms of action including: ataluren (a suppressant of premature stop codons; under clinical evaluation), EPX-100, EPX-200, fenfluramine (serotonin modulators), soticlestat (an 24-hydroxylase cholesterol enzyme inhibitor), SPN-817 (an inhibitor of acetylcholinesterase), verapamil (a voltage-dependent calcium channel inhibitor) and STK-001 (an antisense oligonucleotide). The latter is scheduled for clinical evaluation.
Collapse
Affiliation(s)
- Barbara Miziak
- Department of Pathophysiology, Medical University of Lublin, Lublin, Poland
| | - Stanisław Czuczwar
- Department of Pathophysiology, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
20
|
Pharmacokinetic Variability During Long-Term Therapeutic Drug Monitoring of Valproate, Clobazam, and Levetiracetam in Patients With Dravet Syndrome. Ther Drug Monit 2020; 42:744-753. [DOI: 10.1097/ftd.0000000000000781] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
21
|
Miceli F, Carotenuto L, Barrese V, Soldovieri MV, Heinzen EL, Mandel AM, Lippa N, Bier L, Goldstein DB, Cooper EC, Cilio MR, Taglialatela M, Sands TT. A Novel Kv7.3 Variant in the Voltage-Sensing S 4 Segment in a Family With Benign Neonatal Epilepsy: Functional Characterization and in vitro Rescue by β-Hydroxybutyrate. Front Physiol 2020; 11:1040. [PMID: 33013448 PMCID: PMC7498716 DOI: 10.3389/fphys.2020.01040] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 07/29/2020] [Indexed: 01/09/2023] Open
Abstract
Pathogenic variants in KCNQ2 and KCNQ3, paralogous genes encoding Kv7.2 and Kv7.3 voltage-gated K+ channel subunits, are responsible for early-onset developmental/epileptic disorders characterized by heterogeneous clinical phenotypes ranging from benign familial neonatal epilepsy (BFNE) to early-onset developmental and epileptic encephalopathy (DEE). KCNQ2 variants account for the majority of pedigrees with BFNE and KCNQ3 variants are responsible for a much smaller subgroup, but the reasons for this imbalance remain unclear. Analysis of additional pedigrees is needed to further clarify the nature of this genetic heterogeneity and to improve prediction of pathogenicity for novel variants. We identified a BFNE family with two siblings and a parent affected. Exome sequencing on samples from both parents and siblings revealed a novel KCNQ3 variant (c.719T>G; p.M240R), segregating in the three affected individuals. The M240 residue is conserved among human Kv7.2-5 and lies between the two arginines (R5 and R6) closest to the intracellular side of the voltage-sensing S4 transmembrane segment. Whole cell patch-clamp recordings in Chinese hamster ovary (CHO) cells revealed that homomeric Kv7.3 M240R channels were not functional, whereas heteromeric channels incorporating Kv7.3 M240R mutant subunits with Kv7.2 and Kv7.3 displayed a depolarizing shift of about 10 mV in activation gating. Molecular modeling results suggested that the M240R substitution preferentially stabilized the resting state and possibly destabilized the activated state of the Kv7.3 subunits, a result consistent with functional data. Exposure to β-hydroxybutyrate (BHB), a ketone body generated during the ketogenic diet (KD), reversed channel dysfunction induced by the M240R variant. In conclusion, we describe the first missense loss-of-function (LoF) pathogenic variant within the S4 segment of Kv7.3 identified in patients with BFNE. Studied under conditions mimicking heterozygosity, the M240R variant mainly affects the voltage sensitivity, in contrast to previously analyzed BFNE Kv7.3 variants that reduce current density. Our pharmacological results provide a rationale for the use of KD in patients carrying LoF variants in Kv7.2 or Kv7.3 subunits.
Collapse
Affiliation(s)
- Francesco Miceli
- Department of Neuroscience, University of Naples “Federico II”, Naples, Italy
| | - Lidia Carotenuto
- Department of Neuroscience, University of Naples “Federico II”, Naples, Italy
| | - Vincenzo Barrese
- Department of Neuroscience, University of Naples “Federico II”, Naples, Italy
| | | | - Erin L. Heinzen
- Eshelman School of Pharmacy, Division of Pharmacotherapy and Experimental Therapeutics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, NY, United States
| | - Arthur M. Mandel
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, United States
| | - Natalie Lippa
- Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, NY, United States
| | - Louise Bier
- Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, NY, United States
| | - David B. Goldstein
- Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, NY, United States
| | - Edward C. Cooper
- Departments of Neurology, Neuroscience, and Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
| | - Maria Roberta Cilio
- Department of Pediatrics and Institute of Experimental and Clinical Research, Cliniques universitaires Saint-Luc, Université catholique de Louvain, Brussels, Belgium
| | | | - Tristan T. Sands
- Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, NY, United States
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, United States
| |
Collapse
|
22
|
Heger K, Lund C, Larsen Burns M, Bjørnvold M, Sætre E, Johannessen SI, Johannessen Landmark C. A retrospective review of changes and challenges in the use of antiseizure medicines in Dravet syndrome in Norway. Epilepsia Open 2020; 5:432-441. [PMID: 32913951 PMCID: PMC7469772 DOI: 10.1002/epi4.12413] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/08/2020] [Accepted: 06/13/2020] [Indexed: 01/05/2023] Open
Abstract
OBJECTIVE Dravet syndrome is a developmental and epileptic encephalopathy characterized by severe and drug-resistant seizures in early childhood, followed by developmental delay. The purpose of this study was to investigate aspects of pharmacological treatment of Norwegian patients with Dravet syndrome, focusing on the use of antiseizure medicines (ASMs) and identifying treatment challenges. METHODS Patients were identified through medical registries at the National Center for Epilepsy in Norway and National Center for Rare Epilepsy Related Disorders during 2008-2018. Additional clinical data were obtained from medical records and laboratory request forms. RESULTS We identified 53 patients with Dravet syndrome, 30/23 males/females, aged 2-50 years. The majority of patients with known seizure frequency experienced frequent seizures, 80% (n = 35/44). Only two patients were seizure-free. Valproate (n = 48), clobazam (n = 45), levetiracetam (n = 30), and stiripentol (n = 38) were most commonly used, previous or current use. More than one-third (n = 20) had tried sodium channel blockers (including lamotrigine), but these drugs were used less during the last decade. Polytherapy was common, 81% (n = 43) used two or more ASMs, and eight of these patients used 4-5 drugs (15%). Several challenges were identified: high seizure frequency, comorbidities, treatment changes with a wide range of ASMs, common use of oral gastro-tubes, extensive polypharmacy, and drug interactions. SIGNIFICANCE The use of ASMs has changed over the last decade, in accordance with updated international recommendations. Various treatment challenges were identified. This vulnerable group of patients needs close follow-up for an optimal treatment outcome.
Collapse
Affiliation(s)
- Katrine Heger
- Program for PharmacyDepartment of Life Sciences and HealthFaculty of Health SciencesOslo Metropolitan UniversityOsloNorway
| | - Caroline Lund
- Department of Rare Genetic SyndromesOslo University HospitalOsloNorway
- Department of NeurohabilitationOslo University HospitalOsloNorway
| | - Margrete Larsen Burns
- Section for Clinical PharmacologyThe National Center for EpilepsyDepartment of PharmacologyOslo University HospitalOsloNorway
| | - Marit Bjørnvold
- The National Center for EpilepsyOslo University HospitalOsloNorway
| | - Erik Sætre
- The National Center for EpilepsyOslo University HospitalOsloNorway
| | - Svein I. Johannessen
- Section for Clinical PharmacologyThe National Center for EpilepsyDepartment of PharmacologyOslo University HospitalOsloNorway
- The National Center for EpilepsyOslo University HospitalOsloNorway
| | - Cecilie Johannessen Landmark
- Program for PharmacyDepartment of Life Sciences and HealthFaculty of Health SciencesOslo Metropolitan UniversityOsloNorway
- Section for Clinical PharmacologyThe National Center for EpilepsyDepartment of PharmacologyOslo University HospitalOsloNorway
- The National Center for EpilepsyOslo University HospitalOsloNorway
| |
Collapse
|
23
|
Menezes LFS, Sabiá Júnior EF, Tibery DV, Carneiro LDA, Schwartz EF. Epilepsy-Related Voltage-Gated Sodium Channelopathies: A Review. Front Pharmacol 2020; 11:1276. [PMID: 33013363 PMCID: PMC7461817 DOI: 10.3389/fphar.2020.01276] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 07/31/2020] [Indexed: 12/29/2022] Open
Abstract
Epilepsy is a disease characterized by abnormal brain activity and a predisposition to generate epileptic seizures, leading to neurobiological, cognitive, psychological, social, and economic impacts for the patient. There are several known causes for epilepsy; one of them is the malfunction of ion channels, resulting from mutations. Voltage-gated sodium channels (NaV) play an essential role in the generation and propagation of action potential, and malfunction caused by mutations can induce irregular neuronal activity. That said, several genetic variations in NaV channels have been described and associated with epilepsy. These mutations can affect channel kinetics, modifying channel activation, inactivation, recovery from inactivation, and/or the current window. Among the NaV subtypes related to epilepsy, NaV1.1 is doubtless the most relevant, with more than 1500 mutations described. Truncation and missense mutations are the most observed alterations. In addition, several studies have already related mutated NaV channels with the electrophysiological functioning of the channel, aiming to correlate with the epilepsy phenotype. The present review provides an overview of studies on epilepsy-associated mutated human NaV1.1, NaV1.2, NaV1.3, NaV1.6, and NaV1.7.
Collapse
Affiliation(s)
- Luis Felipe Santos Menezes
- Laboratório de Neurofarmacologia, Departamento de Ciências Fisiológicas, Universidade de Brasília, Brasília, Brazil
| | - Elias Ferreira Sabiá Júnior
- Laboratório de Neurofarmacologia, Departamento de Ciências Fisiológicas, Universidade de Brasília, Brasília, Brazil
| | - Diogo Vieira Tibery
- Laboratório de Neurofarmacologia, Departamento de Ciências Fisiológicas, Universidade de Brasília, Brasília, Brazil
| | - Lilian Dos Anjos Carneiro
- Faculdade de Medicina, Centro Universitário Euro Americano, Brasília, Brazil.,Faculdade de Medicina, Centro Universitário do Planalto Central, Brasília, Brazil
| | - Elisabeth Ferroni Schwartz
- Laboratório de Neurofarmacologia, Departamento de Ciências Fisiológicas, Universidade de Brasília, Brasília, Brazil
| |
Collapse
|
24
|
Strzelczyk A, Schubert-Bast S. Therapeutic advances in Dravet syndrome: a targeted literature review. Expert Rev Neurother 2020; 20:1065-1079. [PMID: 32799683 DOI: 10.1080/14737175.2020.1801423] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
INTRODUCTION Dravet syndrome (DS), a prototypic developmental and genetic epileptic encephalopathy (DEE), is characterized by an early onset of treatment-refractory seizures, together with impairments in motor control, behavior, and cognition. Even with multiple conventional anti-epileptic drugs, seizures remain poorly controlled, and there has been a considerable unmet need for effective and tolerable treatments. AREAS COVERED This targeted literature review aims to highlight recent changes to the therapeutic landscape for DS by summarizing the most up-to-date, evidence-based research, including pivotal data from the clinical development of stiripentol, cannabidiol, and fenfluramine, which are important milestones for DS treatment, together with the latest findings of other pharmacotherapies in development. In phase III, double-blind, placebo-controlled randomized controlled trials stiripentol, cannabidiol, and fenfluramine have shown clinically relevant reductions in convulsive seizure frequency, and are generally well tolerated. Stiripentol was associated with responder rates (greater than 50% reduction in convulsive seizure frequency) of 67%-71%, when added to valproic acid and clobazam; cannabidiol was associated with responder rates of 43%-49% (48%-63% in conjunction with clobazam), and fenfluramine of 54%-68% across studies. Therapies in development include soticlestat, ataluren, verapamil, and clemizole, with strategies to treat the underlying cause of DS, including gene therapy and antisense oligonucleotides beginning to emerge from preclinical studies. EXPERT OPINION Despite the challenges of drug development in rare diseases, this is an exciting time for the treatment of DS, with the promise of new efficacious and well-tolerated therapies, which may pave the way for treatment advances in other DEEs.
Collapse
Affiliation(s)
- Adam Strzelczyk
- Epilepsy Center Frankfurt Rhine-Main, Center of Neurology and Neurosurgery, Goethe-University Frankfurt , Frankfurt am Main, Germany.,LOEWE Center for Personalized and Translational Epilepsy Research (CePTER), Goethe-University Frankfurt , Frankfurt am Main, Germany
| | - Susanne Schubert-Bast
- Epilepsy Center Frankfurt Rhine-Main, Center of Neurology and Neurosurgery, Goethe-University Frankfurt , Frankfurt am Main, Germany.,LOEWE Center for Personalized and Translational Epilepsy Research (CePTER), Goethe-University Frankfurt , Frankfurt am Main, Germany.,Department of Neuropediatrics, Goethe-University Frankfurt , Frankfurt am Main, Germany
| |
Collapse
|
25
|
Visa-Reñé N, Raspall-Chaure M, Paredes-Carmona F, Coromina JS, Macaya-Ruiz A. Clinical experience with brivaracetam in a series of 46 children. Epilepsy Behav 2020; 107:107067. [PMID: 32302941 DOI: 10.1016/j.yebeh.2020.107067] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 03/16/2020] [Accepted: 03/22/2020] [Indexed: 12/27/2022]
Abstract
OBJECTIVES The primary objective of the study was to analyze the efficacy of brivaracetam (BRV) in pediatric patients 12 months after starting treatment. The secondary objective was to establish safety 3, 6, and 12 months after starting treatment. MATERIALS AND METHOD This was an observational and retrospective study. Data were collected from the electronic medical record. Inclusion criteria were as follows: patients under 18 years of age, diagnosis of focal or generalized epilepsy, treatment as an added therapy, initiation of treatment with BRV between June and September 2017, and at least one unprovoked seizure in the year prior to the start of treatment. RESULTS Forty-six patients were included. The response rate was 65%, including 30% seizure-free patients. The rate of adverse effects was 43.5%, resulting in withdrawal in 16 patients (34.7%). The most common adverse effects were drowsiness (17.3%) and irritability (17.3%). CONCLUSIONS Brivaracetam is effective in very diverse childhood epilepsies, including some that present with primarily generalized seizures. Given the characteristics of the population studied, we have not been able to confirm a better tolerability of BRV compared with levetiracetam (LEV).
Collapse
Affiliation(s)
- Núria Visa-Reñé
- Paediatric Department, Hospital Universitari Arnau de Vilanova [Arnau de Vilanova University Hospital], Lleida, Spain.
| | - Miquel Raspall-Chaure
- Paediatric Neurology Department, Hospital Materno-Infantil Vall d'Hebron [Vall d'Hebron Maternity-Neonatal Hospital], Barcelona, Spain
| | - Fernando Paredes-Carmona
- Paediatric Department, Hospital Universitari Arnau de Vilanova [Arnau de Vilanova University Hospital], Lleida, Spain
| | - Júlia Sala Coromina
- Paediatric Neurology Department, Hospital Materno-Infantil Vall d'Hebron [Vall d'Hebron Maternity-Neonatal Hospital], Barcelona, Spain
| | - Alfons Macaya-Ruiz
- Paediatric Neurology Department, Hospital Materno-Infantil Vall d'Hebron [Vall d'Hebron Maternity-Neonatal Hospital], Barcelona, Spain
| |
Collapse
|
26
|
Abstract
Stiripentol (Diacomit®) is an orally-active, structurally unique anti-epileptic drug (AED) with multiple potential mechanisms of action, including enhancement of central γ-aminobutyric acid transmission. In the EU, stiripentol is indicated for use in conjunction with clobazam and valproate as adjunctive therapy of refractory generalized tonic-clonic seizures in patients with Dravet syndrome (DS; previously known as severe myoclonic epilepsy of infancy), whose seizures are not adequately controlled with clobazam and valproate. This approval (and similar DS indications in the USA, Canada and Japan), reflect the results of the STICLO studies, two small, randomized controlled trials in which stiripentol as adjunctive therapy was associated with a markedly superior response rate after 2 months compared with placebo in patients aged between 3 and ≈ 21 years with DS that was inadequately controlled with clobazam and valproate. These short-term results have subsequently been supported and extended by findings from longer-term, open-label, observational studies, including a retrospective longitudinal cohort study, which showed that the efficacy of combining stiripentol with clobazam and valproate when started at paediatric age was maintained in mid-adulthood with up to 24 years of exposure, and up to 40 years of age. Drowsiness, appetite loss, weight loss, ataxia and tremor are the most common adverse events associated with the addition of stiripentol to clobazam and valproate. Based on the available evidence, stiripentol, as an adjunct to clobazam and valproate, is a demonstrably beneficial and generally well-tolerated second-line treatment for patients with DS.
Collapse
Affiliation(s)
- James E Frampton
- Springer Nature, Private Bag 65901, Mairangi Bay, Auckland, 0754, New Zealand.
| |
Collapse
|
27
|
Sherman JJ, Riche DM, Warren CP. Cannabidiol Oral Solution: Challenges as a Treatment for Seizure Syndromes. J Nurse Pract 2020. [DOI: 10.1016/j.nurpra.2019.11.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
28
|
Tiraboschi E, Martina S, van der Ent W, Grzyb K, Gawel K, Cordero-Maldonado ML, Poovathingal SK, Heintz S, Satheesh SV, Brattespe J, Xu J, Suster M, Skupin A, Esguerra CV. New insights into the early mechanisms of epileptogenesis in a zebrafish model of Dravet syndrome. Epilepsia 2020; 61:549-560. [PMID: 32096222 DOI: 10.1111/epi.16456] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 02/03/2020] [Accepted: 02/03/2020] [Indexed: 12/17/2022]
Abstract
OBJECTIVE To pinpoint the earliest cellular defects underlying seizure onset (epileptogenic period) during perinatal brain development in a new zebrafish model of Dravet syndrome (DS) and to investigate potential disease-modifying activity of the 5HT2 receptor agonist fenfluramine. METHODS We used CRISPR/Cas9 mutagenesis to introduce a missense mutation, designed to perturb ion transport function in all channel isoforms, into scn1lab, the zebrafish orthologue of SCN1A (encoding voltage-gated sodium channel alpha subunit 1). We performed behavioral analysis and electroencephalographic recordings to measure convulsions and epileptiform discharges, followed by single-cell RNA-Seq, morphometric analysis of transgenic reporter-labeled γ-aminobutyric acidergic (GABAergic) neurons, and pharmacological profiling of mutant larvae. RESULTS Homozygous mutant (scn1labmut/mut ) larvae displayed spontaneous seizures with interictal, preictal, and ictal discharges (mean = 7.5 per 20-minute recording; P < .0001; one-way analysis of variance). Drop-Seq analysis revealed a 2:1 shift in the ratio of glutamatergic to GABAergic neurons in scn1labmut/mut larval brains versus wild type (WT), with dynamic changes in neuronal, glial, and progenitor cell populations. To explore disease pathophysiology further, we quantified dendritic arborization in GABAergic neurons and observed a 40% reduction in arbor number compared to WT (P < .001; n = 15 mutant, n = 16 WT). We postulate that the significant reduction in inhibitory arbors causes an inhibitory to excitatory neurotransmitter imbalance that contributes to seizures and enhanced electrical brain activity in scn1labmut/mut larvae (high-frequency range), with subsequent GABAergic neuronal loss and astrogliosis. Chronic fenfluramine administration completely restored dendritic arbor numbers to normal in scn1labmut/mut larvae, whereas similar treatment with the benzodiazepine diazepam attenuated seizures, but was ineffective in restoring neuronal cytoarchitecture. BrdU labeling revealed cell overproliferation in scn1labmut/mut larval brains that were rescued by fenfluramine but not diazepam. SIGNIFICANCE Our findings provide novel insights into early mechanisms of DS pathogenesis, describe dynamic cell population changes in the scn1labmut/mut brain, and present first-time evidence for potential disease modification by fenfluramine.
Collapse
Affiliation(s)
- Ettore Tiraboschi
- Chemical Neuroscience Group, Center for Molecular Medicine Norway, University of Oslo, Oslo, Norway
| | - Silvia Martina
- Integrative Cell Signaling Group, Luxembourg Center for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Wietske van der Ent
- Chemical Neuroscience Group, Center for Molecular Medicine Norway, University of Oslo, Oslo, Norway
| | - Kamil Grzyb
- Integrative Cell Signaling Group, Luxembourg Center for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Kinga Gawel
- Chemical Neuroscience Group, Center for Molecular Medicine Norway, University of Oslo, Oslo, Norway.,Department of Experimental and Clinical Pharmacology, Medical University of Lublin, Lublin, Poland
| | - Maria Lorena Cordero-Maldonado
- Integrative Cell Signaling Group, Luxembourg Center for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Suresh Kumar Poovathingal
- Integrative Cell Signaling Group, Luxembourg Center for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Sarah Heintz
- Chemical Neuroscience Group, Center for Molecular Medicine Norway, University of Oslo, Oslo, Norway
| | | | - Jarle Brattespe
- Department of Biological Sciences, University of Bergen, Bergen, Norway
| | - Ju Xu
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | | | - Alexander Skupin
- Integrative Cell Signaling Group, Luxembourg Center for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Camila V Esguerra
- Chemical Neuroscience Group, Center for Molecular Medicine Norway, University of Oslo, Oslo, Norway.,Department of Pharmacy, University of Oslo, Oslo, Norway
| |
Collapse
|
29
|
Cross JH, Caraballo RH, Nabbout R, Vigevano F, Guerrini R, Lagae L. Dravet syndrome: Treatment options and management of prolonged seizures. Epilepsia 2019; 60 Suppl 3:S39-S48. [DOI: 10.1111/epi.16334] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 08/10/2019] [Accepted: 08/11/2019] [Indexed: 12/22/2022]
Affiliation(s)
- J. Helen Cross
- University College London National Institute for Health Research Biomedical Research Centre Great Ormond Street Institute of Child Health London UK
- Great Ormond Street Hospital for Children London UK
| | - Roberto H. Caraballo
- Department of Neurology J. P. Garrahan Hospital, Capital Federal Buenos Aires Argentina
| | - Rima Nabbout
- Référence centre for rare épilepsies, department of pediatric neurology necker enfants malades hospital, aphp, Paris Descartes university Paris France
- Imagine institute insermU1163Paris France
| | - Federico Vigevano
- Pediatric Neurology Unit Bambino Gesù Children's Hospital Rome Italy
| | - Renzo Guerrini
- Pediatric Neurology Unit A. Meyer Children's HospitalUniversity of Florence Florence Italy
| | - Lieven Lagae
- Department of Pediatric Neurology UZ Leuven Leuven Belgium
| |
Collapse
|
30
|
Griffin AL, Jaishankar P, Grandjean JM, Olson SH, Renslo AR, Baraban SC. Zebrafish studies identify serotonin receptors mediating antiepileptic activity in Dravet syndrome. Brain Commun 2019; 1:fcz008. [PMID: 31667472 PMCID: PMC6798786 DOI: 10.1093/braincomms/fcz008] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Revised: 07/10/2019] [Accepted: 07/15/2019] [Indexed: 02/06/2023] Open
Abstract
Dravet syndrome is a life-threatening early-onset epilepsy not well controlled by antiepileptic drugs. Drugs that modulate serotonin (5-HT) signalling, including clemizole, locaserin, trazodone and fenfluramine, have recently emerged as potential treatment options for Dravet syndrome. To investigate the serotonin receptors that could moderate this antiepileptic activity, we designed and synthesized 28 novel analogues of clemizole, obtained receptor binding affinity profiles, and performed in vivo screening in a scn1lab mutant zebrafish (Danio rerio) model which recapitulates critical clinical features of Dravet syndrome. We discovered three clemizole analogues with 5-HT receptor binding that exert powerful antiepileptic activity. Based on structure–activity relationships and medicinal chemistry-based analysis, we then screened an additional set of known 5-HT receptor specific drug candidates. Integrating our in vitro and in vivo data implicates 5-HT2B receptors as a critical mediator in the mechanism of seizure suppression observed in Dravet syndrome patients treated with 5-HT modulating drugs.
Collapse
Affiliation(s)
- Aliesha L Griffin
- Epilepsy Research Laboratory and Weill Institute for Neuroscience, Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94122, USA
| | - Priyadarshini Jaishankar
- Department of Pharmaceutical Chemistry and Small Molecule Discovery Center, University of California San Francisco, CA 94143, USA
| | - Jean-Marc Grandjean
- Department of Neurology, Institute for Neurodegenerative Diseases and Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94143, USA
| | - Steven H Olson
- Department of Neurology, Institute for Neurodegenerative Diseases and Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94143, USA
| | - Adam R Renslo
- Department of Pharmaceutical Chemistry and Small Molecule Discovery Center, University of California San Francisco, CA 94143, USA
| | - Scott C Baraban
- Epilepsy Research Laboratory and Weill Institute for Neuroscience, Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94122, USA
| |
Collapse
|
31
|
Lazaridis D, Eraikhuemen N, Williams K, Lovince J. Treatment of Seizures Associated with Lennox-Gastaut and Dravet Syndromes: A Focus on Cannabidiol Oral Solution. P & T : A PEER-REVIEWED JOURNAL FOR FORMULARY MANAGEMENT 2019; 44:255-266. [PMID: 31080333 PMCID: PMC6487974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Cannabidiol oral solution for seizures associated with Lennox-Gastaut and Dravet syndromes.
Collapse
|
32
|
Jagadish S, Payne ET, Wong-Kisiel L, Nickels KC, Eckert S, Wirrell EC. The Ketogenic and Modified Atkins Diet Therapy for Children With Refractory Epilepsy of Genetic Etiology. Pediatr Neurol 2019; 94:32-37. [PMID: 30803845 DOI: 10.1016/j.pediatrneurol.2018.12.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 12/21/2018] [Accepted: 12/23/2018] [Indexed: 02/08/2023]
Abstract
BACKGROUND The ketogenic diet is an accepted treatment modality in refractory childhood epilepsy. In this study, we analyzed the efficacy and tolerability of the ketogenic and modified Atkins diets in children with refractory epilepsy of genetic etiology and studied the effect of the diet on seizure frequency. METHODS The records of children with a genetic etiology for refractory epilepsy treated with ketogenic and modified Atkins diet between September 2005 and July 2016 were reviewed. We documented age of seizure and diet onset, seizure characteristics, and specific genetic etiology. The proportion of children remaining on the diet and responder rates (greater than 50% seizure reduction) were noted at one, three, six, 12, and 24 months after diet initiation. Tolerability and safety profile were also recorded. RESULTS Fifty-nine children with a genetic etiology (63% females, median age at diet onset 2.2 years) were initiated on the diet at our center. Fifty-three (90%) were started on a traditional ketogenic diet, whereas six started a modified Atkins diet. The adverse events at the initiation of diet were vomiting (24%), hypoglycemia (15%), and refusal to feed (11%). Three children stopped the diet before discharge because of poor compliance, severe reflux, and ketoacidosis (n = 1 each). The proportion of children remaining on the diet at one, three, six, 12, and 24 months was 95%, 86%, 69%, 64%, and 47%. The responder rates were 63%, 61%, 54%, 53%, and 41% at one, three, six, 12, and 24 months, respectively. CONCLUSIONS The ketogenic diet is an effective treatment modality in children with refractory epilepsy of genetic etiology.
Collapse
|
33
|
Chiron C. Stiripentol for the treatment of seizures associated with Dravet syndrome. Expert Rev Neurother 2019; 19:301-310. [DOI: 10.1080/14737175.2019.1593142] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Catherine Chiron
- Inserm NeuroDiderot, InDev, Paris, France
- Neurospin, CEA, Paris, France
| |
Collapse
|
34
|
|
35
|
Thijs RD, Surges R, O'Brien TJ, Sander JW. Epilepsy in adults. Lancet 2019; 393:689-701. [PMID: 30686584 DOI: 10.1016/s0140-6736(18)32596-0] [Citation(s) in RCA: 1064] [Impact Index Per Article: 177.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 09/08/2018] [Accepted: 10/11/2018] [Indexed: 12/11/2022]
Abstract
Epilepsy is one of the most common serious brain conditions, affecting over 70 million people worldwide. Its incidence has a bimodal distribution with the highest risk in infants and older age groups. Progress in genomic technology is exposing the complex genetic architecture of the common types of epilepsy, and is driving a paradigm shift. Epilepsy is a symptom complex with multiple risk factors and a strong genetic predisposition rather than a condition with a single expression and cause. These advances have resulted in the new classification of epileptic seizures and epilepsies. A detailed clinical history and a reliable eyewitness account of a seizure are the cornerstones of the diagnosis. Ancillary investigations can help to determine cause and prognosis. Advances in brain imaging are helping to identify the structural and functional causes and consequences of the epilepsies. Comorbidities are increasingly recognised as important aetiological and prognostic markers. Antiseizure medication might suppress seizures in up to two-thirds of all individuals but do not alter long-term prognosis. Epilepsy surgery is the most effective way to achieve long-term seizure freedom in selected individuals with drug-resistant focal epilepsy, but it is probably not used enough. With improved understanding of the gradual development of epilepsy, epigenetic determinants, and pharmacogenomics comes the hope for better, disease-modifying, or even curative, pharmacological and non-pharmacological treatment strategies. Other developments are clinical implementation of seizure detection devices and new neuromodulation techniques, including responsive neural stimulation.
Collapse
Affiliation(s)
- Roland D Thijs
- Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede, Netherlands; Department of Neurology, Leiden University Medical Centre, Leiden, Netherlands; NIHR University College London Hospitals Biomedical Research Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Rainer Surges
- Section of Epileptology, Department of Neurology, University Hospital RWTH Aachen, Germany
| | - Terence J O'Brien
- Melbourne Brain Centre, Departments of Medicine and Neurology, Royal Melbourne Hospital, University of Melbourne, VIC, Australia; Departments of Neuroscience and Neurology, Central Clinical School, Monash University, The Alfred Hospital, Melbourne, VIC, Australia
| | - Josemir W Sander
- Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede, Netherlands; NIHR University College London Hospitals Biomedical Research Centre, UCL Queen Square Institute of Neurology, London, UK; Chalfont Centre for Epilepsy, Chalfont St Peter, UK.
| |
Collapse
|
36
|
|
37
|
Brigo F, Striano P, Balagura G, Belcastro V. Emerging drugs for the treatment of Dravet syndrome. Expert Opin Emerg Drugs 2018; 23:261-269. [DOI: 10.1080/14728214.2018.1552937] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Francesco Brigo
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
- Division of Neurology, “Franz Tappeiner” Hospital, Merano, Italy
| | - Pasquale Striano
- Pediatric Neurology and Muscular Diseases Unit, Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, “G. Gaslini” Institute, Genova, Italy
| | - Ganna Balagura
- Pediatric Neurology and Muscular Diseases Unit, Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, “G. Gaslini” Institute, Genova, Italy
| | | |
Collapse
|
38
|
Abstract
PURPOSE OF REVIEW Dravet syndrome is a rare but severe genetic epilepsy that has unique treatment challenges. This is a review of current and future potential treatment options. RECENT FINDINGS Treatment for Dravet syndrome should encompass many aspects of the syndrome such as gait, behavior, and nutrition, as well as focus on seizure control. Many sodium channel blockers should be avoided as they are likely to exacerbate seizures. Current options for treatment include valproic acid, clobazam, stiripentol, and ketogenic diet. Testing is underway for several new treatment options with unique mechanisms of action and therapeutic targets, including the serotonin system and genetic modulation. Accurate and early diagnosis of Dravet syndrome will lead to avoidance of medications that may exacerbate seizures. Additionally, a multi-disciplinary approach and careful planning for management of episodes of status epilepticus may lead to improved outcomes. Ongoing research for novel approaches to treatment creates optimism for future improvement in outcomes.
Collapse
|
39
|
Ruffolo G, Cifelli P, Roseti C, Thom M, van Vliet EA, Limatola C, Aronica E, Palma E. A novel GABAergic dysfunction in human Dravet syndrome. Epilepsia 2018; 59:2106-2117. [DOI: 10.1111/epi.14574] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 09/11/2018] [Accepted: 09/11/2018] [Indexed: 01/09/2023]
Affiliation(s)
- Gabriele Ruffolo
- Department of Physiology and Pharmacology; Pasteur Institute-Cenci Bolognetti Foundation; Sapienza University of Rome; Rome Italy
| | - Pierangelo Cifelli
- Department of Physiology and Pharmacology; Pasteur Institute-Cenci Bolognetti Foundation; Sapienza University of Rome; Rome Italy
- IRCCS Neuromed; Pozzilli Italy
| | | | - Maria Thom
- Department of Clinical and Experimental Epilepsy; University College London Institute of Neurology; London UK
| | - Erwin A. van Vliet
- Department of (Neuro)Pathology; Amsterdam UMC; University of Amsterdam; Amsterdam Neuroscience; Amsterdam The Netherlands
- Center for Neuroscience; Swammerdam Institute for Life Sciences; University of Amsterdam; Amsterdam The Netherlands
| | - Cristina Limatola
- Department of Physiology and Pharmacology; Pasteur Institute-Cenci Bolognetti Foundation; Sapienza University of Rome; Rome Italy
- IRCCS Neuromed; Pozzilli Italy
| | - Eleonora Aronica
- Department of (Neuro)Pathology; Amsterdam UMC; University of Amsterdam; Amsterdam Neuroscience; Amsterdam The Netherlands
- Stichting Epilepsie Instellingen Nederland (SEIN); Heemstede The Netherlands
| | - Eleonora Palma
- Department of Physiology and Pharmacology; Pasteur Institute-Cenci Bolognetti Foundation; Sapienza University of Rome; Rome Italy
- IRCCS San Raffaele Pisana; Rome Italy
| |
Collapse
|
40
|
Lin KL, Lin JJ, Chou ML, Hung PC, Hsieh MY, Chou IJ, Lim SN, Wu T, Wang HS. Efficacy and tolerability of perampanel in children and adolescents with pharmacoresistant epilepsy: The first real-world evaluation in Asian pediatric neurology clinics. Epilepsy Behav 2018; 85:188-194. [PMID: 30032806 DOI: 10.1016/j.yebeh.2018.06.033] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 06/09/2018] [Accepted: 06/17/2018] [Indexed: 12/25/2022]
Abstract
AIM This study investigated the efficacy and safety of perampanel (PER) adjunctive therapy in pediatric patients with epilepsy whose seizures are pharmacoresistant to existing antiepileptic drugs. METHODS A clinical retrospective study was conducted from 2016 to 2017 in the pediatric neurology clinic at a tertiary children's hospital. We reviewed the data obtained from 66 children whose seizures were pharmacoresistant to more than two antiepileptic drugs, and could be followed up for a minimum of 3 months after PER adjunctive therapy initiation. The efficacy was estimated by the PER response rate at 3-, 6-, and 12-month follow-up evaluations, and adverse events were also recorded. RESULTS The rate of seizure reduction of >50% was 30.3%, 37.5%, and 34.7% for all seizure types at 3, 6, and 12 months, in which 7.6%, 8.9%, and 14.3% of the patients became seizure-free at these time points, respectively. No significant differences were found between enzyme-inducing and nonenzyme-inducing antiepileptic drugs in combination with PER with regard to the responder rate. Five patients with Dravet syndrome were included in the study. Four of them (80%) exhibited 50% seizure reduction at the last visit, at which point, two patients (40.0%) were seizure-free. The retention rate was 51% at 12 months. Adverse events were documented in 25 patients (35.7%) and led to PER discontinuation in eight patients (12.1%). The most common adverse events comprised irritability, skin rash, dizziness, and somnolence; however, all were transient and successfully managed after PER dose reduction or discontinuation. CONCLUSION The current data support the value of adjunctive PER in child and adolescent patients with pharmacoresistant epilepsy in daily clinical practice. Perampanel was efficacious and generally well-tolerated as an add-on treatment for epilepsy.
Collapse
Affiliation(s)
- Kuang-Lin Lin
- Division of Pediatric Neurology, Chang Gung Children's Hospital and Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Jainn-Jim Lin
- Division of Pediatric Neurology, Chang Gung Children's Hospital and Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan; Division of Pediatric Critical Care and Pediatric Neurocritical Care Center, Chang Gung Children's Hospital and Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan; Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ming-Liang Chou
- Division of Pediatric Neurology, Chang Gung Children's Hospital and Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Po-Cheng Hung
- Division of Pediatric Neurology, Chang Gung Children's Hospital and Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Meng-Ying Hsieh
- Division of Pediatric Neurology, Chang Gung Children's Hospital and Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - I-Jun Chou
- Division of Pediatric Neurology, Chang Gung Children's Hospital and Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Siew-Na Lim
- Department of Neurology, Section of Epilepsy, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Tony Wu
- Department of Neurology, Section of Epilepsy, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Huei-Shyong Wang
- Division of Pediatric Neurology, Chang Gung Children's Hospital and Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan.
| |
Collapse
|